WO2011110861A2 - Therapy for influenza like illness - Google Patents

Therapy for influenza like illness Download PDF

Info

Publication number
WO2011110861A2
WO2011110861A2 PCT/GB2011/050480 GB2011050480W WO2011110861A2 WO 2011110861 A2 WO2011110861 A2 WO 2011110861A2 GB 2011050480 W GB2011050480 W GB 2011050480W WO 2011110861 A2 WO2011110861 A2 WO 2011110861A2
Authority
WO
WIPO (PCT)
Prior art keywords
ifn
agent
influenza
respiratory tract
treatment
Prior art date
Application number
PCT/GB2011/050480
Other languages
French (fr)
Other versions
WO2011110861A3 (en
Inventor
Victoria Jane Tear
James Jonathan Welch Roberts
Phillip David Monk
Original Assignee
Synairgen Research Limited
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 GBGB1004144.0A external-priority patent/GB201004144D0/en
Priority claimed from GBGB1008114.9A external-priority patent/GB201008114D0/en
Priority to ES11730046.7T priority Critical patent/ES2592528T3/en
Priority to CA2792727A priority patent/CA2792727C/en
Priority to RS20160753A priority patent/RS55154B1/en
Priority to SI201130956A priority patent/SI2544705T1/en
Priority to LTEP11730046.7T priority patent/LT2544705T/en
Priority to CN2011800135925A priority patent/CN102883741A/en
Application filed by Synairgen Research Limited filed Critical Synairgen Research Limited
Priority to US13/583,976 priority patent/US9421243B2/en
Priority to JP2012557607A priority patent/JP5926200B2/en
Priority to DK11730046.7T priority patent/DK2544705T3/en
Priority to EP11730046.7A priority patent/EP2544705B1/en
Publication of WO2011110861A2 publication Critical patent/WO2011110861A2/en
Publication of WO2011110861A3 publication Critical patent/WO2011110861A3/en
Priority to HRP20161158TT priority patent/HRP20161158T1/en
Priority to CY20161100897T priority patent/CY1117985T1/en
Priority to SM201600334T priority patent/SMT201600334B/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/215IFN-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/351Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom not condensed with another ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/565IFN-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • C07K14/08RNA viruses
    • C07K14/11Orthomyxoviridae, e.g. influenza virus

Definitions

  • the invention relates to the treatment of individuals with lower respiratory tract illness that has developed during or following an established influenza-like illness (ILI).
  • established influenza like illness we mean an illness in which the symptoms have been apparent, e,g., to the individual or to a carer, for at least 24 hours, more particularly at least 48 hours.
  • the invention relates to the treatment of individuals with lower respiratory tract illness that have developed during or following an ILI, particularly hospitalized patients, by delivery of interferon- ⁇ (IFN- ⁇ ) or an agent that increases IFN- ⁇ expression by aerosol to the lower respiratory tract.
  • ILI interferon- ⁇
  • Influenza like illness is a disease characterised by two of the following symptoms: headache, cough, sore throat, and myalgia and/or confirmed influenza.
  • ILI may be defined as a fever >37.8°C plus two of the following symptoms: (headache, cough, sore throat, and myalgia) and/or confirmed influenza infection.
  • Seasonal influenza is a common infection, especially during winter. Every year strains of influenza (type A or B) circulate, giving rise to clinical consultations in primary care, episodes of hospital treatment (mainly in older persons and young children, but occasionally in middle aged adults), and deaths (mainly in the elderly). Treatment in primary care and hospital may be required due to the direct effects of influenza virus infection or its possible complications, most commonly secondary bacterial infection. Increases in primary care consultations for ILI and winter bed pressures are frequently associated with periods of known community influenza activity.
  • Pandemic influenza occurs when a new influenza virus subtype emerges which is markedly different from recently circulating subtypes and strains, and is able to: • infect humans;
  • Highly pathogenic influenza is a virulent form of influenza, such as H5N1 , which leads to a rapid and high level of morbidity.
  • ILI includes seasonal influenza, pandemic influenza and highly pathogenic influenza.
  • the invention provides an agent selected from:
  • the invention further provides a method of treating an individual diagnosed with lower respiratory tract illness that has developed during or following an established ILI comprising aerosol delivery to the lower respiratory tract of the individual of an agent selected from the group consisting of:
  • MxA myxovirus resistance protein 1
  • Whole blood was treated with IFN- ⁇ (0-1000 lU/ml) for 4 hours, RNA extracted and effect on 2'-5' OAS gene expression measured.
  • SEQ ID NO: 1 shows the nucleotide sequence of human IFN ⁇ I a.
  • SEQ ID NO: 2 shows the amino acid sequence of human IFN ⁇ Ia.
  • SEQ ID NO: 3 shows the nucleotide sequence of human IFN- ⁇ b.
  • SEQ ID NO: 4 shows the amino acid sequence of human IFN- ⁇ b.
  • IFN- ⁇ b is identical to human IFN- ⁇ a except for replacement of the cysteine at residue 17 with serine.
  • the present invention relates to new therapeutic uses for IFN- ⁇ .
  • it relates, for example, to therapeutic use of IFN- ⁇ by aerosol delivery to the lower respiratory tract for the treatment of individuals diagnosed as having lower respiratory tract illness that has developed during or following an influenza like illness, particularly those patients who have been admitted to hospital.
  • interferons have been proposed previously for the prophylaxis or early stage treatment of ILI. Accordingly, interferons have been proposed for use either before symptoms of ILI are manifested or on the initial occurrence of such symptoms. In general, these interferons have been administered to the upper respiratory tract, eg the nasal pharynx. Usually initiation of treatment with interferons begins within 24 hours of the occurrence of symptoms.
  • IFN- ⁇ as used herein will be understood to refer to any form or analog of IFN- ⁇ that retains the biological activity of native IFN- ⁇ and preferably retains the activity of IFN- ⁇ that is present in the lung and, in particular, the bronchial and/or alveolar epithelium.
  • the IFN- ⁇ may be identical to or comprise the sequence of human IFN- ⁇ a (SEQ ID NO: 2) or human IFN- ⁇ b (SEQ ID NO: 4).
  • IFN- ⁇ also refers to a variant polypeptide having an amino acid sequence which varies from that of SEQ ID NO: 2 or 4.
  • IFN- ⁇ may be chemically-modified.
  • a variant of IFN- ⁇ may be a naturally occurring variant, for example a variant which is expressed by a non-human species.
  • variants of IFN- ⁇ include sequences which vary from SEQ ID NO: 2 or 4 but are not necessarily naturally occurring. Over the entire length of the amino acid sequence of SEQ ID NO: 2 or 4, a variant will preferably be at least 80% homologous to that sequence based on amino acid identity. More preferably, the polypeptide is at least 85% or 90% and more preferably at least 95%, 97% or 99% homologous based on amino acid identity to the amino acid sequence of SEQ ID NO: 2 or 4 over the entire sequence.
  • Homology may be determined using any method known in the art.
  • the UWGCG Package provides the BESTFIT program which can be used to calculate homology, for example used on its default settings (Devereux et al (1984) Nucleic Acids Research 12, p387-395).
  • the PILEUP and BLAST algorithms can be used to calculate homology or line up sequences (such as identifying equivalent residues or corresponding sequences (typically on their default settings)), for example as described in Altschul S. F.
  • the word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Extensions for the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment.
  • the BLAST program uses as defaults a word length (W) of 1 1 , the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1992) Proc. Natl. Acad.
  • a sequence is considered similar to another sequence if the smallest sum probability in comparison of the first sequence to the second sequence is less than about 1 , preferably less than about 0.1 , more preferably less than about 0.01 , and most preferably less than about 0.001 .
  • Amino acid substitutions may be made to the amino acid sequence of SEQ ID NO: 1 or 2, for example from 1 , 2, 3, 4 or 5 to 10, 20 or 30 substitutions. Conservative substitutions may be made, for example, according to Table 1 .
  • Amino acids in the same block in the second column and preferably in the same line in the third column may be substituted for each other: Table 1 - Conservative amino acid substitutions NON- AROMATIC Non-polar G A P I L V Polar - uncharged C S T M N Q Polar - charged D E H KR AROMATIC H F W Y
  • IFN- ⁇ also includes fragments of the above-mentioned sequences. Such fragments retain IFN- ⁇ activity. Fragments may be at least from 120 or 140 amino acids in length. Such fragments may be used to produce chimeric agents as described in more detail below. IFN- ⁇ includes chimeric proteins comprising fragments or portions of SEQ ID NO: 2 or 4. One or more amino acids may be alternatively or additionally added to the polypeptides described above.
  • An extension may be provided at the N-terminus or C-terminus of the amino acid sequence of SEQ ID NO: 2 or 4 or polypeptide variant or fragment thereof.
  • the extension may be quite short, for example from 1 to 10 amino acids in length. Alternatively, the extension may be longer.
  • a carrier protein may be fused to an amino acid sequence described above.
  • a fusion protein incorporating one of the polypeptides described above can thus be used in the invention.
  • IFN- ⁇ also includes SEQ ID NO: 2 or 4 or variants thereof that have been chemically-modified. A number of side chain modifications are known in the art and may be made to the side chains of the proteins or peptides discussed above.
  • Such modifications include, for example, glycosylation, phosphorylation, modifications of amino acids by reductive alkylation by reaction with an aldehyde followed by reduction with NaBH , amidination with methylacetimidate or acylation with acetic anhydride.
  • the modification is preferably glycosylation.
  • the IFN- ⁇ may be made synthetically or by recombinant means using methods known in the art.
  • the amino acid sequence of proteins and polypeptides may be modified to include non- naturally occurring amino acids or to increase the stability of the compound. When the proteins or peptides are produced by synthetic means, such amino acids may be introduced during production.
  • the proteins or peptides may also be modified following either synthetic or recombinant production.
  • the IFN- ⁇ may also be produced using D-amino acids. In such cases the amino acids will be linked in reverse sequence in the C to N orientation. This is conventional in the art for producing such proteins or peptides.
  • the IFN- ⁇ may be produced in a cell by in situ expression of the polypeptide from a recombinant expression vector.
  • the expression vector optionally carries an inducible promoter to control the expression of the polypeptide.
  • the IFN- ⁇ or analog thereof may be produced in large scale following purification by any protein liquid chromatography system after recombinant expression.
  • Preferred protein liquid chromatography systems include FPLC, AKTA systems, the Bio-Cad system, the Bio- RadBioLogic system and the Gilson HPLC system.
  • Commercially available forms of IFN- ⁇ or analogs thereof may be used in the invention. Examples include Betaseron® and Avonex®. Agents that increase IFN- ⁇ expression
  • the invention may also involve using an agent that increases endogenous expression of IFN- ⁇ in the lung or preferably the bronchial and/or alveolar epithelium.
  • the agents may act directly on the promoter or other regulatory sequences of the IFN- ⁇ gene. Such agents may act to reduce the constitutive silencing of the IFN- ⁇ promoter.
  • the agent may stimulate cells to produce endogenous IFN- ⁇ by acting at receptors at the cell surface.
  • Agents that increase endogenous expression of IFN- ⁇ of interest in relation to the present invention include, but are not limited to, poly(inosinic acid)-poly(cytidylic acid) (poly(IC)), ANA773, perindopril, BL-20803, Tilorone, ABMP, DRB, Atabrine, 10-carboxy-9acridone, CP-28888, Bropirimine, and Imiquimod.
  • the invention may also involve using a polynucleotide which is capable of expressing IFN- ⁇ or an agent that increases endogenous expression of IFN- ⁇ in lung airways.
  • a polynucleotide may preferably be in the form of a vector capable of directing expression of IFN- ⁇ or an agent that induces IFN- ⁇ in the bronchial and/or alveolar epithelium.
  • the resulting IFN- ⁇ or agent may then have a therapeutic effect ("gene therapy").
  • the polynucleotide may encode any of the forms of IFN- ⁇ discussed above including the variants, fragments and chimeric proteins thereof.
  • the polynucleotide encoding IFN- ⁇ may comprise the human sequence (SEQ ID NO: 1 or 3) or a naturally occurring sequence variant, for example a variant which is expressed by a non-human species. Also, a polynucleotide encoding IFN- ⁇ includes sequences which vary from SEQ ID NO: 1 or 3 but are not necessarily naturally occurring. Over the entire length of the amino acid sequence of SEQ ID NO: 1 or 3, a variant will preferably be at least 80% homologous to that sequence based on nucleotide identity.
  • the polynucleotide is at least 85% or 90% and more preferably at least 95%, 97% or 99% homologous based on nucleotide identity to the nucleotide of SEQ ID NO: 1 or 3 over the entire sequence.
  • the polynucleotides may comprise DNA or RNA but preferably comprise DNA. They may also be polynucleotides which include within them synthetic or modified nucleotides.
  • polynucleotides A number of different types of modification to polynucleotides are known in the art. These include methylphosphate and phosphorothioate backbones, addition of acridine or polylysine chains at the 3' and/or 5' ends of the molecule.
  • polynucleotides described herein may be modified by any method available in the art. Polynucleotides such as a DNA polynucleotide may be produced recombinantly, synthetically, or by any means available to those of skill in the art. They may also be cloned by standard techniques. The polynucleotides are typically provided in isolated and/or purified form.
  • Polynucleotides will generally be produced using recombinant means, for example using PCR (polymerase chain reaction) cloning techniques. This will involve making a pair of primers (e.g. of about 15- 30 nucleotides) to a region of the required gene which it is desired to clone, bringing the primers into contact with DNA obtained from a suitable cell, performing a polymerase chain reaction under conditions which bring about amplification of the desired region, isolating the amplified fragment (e.g. by purifying the reaction mixture on an agarose gel) and recovering the amplified DNA.
  • the primers may be designed to contain suitable restriction enzyme recognition sites so that the amplified DNA can be cloned into a suitable cloning vector.
  • the polynucleotide is used in an expression vector wherein it is operably linked to a control sequence which is capable of providing for the expression of the coding sequence in the airways of human lung.
  • Expression vectors for use in accordance with the invention may be any type of vector conventionally employed for gene therapy. It may be a plasmid expression vector administered as naked DNA or complexed with one or more cationic amphiphiles, e.g one or more cationic lipids, e.g. in the form of DNA/liposomes.
  • a viral vector may alternatively be employed.
  • the IFN- ⁇ , agent or polynucleotide may be administered in a medicament or pharmaceutical composition suitable for airway delivery which will typically also include a pharmaceutically acceptable excipient.
  • a pharmaceutically acceptable excipient generally refers to a substantially inert material that is nontoxic and does not interact with other components of the composition in a deleterious manner.
  • Pharmaceutically acceptable excipients include, but are not limited to, liquids such as water, saline, polyethyleneglycol, hyaluronic acid, glycerol and ethanol.
  • compositions or medicaments comprising the therapeutic agent will contain a pharmaceutically acceptable carrier that serves as a stabilizer, particularly for peptide, protein, polynucleotide or other like agents.
  • suitable carriers that also act as stabilizers for peptides include, without limitation, pharmaceutical grades of dextrose, sucrose, lactose, trehalose, mannitol, sorbitol, inositol, dextran, and the like.
  • Other suitable carriers include, again without limitation, starch, cellulose, sodium or calcium phosphates, citric acid, tartaric acid, glycine, high molecular weight polyethylene glycols (PEGs), and combination thereof. It may also be useful to employ a charged lipid and/or detergent.
  • Suitable charged lipids include, without limitation, phosphatidylcholines (lecithin), and the like.
  • Detergents will typically be a nonionic, anionic, cationic or amphoteric surfactant.
  • suitable surfactants include, for example, Tergitol® and Triton® surfactants (Union Carbide Chemicals anaplastics, Danbury, CT), polyoxyethylenesorbitans, for example, TWEEN® surfactants (Atlas Chemical Industries, Wilmington, DE), polyoxyethylene ethers, for example Brij, pharmaceutically acceptable fatty acid esters, for example, lauryl sulfate and salts thereof (SDS), and like materials.
  • SDS lauryl sulfate and salts thereof
  • a suitable composition for airway delivery of IFN- ⁇ may, for example, be formulated as described in US Patent no 6,030,609 by dissolving lyophilised IFN- ⁇ in a pharmaceutically acceptable vehicle such as sterile distilled water or sterile physiological saline, optionally with addition of one or more carriers, stabilizers, surfactants or other agents in order to enhance effectiveness of the IFN- ⁇ active agent.
  • a pharmaceutically acceptable vehicle such as sterile distilled water or sterile physiological saline
  • a suitable composition has a pH of from 5 to 8, more preferably 5.5 to 7.5.
  • the composition is buffered, e.g. using a citrate buffer.
  • the Rentschler composition which is based on formulations disclosed in US 6,030,609, has a pH of 6.5 and osmolarity of 290 mOsm/kg.
  • the composition is preferably provided as a sterile, clear and colourless, ready-to-use aqueous nebuliser solution presented in a disposable glass syringe.
  • the composition preferably comprises a buffering system to maintain the pH at between 5 and 8, more preferably 5.5 and 7.5, especially 6.5.
  • the composition also preferably comprises an antioxidant, for example, DL-methionine.
  • a composition comprising a therapeutically effective amount of the IFN- ⁇ , agent or polynucleotide described herein may conveniently be delivered to the lung airways by means of an inhalation device that is capable of delivering fine particles of the active ingredient to the lower respiratory tract or airways.
  • particles of the active ingredient will have a mass median diameter of 1 -10 microns.
  • Suitable inhalation devices include dry powder inhalation (DPI) devices, pressurised metered dose inhalers (pMDI) and aerosol nebulisers.
  • the inhalation device will produce an aerosol with a particle size, as determined using a Malvern Masterizer S, with a mass median diameter of 1 -10 micron, preferably 3-8 micron, in which mass per cent have a diameter below 5 micron is from 25-80%, preferably 30-65%.
  • a suitable nebulizier is the l-neb device, a CE-marked nebuliser manufactured by Philips Respironics.
  • An appropriate effective amount may be determined by appropriate clinical testing and will vary with for example the activity of the IFN- ⁇ administered or induced.
  • the IFN- ⁇ , agent or polynucleotide may for example, be administered in microgram amounts.
  • the amount to be delivered per dose may be 0.1 g to 500ig, for example 1 to 50 g, depending on the subject to be treated.
  • the treatment normally lasts from 5-7 days and may continue to 14 days if symptoms persist. Treatment may be given from every other day to several times a day. Preferably, treatment is given once per day.
  • the IFN- ⁇ , agent or polynucleotide may be administered on its own or simultaneously, sequentially or separately in combination with another therapeutic compound.
  • the IFN- ⁇ , agent or polynucleotide may be administered in conjunction with a therapeutic compound used to treat the respiratory disease or antiviral to the individual.
  • the IFN- ⁇ , agent or polynucleotide and additional therapeutic compound may be formulated in the same or different compositions.
  • the IFN- ⁇ , agent or polynucleotide is administered to an individual with asthma in combination with an inhaled corticosteroid.
  • the IFN- ⁇ , agent or polynucleotide may be administered simultaneously, sequentially or separately with an inhaled neuraminidase inhibitor.
  • a product for treatment of individuals diagnosed as having lower respiratory tract disease (or symptoms) that have developed during or following an influenza like illness comprising for simultaneous, separate or sequential lower respiratory tract administration (i) a first agent selected from (a) IFN- ⁇ , (b) an agent that increases IFN- ⁇ expression and (c) a polynucleotide capable of expressing (a) or (b) and (ii) an inhaled neuraminidase inhibitor.
  • such a product will provide for simultaneous, separate or sequential administration of IFN- ⁇ and an inhaled neuraminidase inhibitor, for example, zanamivir.
  • a first agent as defined above and an inhaled neuraminidase inhibitor may, for example, be provided in the form of a single pharmaceutical composition suitable for aerosol delivery to the airways, e.g., by means of a dry powder inhaler, a pressurised metered dose inhaler or an aerosol nebulizer.
  • the IFN- ⁇ , agent or polynucleotide may be administered simultaneously, sequentially or separately with an oral neuraminidase inhibitor, such as oseltamivir.
  • an oral neuraminidase inhibitor such as oseltamivir.
  • the IFN- ⁇ , agent or polynucleotide may be administered simultaneously, sequentially or separately with a systemically administered neuraminidase inhibitor, such as peramivir.
  • a systemically administered neuraminidase inhibitor such as peramivir.
  • the IFN- ⁇ , agent or polynucleotide may be administered simultaneously, sequentially or separately with an inhaled influenza virus attachment inhibitor.
  • a product for treatment of individuals diagnosed as having lower respiratory tract disease (or symptoms) that have developed during or following an influenza like illness comprising for simultaneous, separate or sequential lower respiratory tract administration (i) a first agent selected from (a) IFN- ⁇ , (b) an agent that increases IFN- ⁇ expression and (c) a polynucleotide capable of expressing (a) or (b) and (ii) an inhaled influenza virus attachment inhibitor.
  • a product will provide for simultaneous, separate or sequential administration of IFN- ⁇ and an influenza virus attachment inhibitor, for example, the sialidase fusion protein, DAS181 (Fludase®).
  • a first agent as defined above and an inhaled influenza virus attachment inhibitor may, for example, be provided in the form of a single pharmaceutical composition suitable for aerosol delivery to the airways.
  • the IFN- ⁇ , agent or polynucleotide may be administered simultaneously, sequentially or separately with an antibacterial antibiotic.
  • the antibacterial antibiotic is administered by inhalation.
  • the IFN- ⁇ , agent or polynucleotide may be administered simultaneously, sequentially or separately with an antifungal antibiotic.
  • the antifungal antibiotic is administered by inhalation.
  • a further at risk group are patients who have not previously been exposed to an ILL Example 1
  • exogenous IFN- ⁇ to suppress influenza A infection in an in vitro model of established infection of the lower respiratory tract.
  • HBE cells Human lung bronchial epithelial (HBE) cells were cultured in minimum essential medium (MEM) containing glutamax (Invitrogen), 10% fetal bovine serum (FBS) (Invitrogen), penicillin (50 U/ml) and streptomycin (50 pg/ml) (Invitrogen). HBE cells were plated at 1 .5x10 5 /well, passage 41 -49, in a 24 well plate. At approximately 60-70% confluency, the cells were cultured in reduced serum medium (MEM containing glutamax (Invitrogen) and 0.75-1 % FBS (Invitrogen)) for 24 hours.
  • MEM minimum essential medium
  • FBS fetal bovine serum
  • penicillin 50 U/ml
  • streptomycin 50 pg/ml
  • Cell media was collected at 24, 48, 72, and 96 hours post-infection and stored at -80°C. At 24, 48, 72 and 96 hours post-infection cells were trypsinised, counted and washed in phosphate buffered saline (PBS). Up to 2x10 5 cells were taken for analysis by flow cytometry. The cells were centrifuged at 400g and the supernatant discarded. Live/dead cell stain (Molecular Probes) was reconstituted in dimethyl sulfoxide (DMSO) (500 ⁇ ) and diluted 1 :100 in PBS. Diluted live/dead cell stain (100 ⁇ ) was incubated with cells at room temperature for 30 minutes.
  • DMSO dimethyl sulfoxide
  • MDCK cells Madin Darby canine kidney (MDCK) cells were cultured in MEM containing glutamax (Invitrogen), 10% FBS (Invitrogen), non-essential amino acids (Invitrogen), and penicillin/streptomycin (Invitrogen). MDCK cells were plated at 1 x10 5 cells/well, passage 25-30, in a 12 well plate. On reaching 100% confluence cells were washed twice with PBS.
  • DMEM serum-free media
  • penicillin/streptomycin Invitrogen
  • L-glutamine Invitrogen
  • Non-essential amino acids Invitrogen
  • sodium pyruvate Invitrogen
  • Overlay medium 100 ml 10 times concentrated MEM (Invitrogen), 20 ml 7.5% sodium bicarbonate (Sigma), 10 ml 1 M HEPES (Fluka), 28 ml 7.5% bovine serum albumin (BSA) fraction V (Sigma), 5 ml 1 % DEAE-dextran (Sigma), 20 ml penicillin/streptomycin (Invitrogen), 10 ml 200 mM glutamine (Invitrogen), 507 ml d.H 2 O) was prepared and 17.5 ml added to 12.5 ⁇ 1 mg/ml trypsin treated with L-(tosylamido-2- phenyl) ethyl chloromethyl ketone (TRTPCK trypsin) (Worthington Biochemical Corp.) and 7.5 ml Avicel suspension (4.8g Avicel (FMC Biopolymer) in 186 ml d.H 2 O).
  • TRTPCK trypsin L-(tosylamid
  • the overlay was mixed by inverting and 1 ml added per well. Plates were incubated for 2 days without moving or shaking. Overlay medium was aspirated and cells washed twice with PBS. Crystal violet solution was added (0.5 ml) (0.65g crystal violet (Sigma), 25 ml formaldehyde (Sigma), 25 ml 100% ethanol (Sigma), 450 ml 1 xPBS (5 DulA PBS tablets (Sigma) + 500 ml d.H 2 0)) for 30 minutes at room temperature. Crystal violet was removed and plates washed in water. Plates were allowed to dry and plaques counted. Three independent experiments were conducted. Data from the first experiment and summary data are presented.
  • IFN- ⁇ treatment has the potential to alter the course of established influenza infection in the lungs and thus to reduce respiratory tract illness that develops during or following an established influenza-like illness (ILI).
  • ILI influenza-like illness
  • influenza-like illness The main complication of influenza-like illness is viral pneumonia that develops following the spread of the virus to the lower respiratory tract.
  • Human IFN- ⁇ is highly species specific in its biological activity.
  • the cynomolgus macaque monkey is an appropriate species to study the effects of human IFN- ⁇ .
  • Human IFN- ⁇ has been shown to upregulate the interferon- sensitive antiviral genes myxovirus resistance protein 1 (MxA) ( Figure 3) and 2'-5' oligoadenylate synthetase (2'-5' OAS) ( Figure 4) to the same extent in cynomolgus macaque blood cells and human blood cells.
  • MxA myxovirus resistance protein 1
  • OAS 2'-5' oligoadenylate synthetase
  • the study comprises two stages.
  • successful delivery of inhaled IFN- ⁇ is established by showing upregulation of IFN ⁇ -dependent antiviral markers in lung cells obtained by broncho-alveolar lavage (BAL).
  • BAL broncho-alveolar lavage
  • Similar data in clinical studies have supported further development of inhaled IFN- ⁇ as an antiviral therapy.
  • the second stage the effect of prophylactic and therapeutic treatment with lung delivered IFN- ⁇ on influenza induced lung pathology and lung viral load is investigated.
  • Stage 1 Confirmation of successful delivery of IFN- ⁇ to the lung
  • IFN- ⁇ in solution, is administered by nebuliser on two occasions to the lungs of cynomolgus macaque monkeys.
  • BAL is collected prior to and following each dose.
  • Gene expression of IFN- -dependent antiviral markers (MxA, 2'-5' OAS and Interferon gamma-induced protein 10 kDa (IP-10) in BAL cells is determined using quantitative PCR methods. Levels of upregulation are compared to the data generated in clinical studies in order to select the dose for the next stage. The study schedule and more details of the methods are described below.
  • BAL blood and broncho-alveolar lavage
  • IFN- ⁇ nebuliser solution is delivered by inhalation using the l-neb device (Philips Respironics) coupled to a pediatric face mask (Philips Respironics, part number HS81 1 10EU-001 ).
  • the l-neb device is programmed to generate a continuous stream of aerosol when switched on.
  • IFN- ⁇ On day 8, under anaesthesia, blood is collected, weight measured and 1 .5 ml or 4.5 ml inhaled IFN- ⁇ delivered to each macaque. The dose of IFN- ⁇ selected will be dependent on the results obtained from dose 1 .
  • Stage 2 Effect of prophylactic and therapeutic treatment with lung delivered IFN- ⁇ on influenza induced lung pathology and lung viral load
  • the second stage of the study includes three treatment groups: Groupl receives placebo; Group 2 receives inhaled IFN- ⁇ treatment 24hrs prior to being infected with challenge virus and subsequent daily treatment with IFN- ⁇ ; Group 3 receives inhaled IFN- ⁇ treatment 4 hours after being infected with challenge virus and subsequent daily treatment with IFN- ⁇ .
  • Therapeutic treatments are administered 4 hours post infection as in this model a relatively high dose of virus is administered directly to the lower respiratory tract to model established lower airways disease.
  • a sample size calculation indicated that 9 animals per group is required to see a treatment effect of a 1 .5 log difference in viral titre between treated and control (untreated) animals. This is considered to be clinically significant on the basis of results of clinical studies with the neuraminidase inhibitors (Barnett et al. (2000) Antimicrob. Agents Chemother. 44: 78-87).
  • the study schedule is shown below:
  • a block design is used. The study is conducted in three blocks, with each block consisting of nine animals, three animals from each treatment group. Fourteen days prior to virus challenge nine cynomolgus macaques confirmed as seronegative for circulating influenza viruses are equipped with a preprogrammed temperature sensor in the abdominal cavity and blood is collected under ketamine and dormitor anaesthesia.
  • mice On day -1 , after full recovery from surgery, all animals are anaesthetised, weighed and samples (whole blood for serum and nasal/throat swabs) collected. Animals belonging to Groups 1 and 2 are administered nebulised placebo or IFN- ⁇ (at the dose determined in stage 1 ) under anaesthesia.
  • mice of Group 1 and 2 are treated with nebulised placebo or IFN- ⁇ as described above. All animals are then challenged intratracheally with influenza virus (10 5 TCID 50 H5N1 or 10 7 TCID 50 H1 N1 ). Four hours after intratracheal challenge animals of Group 3 are administered nebulised IFN- ⁇ as described above.
  • the main endpoints are lung viral load and lung pathology score. Throat swabs taken daily will give an indication of the time course of the viral infection. Positive data, a reduction in lung viral load or lung pathology, particularly following therapeutic treatment with IFN- ⁇ , would support the development of inhaled IFN- ⁇ for the treatment of lower respiratory tract illness caused by influenza.

Abstract

An agent selected from: (a) interferon-β (IFN-β); (b) an agent that increases IFN-β expression; or (c) a polynucleotide which is capable of expressing (a) or (b); for use in the treatment of individuals with lower respiratory tract illness that has developed during or following an established ILI, wherein said treatment is by airway delivery of said medicament.

Description

THERAPY FOR INFLUENZA LIKE ILLNESS
Field of the Invention The invention relates to the treatment of individuals with lower respiratory tract illness that has developed during or following an established influenza-like illness (ILI). By established influenza like illness, we mean an illness in which the symptoms have been apparent, e,g., to the individual or to a carer, for at least 24 hours, more particularly at least 48 hours. More specifically, the invention relates to the treatment of individuals with lower respiratory tract illness that have developed during or following an ILI, particularly hospitalized patients, by delivery of interferon-β (IFN-β) or an agent that increases IFN-β expression by aerosol to the lower respiratory tract. Influenza like illness is a disease characterised by two of the following symptoms: headache, cough, sore throat, and myalgia and/or confirmed influenza. More specifically, ILI may be defined as a fever >37.8°C plus two of the following symptoms: (headache, cough, sore throat, and myalgia) and/or confirmed influenza infection.
Background of the invention
Seasonal influenza is a common infection, especially during winter. Every year strains of influenza (type A or B) circulate, giving rise to clinical consultations in primary care, episodes of hospital treatment (mainly in older persons and young children, but occasionally in middle aged adults), and deaths (mainly in the elderly). Treatment in primary care and hospital may be required due to the direct effects of influenza virus infection or its possible complications, most commonly secondary bacterial infection. Increases in primary care consultations for ILI and winter bed pressures are frequently associated with periods of known community influenza activity.
Pandemic influenza occurs when a new influenza virus subtype emerges which is markedly different from recently circulating subtypes and strains, and is able to: • infect humans;
• spread efficiently from person to person;
• cause significant clinical illness in a high proportion of those infected. Because the virus is novel in humans, a high proportion of the population will have little or no immunity, producing a large pool of susceptible persons; accordingly the disease spreads widely and rapidly.
Interferons have previously been proposed for either prophylactic or early stage intervention in the treatment of influenza, but with limited success.
Highly pathogenic influenza is a virulent form of influenza, such as H5N1 , which leads to a rapid and high level of morbidity. ILI includes seasonal influenza, pandemic influenza and highly pathogenic influenza.
We have now found a new method of treating individuals, particularly hospitalized patients, with lower respiratory tract illness that has developed during or following an ILI.
Summary of the Invention
Accordingly, the invention provides an agent selected from:
(a) interferon-β (IFN-β);
(b) an agent that increases IFN-β expression; or
(c) a polynucleotide which is capable of expressing (a) or (b);
for use in the treatment of individuals with lower respiratory tract illness that have developed during or following an established ILI, wherein said treatment is by aerosol delivery to the lower respiratory tract of said agent.
The invention further provides a method of treating an individual diagnosed with lower respiratory tract illness that has developed during or following an established ILI comprising aerosol delivery to the lower respiratory tract of the individual of an agent selected from the group consisting of:
(a) interferon-β (IFN-β);
(b) an agent that increases IFN-β expression;
(c) a polynucleotide which is capable of expressing (a) or (b).
Also provided is the use of an agent selected from:
(a) interferon-β (IFN-β);
(b) an agent that increases IFN-β expression; or
(c) a polynucleotide which is capable of expressing (a) or (b);
In the preparation of a medicament for the treatment of lower respiratory tract illness that has developed during or following an established ILI, wherein said treatment is by aerosol delivery to the lower respiratory tract of said agent. Brief description of the Figures
Figure 1 a shows the increase in the percentage of human lung bronchial epithelial cells infected over a 96 hour period following infection with influenza A virus and the effect of IFN-β treatment from 48 hours post-infection (n=1 experiment).
Figure 1 b shows the increase in the percentage of human lung bronchial epithelial cells infected over a 96 hour period following infection with influenza A virus and the effect of IFN-β treatment from 48 hours post-infection (n=3 experiments including original data).
Figure 2a shows viral shedding over a 96 hour period following infection of human lung bronchial epithelial cells with influenza A virus and the effect of IFN-β treatment from 48 hours post-infection. Viral shedding plateaus between 48 and 96 hours. (n=1 experiment).
Figure 2b shows viral shedding over a 96 hour period following infection of human lung bronchial epithelial cells with influenza A virus and the effect of IFN-β treatment from 48 hours post-infection. Viral shedding plateaus between 48 and 96 hours. (n=3 experiments including original data).
Figure 3 shows the effect of human IFN-β treatment on the expression of the IFN- -dependent antiviral gene myxovirus resistance protein 1 (MxA) (n=3) in blood cells from healthy human volunteers or cynomolgus macaques. Whole blood was treated with IFN-β (0-1000 lU/ml) for 4 hours, RNA extracted and effect on MxA gene expression measured. Figure 4 shows the effect of human IFN-β treatment on the expression of the IFN- -dependent antiviral gene 2'-5' oligoadenylate synthetase (2'-5' OAS) (n=3) in blood cells from healthy human volunteers or cynomolgus macaques. Whole blood was treated with IFN-β (0-1000 lU/ml) for 4 hours, RNA extracted and effect on 2'-5' OAS gene expression measured.
.
Figure 5 shows the protective effect of human IFN-β in cynomolgus macaque cells infected with seasonal influenza (n=4). Cynomolgus macaque cells were pre-treated with IFN-β (0, 100 or 1000 lU/ml) for 24hrs prior to infection with the flu strain AVictoria/3/75 (H3N2) at an MOI of 0.01 . Supernatants were collected 48hrs after virus infection and viral shedding was measured by plaque assay. * indicates p<0.05.
Brief description of the Sequence Listing SEQ ID NO: 1 shows the nucleotide sequence of human IFN^I a. SEQ ID NO: 2 shows the amino acid sequence of human IFN^Ia. SEQ ID NO: 3 shows the nucleotide sequence of human IFN-βΙ b. SEQ ID NO: 4 shows the amino acid sequence of human IFN-βΙ b. IFN-βΙ b is identical to human IFN-βΙ a except for replacement of the cysteine at residue 17 with serine.
Detailed description of the invention
As hereinbefore indicated, the present invention relates to new therapeutic uses for IFN-β. In particular, it relates, for example, to therapeutic use of IFN-β by aerosol delivery to the lower respiratory tract for the treatment of individuals diagnosed as having lower respiratory tract illness that has developed during or following an influenza like illness, particularly those patients who have been admitted to hospital.
As noted above, interferons have been proposed previously for the prophylaxis or early stage treatment of ILI. Accordingly, interferons have been proposed for use either before symptoms of ILI are manifested or on the initial occurrence of such symptoms. In general, these interferons have been administered to the upper respiratory tract, eg the nasal pharynx. Usually initiation of treatment with interferons begins within 24 hours of the occurrence of symptoms.
In contrast we have found that patients who have been hospitalized with complications arising from ILI, in particular with lower respiratory tract illness may be treated by the aerosolized delivery of an interferon-β agent to the lower respiratory tract. Usually hospitalization occurs typically 48 hours after the first occurrence of symptoms.
Definition of IFN-β.
The term IFN-β as used herein will be understood to refer to any form or analog of IFN-β that retains the biological activity of native IFN-β and preferably retains the activity of IFN-β that is present in the lung and, in particular, the bronchial and/or alveolar epithelium. The IFN-β may be identical to or comprise the sequence of human IFN-βΙ a (SEQ ID NO: 2) or human IFN-βΙ b (SEQ ID NO: 4). IFN-β also refers to a variant polypeptide having an amino acid sequence which varies from that of SEQ ID NO: 2 or 4. Alternatively, IFN-β may be chemically-modified. A variant of IFN-β may be a naturally occurring variant, for example a variant which is expressed by a non-human species. Also, variants of IFN-β include sequences which vary from SEQ ID NO: 2 or 4 but are not necessarily naturally occurring. Over the entire length of the amino acid sequence of SEQ ID NO: 2 or 4, a variant will preferably be at least 80% homologous to that sequence based on amino acid identity. More preferably, the polypeptide is at least 85% or 90% and more preferably at least 95%, 97% or 99% homologous based on amino acid identity to the amino acid sequence of SEQ ID NO: 2 or 4 over the entire sequence. There may be at least 80%, for example at least 85%, 90% or 95%, amino acid identity over a stretch of 40 or more, for example 60, 80, 100, 120, 140 or 160 or more, contiguous amino acids ("hard homology"). Homology may be determined using any method known in the art. For example the UWGCG Package provides the BESTFIT program which can be used to calculate homology, for example used on its default settings (Devereux et al (1984) Nucleic Acids Research 12, p387-395). The PILEUP and BLAST algorithms can be used to calculate homology or line up sequences (such as identifying equivalent residues or corresponding sequences (typically on their default settings)), for example as described in Altschul S. F. (1993) J Mol Evol 36:290-300; Altschul, S.F et al (1990) J Mol Biol 215:403-10. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nhn.nih.gov/). This algorithm involves first identifying high scoring sequence pan: (HSPs) by identifying short words of length W in the query sequence that either match or satisfy some positive- valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighbourhood word score threshold (Altschul et al, supra). These initial neighbourhood word hits act as seeds for initiating searches to find HSP's containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Extensions for the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment. The BLAST program uses as defaults a word length (W) of 1 1 , the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1992) Proc. Natl. Acad. ScL USA 89: 10915-10919) alignments (B) of 50, expectation (E) of 10, M=5, N=4, and a comparison of both strands . The BLAST algorithm performs a statistical analysis of the similarity between two sequences; see e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-5787. One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P (N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a sequence is considered similar to another sequence if the smallest sum probability in comparison of the first sequence to the second sequence is less than about 1 , preferably less than about 0.1 , more preferably less than about 0.01 , and most preferably less than about 0.001 . Amino acid substitutions may be made to the amino acid sequence of SEQ ID NO: 1 or 2, for example from 1 , 2, 3, 4 or 5 to 10, 20 or 30 substitutions. Conservative substitutions may be made, for example, according to Table 1 . Amino acids in the same block in the second column and preferably in the same line in the third column may be substituted for each other: Table 1 - Conservative amino acid substitutions NON- AROMATIC Non-polar G A P I L V Polar - uncharged C S T M N Q Polar - charged D E H KR AROMATIC H F W Y
Table 1 - Conservative amino acid substitutions
Figure imgf000008_0001
One or more amino acid residues of the amino acid sequence of SEQ ID NO: 1 or 2 may alternatively or additionally be deleted. From 1 , 2, 3, 4 or 5 to 10, 20 or 30 residues may be deleted, or more. IFN-β also includes fragments of the above-mentioned sequences. Such fragments retain IFN-β activity. Fragments may be at least from 120 or 140 amino acids in length. Such fragments may be used to produce chimeric agents as described in more detail below. IFN-β includes chimeric proteins comprising fragments or portions of SEQ ID NO: 2 or 4. One or more amino acids may be alternatively or additionally added to the polypeptides described above. An extension may be provided at the N-terminus or C-terminus of the amino acid sequence of SEQ ID NO: 2 or 4 or polypeptide variant or fragment thereof. The extension may be quite short, for example from 1 to 10 amino acids in length. Alternatively, the extension may be longer. A carrier protein may be fused to an amino acid sequence described above. A fusion protein incorporating one of the polypeptides described above can thus be used in the invention. IFN-β also includes SEQ ID NO: 2 or 4 or variants thereof that have been chemically-modified. A number of side chain modifications are known in the art and may be made to the side chains of the proteins or peptides discussed above. Such modifications include, for example, glycosylation, phosphorylation, modifications of amino acids by reductive alkylation by reaction with an aldehyde followed by reduction with NaBH , amidination with methylacetimidate or acylation with acetic anhydride. The modification is preferably glycosylation. The IFN-β may be made synthetically or by recombinant means using methods known in the art. The amino acid sequence of proteins and polypeptides may be modified to include non- naturally occurring amino acids or to increase the stability of the compound. When the proteins or peptides are produced by synthetic means, such amino acids may be introduced during production. The proteins or peptides may also be modified following either synthetic or recombinant production. The IFN-β may also be produced using D-amino acids. In such cases the amino acids will be linked in reverse sequence in the C to N orientation. This is conventional in the art for producing such proteins or peptides. The IFN-β may be produced in a cell by in situ expression of the polypeptide from a recombinant expression vector. The expression vector optionally carries an inducible promoter to control the expression of the polypeptide. The IFN-β or analog thereof may be produced in large scale following purification by any protein liquid chromatography system after recombinant expression. Preferred protein liquid chromatography systems include FPLC, AKTA systems, the Bio-Cad system, the Bio- RadBioLogic system and the Gilson HPLC system. Commercially available forms of IFN-β or analogs thereof may be used in the invention. Examples include Betaseron® and Avonex®. Agents that increase IFN-β expression
The invention may also involve using an agent that increases endogenous expression of IFN-β in the lung or preferably the bronchial and/or alveolar epithelium. The agents may act directly on the promoter or other regulatory sequences of the IFN-β gene. Such agents may act to reduce the constitutive silencing of the IFN-β promoter. Alternatively, the agent may stimulate cells to produce endogenous IFN-β by acting at receptors at the cell surface. Agents that increase endogenous expression of IFN-β of interest in relation to the present invention include, but are not limited to, poly(inosinic acid)-poly(cytidylic acid) (poly(IC)), ANA773, perindopril, BL-20803, Tilorone, ABMP, DRB, Atabrine, 10-carboxy-9acridone, CP-28888, Bropirimine, and Imiquimod.
The invention may also involve using a polynucleotide which is capable of expressing IFN-β or an agent that increases endogenous expression of IFN-β in lung airways. Such a polynucleotide may preferably be in the form of a vector capable of directing expression of IFN-β or an agent that induces IFN-β in the bronchial and/or alveolar epithelium. The resulting IFN-β or agent may then have a therapeutic effect ("gene therapy"). The polynucleotide may encode any of the forms of IFN-β discussed above including the variants, fragments and chimeric proteins thereof. The polynucleotide encoding IFN-β may comprise the human sequence (SEQ ID NO: 1 or 3) or a naturally occurring sequence variant, for example a variant which is expressed by a non-human species. Also, a polynucleotide encoding IFN-β includes sequences which vary from SEQ ID NO: 1 or 3 but are not necessarily naturally occurring. Over the entire length of the amino acid sequence of SEQ ID NO: 1 or 3, a variant will preferably be at least 80% homologous to that sequence based on nucleotide identity. More preferably, the polynucleotide is at least 85% or 90% and more preferably at least 95%, 97% or 99% homologous based on nucleotide identity to the nucleotide of SEQ ID NO: 1 or 3 over the entire sequence. There may be at least 80%, for example at least 85%, 90% or 95%, nucleotide identity over a stretch of 40 or more, for example 60, 80, 100, 120, 140 or 160 or more, contiguous nucleotides ("hard homology"). Homology may be determined as discussed above. The polynucleotides may comprise DNA or RNA but preferably comprise DNA. They may also be polynucleotides which include within them synthetic or modified nucleotides. A number of different types of modification to polynucleotides are known in the art. These include methylphosphate and phosphorothioate backbones, addition of acridine or polylysine chains at the 3' and/or 5' ends of the molecule. For the purposes of the present invention, it is to be understood that the polynucleotides described herein may be modified by any method available in the art. Polynucleotides such as a DNA polynucleotide may be produced recombinantly, synthetically, or by any means available to those of skill in the art. They may also be cloned by standard techniques. The polynucleotides are typically provided in isolated and/or purified form. Polynucleotides will generally be produced using recombinant means, for example using PCR (polymerase chain reaction) cloning techniques. This will involve making a pair of primers (e.g. of about 15- 30 nucleotides) to a region of the required gene which it is desired to clone, bringing the primers into contact with DNA obtained from a suitable cell, performing a polymerase chain reaction under conditions which bring about amplification of the desired region, isolating the amplified fragment (e.g. by purifying the reaction mixture on an agarose gel) and recovering the amplified DNA. The primers may be designed to contain suitable restriction enzyme recognition sites so that the amplified DNA can be cloned into a suitable cloning vector. Although in general the techniques mentioned herein are well known in the art, reference may be made in particular to Sambrook et al, 1989. As hereinbefore indicated, preferably the polynucleotide is used in an expression vector wherein it is operably linked to a control sequence which is capable of providing for the expression of the coding sequence in the airways of human lung. Expression vectors for use in accordance with the invention may be any type of vector conventionally employed for gene therapy. It may be a plasmid expression vector administered as naked DNA or complexed with one or more cationic amphiphiles, e.g one or more cationic lipids, e.g. in the form of DNA/liposomes. A viral vector may alternatively be employed. Vectors for expression of therapeutic proteins in the airways of human lung have previously been described. For example, Published International Application WO 01/91800 (Isis Innovation Limited) describes for such purpose expression vectors including the human ubiquitin C promoter or functional analogues thereof. The human ubiquitin C promoter has been shown to be capable of producing high level protein expression in the airways of mice over many weeks and hence has been proposed as a favoured promoter for use in airway gene therapy for a variety of respiratory diseases. Examples of expression vectors for use in directing transgene expression in airway epithelia have also been described in Chow et al. Proc. Natl. Acad. Sci. USA 1997; 94: 14695- 14700. Such expression vectors can be administered via the airways, e.g into the nasal cavity or trachea.
The IFN-β, agent or polynucleotide may be administered in a medicament or pharmaceutical composition suitable for airway delivery which will typically also include a pharmaceutically acceptable excipient. Such an "excipient" generally refers to a substantially inert material that is nontoxic and does not interact with other components of the composition in a deleterious manner. Pharmaceutically acceptable excipients include, but are not limited to, liquids such as water, saline, polyethyleneglycol, hyaluronic acid, glycerol and ethanol. Pharmaceutically acceptable salts can be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulphates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. It is also preferred, although not required, that a composition or medicament comprising the therapeutic agent will contain a pharmaceutically acceptable carrier that serves as a stabilizer, particularly for peptide, protein, polynucleotide or other like agents. Examples of suitable carriers that also act as stabilizers for peptides include, without limitation, pharmaceutical grades of dextrose, sucrose, lactose, trehalose, mannitol, sorbitol, inositol, dextran, and the like. Other suitable carriers include, again without limitation, starch, cellulose, sodium or calcium phosphates, citric acid, tartaric acid, glycine, high molecular weight polyethylene glycols (PEGs), and combination thereof. It may also be useful to employ a charged lipid and/or detergent. Suitable charged lipids include, without limitation, phosphatidylcholines (lecithin), and the like. Detergents will typically be a nonionic, anionic, cationic or amphoteric surfactant. Examples of suitable surfactants include, for example, Tergitol® and Triton® surfactants (Union Carbide Chemicals anaplastics, Danbury, CT), polyoxyethylenesorbitans, for example, TWEEN® surfactants (Atlas Chemical Industries, Wilmington, DE), polyoxyethylene ethers, for example Brij, pharmaceutically acceptable fatty acid esters, for example, lauryl sulfate and salts thereof (SDS), and like materials. A thorough discussion of pharmaceutically acceptable excipients, carriers, stabilizers and other auxiliary substances is available in Remingtons Pharmaceutical Sciences (Mack Pub. Co., N. J.1991 ).
A suitable composition for airway delivery of IFN-β may, for example, be formulated as described in US Patent no 6,030,609 by dissolving lyophilised IFN-β in a pharmaceutically acceptable vehicle such as sterile distilled water or sterile physiological saline, optionally with addition of one or more carriers, stabilizers, surfactants or other agents in order to enhance effectiveness of the IFN-β active agent.
A suitable composition has a pH of from 5 to 8, more preferably 5.5 to 7.5. Preferably, the composition is buffered, e.g. using a citrate buffer.
The Rentschler composition, which is based on formulations disclosed in US 6,030,609, has a pH of 6.5 and osmolarity of 290 mOsm/kg. The composition is preferably provided as a sterile, clear and colourless, ready-to-use aqueous nebuliser solution presented in a disposable glass syringe.
In addition to the active ingredient, the composition preferably comprises a buffering system to maintain the pH at between 5 and 8, more preferably 5.5 and 7.5, especially 6.5. The composition also preferably comprises an antioxidant, for example, DL-methionine.
Figure imgf000014_0001
A composition comprising a therapeutically effective amount of the IFN-β, agent or polynucleotide described herein may conveniently be delivered to the lung airways by means of an inhalation device that is capable of delivering fine particles of the active ingredient to the lower respiratory tract or airways. Typically particles of the active ingredient will have a mass median diameter of 1 -10 microns. Suitable inhalation devices include dry powder inhalation (DPI) devices, pressurised metered dose inhalers (pMDI) and aerosol nebulisers.
Typically, the inhalation device will produce an aerosol with a particle size, as determined using a Malvern Masterizer S, with a mass median diameter of 1 -10 micron, preferably 3-8 micron, in which mass per cent have a diameter below 5 micron is from 25-80%, preferably 30-65%. A suitable nebulizier is the l-neb device, a CE-marked nebuliser manufactured by Philips Respironics.
An appropriate effective amount may be determined by appropriate clinical testing and will vary with for example the activity of the IFN-β administered or induced. The IFN-β, agent or polynucleotide may for example, be administered in microgram amounts.
They are administered to the subject to be treated in a manner compatible with the dosage formulation, and in an amount that will be effective to bring about the desired effect. The amount to be delivered per dose may be 0.1 g to 500ig, for example 1 to 50 g, depending on the subject to be treated.
The treatment normally lasts from 5-7 days and may continue to 14 days if symptoms persist. Treatment may be given from every other day to several times a day. Preferably, treatment is given once per day.
The IFN-β, agent or polynucleotide may be administered on its own or simultaneously, sequentially or separately in combination with another therapeutic compound. In particular, the IFN-β, agent or polynucleotide may be administered in conjunction with a therapeutic compound used to treat the respiratory disease or antiviral to the individual. The IFN-β, agent or polynucleotide and additional therapeutic compound may be formulated in the same or different compositions.
In one embodiment, the IFN-β, agent or polynucleotide is administered to an individual with asthma in combination with an inhaled corticosteroid.
In a further embodiment, the IFN-β, agent or polynucleotide may be administered simultaneously, sequentially or separately with an inhaled neuraminidase inhibitor. Thus, in a further aspect of the present invention there is provided a product for treatment of individuals diagnosed as having lower respiratory tract disease (or symptoms) that have developed during or following an influenza like illness comprising for simultaneous, separate or sequential lower respiratory tract administration (i) a first agent selected from (a) IFN-β, (b) an agent that increases IFN-β expression and (c) a polynucleotide capable of expressing (a) or (b) and (ii) an inhaled neuraminidase inhibitor. Preferably, such a product will provide for simultaneous, separate or sequential administration of IFN-β and an inhaled neuraminidase inhibitor, for example, zanamivir. A first agent as defined above and an inhaled neuraminidase inhibitor may, for example, be provided in the form of a single pharmaceutical composition suitable for aerosol delivery to the airways, e.g., by means of a dry powder inhaler, a pressurised metered dose inhaler or an aerosol nebulizer.
Alternatively, the IFN-β, agent or polynucleotide may be administered simultaneously, sequentially or separately with an oral neuraminidase inhibitor, such as oseltamivir.
Alternatively, the IFN-β, agent or polynucleotide may be administered simultaneously, sequentially or separately with a systemically administered neuraminidase inhibitor, such as peramivir. In a further embodiment, the IFN-β, agent or polynucleotide may be administered simultaneously, sequentially or separately with an inhaled influenza virus attachment inhibitor. Thus, in a further aspect of the present invention there is provided a product for treatment of individuals diagnosed as having lower respiratory tract disease (or symptoms) that have developed during or following an influenza like illness comprising for simultaneous, separate or sequential lower respiratory tract administration (i) a first agent selected from (a) IFN-β, (b) an agent that increases IFN-β expression and (c) a polynucleotide capable of expressing (a) or (b) and (ii) an inhaled influenza virus attachment inhibitor. Preferably, such a product will provide for simultaneous, separate or sequential administration of IFN-β and an influenza virus attachment inhibitor, for example, the sialidase fusion protein, DAS181 (Fludase®). A first agent as defined above and an inhaled influenza virus attachment inhibitor may, for example, be provided in the form of a single pharmaceutical composition suitable for aerosol delivery to the airways.
In a further embodiment, the IFN-β, agent or polynucleotide may be administered simultaneously, sequentially or separately with an antibacterial antibiotic. Preferably the antibacterial antibiotic is administered by inhalation. In a further embodiment, the IFN-β, agent or polynucleotide may be administered simultaneously, sequentially or separately with an antifungal antibiotic. Preferably the antifungal antibiotic is administered by inhalation.
Children younger than 5, but especially children younger than 2 years old, adults 65 years of age and older, pregnant women and those with comorbidities (e.g. chronic lung disease, neurological and neurodevelopmental conditions, heart disease, blood disorders, endocrine disorders, kidney disorders, liver disorders, metabolic disorders, malignancy and weakened immune system due to disease or medication) are at greatest risk of developing complications resulting in hospitalisation and have the poorest outcomes and represent a particular target group for the invention. Elderly individuals, for example those more than 60 years of age are a particular target group, as are patients suffering from asthma and/or COPD.
A further at risk group are patients who have not previously been exposed to an ILL Example 1
The ability of exogenous IFN-β to suppress influenza A infection in an in vitro model of established infection of the lower respiratory tract is illustrated in the following example.
Overview
Lower respiratory tract disease (or symptoms) that develop during or following an influenza-like illness (ILI) are often precipitated by the spread of virus infections from the upper to the lower respiratory tract. Furthermore, prolonged symptoms and, in hospitalised patients, extended hospital stays are associated with persistent viral shedding in the lower airways. Using human lung bronchial epithelial cells, an important site of influenza replication in man, we have developed a model of established influenza A infection of the lower respiratory tract. The effect of IFN-β in this model was investigated.
Method
Cell culture and infection protocol
Human lung bronchial epithelial (HBE) cells were cultured in minimum essential medium (MEM) containing glutamax (Invitrogen), 10% fetal bovine serum (FBS) (Invitrogen), penicillin (50 U/ml) and streptomycin (50 pg/ml) (Invitrogen). HBE cells were plated at 1 .5x105/well, passage 41 -49, in a 24 well plate. At approximately 60-70% confluency, the cells were cultured in reduced serum medium (MEM containing glutamax (Invitrogen) and 0.75-1 % FBS (Invitrogen)) for 24 hours. Cells were infected with an activated influenza strain (influenza A/WSN/33 (H1 N1 ) (Retroscreen)) at 0.0001 MOI at 37°C. After one hour, unbound virus was removed by washing. Reduced serum medium was added and the cells were incubated at 37°C for a further four days. Cells were treated with 1000IU/nnl of human IFN- i a (Rentschler) 48 hrs post-infection with virus. Once IFN-β treatment commenced the dose was repeated every 24 hrs. Flow cytometry to determine percentage influenza positive cells
Cell media was collected at 24, 48, 72, and 96 hours post-infection and stored at -80°C. At 24, 48, 72 and 96 hours post-infection cells were trypsinised, counted and washed in phosphate buffered saline (PBS). Up to 2x105 cells were taken for analysis by flow cytometry. The cells were centrifuged at 400g and the supernatant discarded. Live/dead cell stain (Molecular Probes) was reconstituted in dimethyl sulfoxide (DMSO) (500 μΙ) and diluted 1 :100 in PBS. Diluted live/dead cell stain (100 μΙ) was incubated with cells at room temperature for 30 minutes. Cells were washed in PBS, centrifuged at 400g, and the supernatant discarded. 250 μΙ Cytofix/Cytoperm (BD Biosciences) was added per tube and the cells left at 4°C for 20 minutes. Cells were washed twice with 2 ml Perm/Wash (BD Biosciences) solution by centrifugation at 500g. After the second wash, cells were re-suspended in 100 μΙ Perm/Wash solution and left at room temperature for 15 minutes. Anti-influenza A matrix protein antibody (Serotec) was diluted to 2.5 pg/rril in Perm/Wash solution and 100 μΙ added to cells for 30 minutes at room temperature. Cells were washed twice with 2 ml Perm/Wash solution by centrifugation at 500g . Anti-mouse IgG-FITC (Sigma) was diluted to 5 g/ml in Perm/Wash solution and 100 μΙ added to cells for 30 minutes at room temperature protected from light. Cells were washed twice with 2 ml Perm/Wash solution by centrifugation at 500g. Cells were resuspended in Perm/Wash solution and analysed by flow cytometry using the FACSCalibur (BD Biosciences) and CellQuest Pro software. Data was subsequently analysed using WinMDI software.
Plaque assay to determine viral shedding
Madin Darby canine kidney (MDCK) cells were cultured in MEM containing glutamax (Invitrogen), 10% FBS (Invitrogen), non-essential amino acids (Invitrogen), and penicillin/streptomycin (Invitrogen). MDCK cells were plated at 1 x105 cells/well, passage 25-30, in a 12 well plate. On reaching 100% confluence cells were washed twice with PBS. Cell media collected during the infection experiment was diluted in serum-free media (DMEM (Invitrogen), penicillin/streptomycin (Invitrogen), L-glutamine (Invitrogen), Non-essential amino acids (Invitrogen), and sodium pyruvate (Invitrogen)) to give a dilution range of ten-fold dilutions between 1 :10 to 1 :106. Diluted cell media (200 μΙ) was added to duplicate wells. Plates were incubated at 37°C for 1 hr. Virus was aspirated and cells washed with PBS. Overlay medium (100 ml 10 times concentrated MEM (Invitrogen), 20 ml 7.5% sodium bicarbonate (Sigma), 10 ml 1 M HEPES (Fluka), 28 ml 7.5% bovine serum albumin (BSA) fraction V (Sigma), 5 ml 1 % DEAE-dextran (Sigma), 20 ml penicillin/streptomycin (Invitrogen), 10 ml 200 mM glutamine (Invitrogen), 507 ml d.H2O) was prepared and 17.5 ml added to 12.5 μΙ 1 mg/ml trypsin treated with L-(tosylamido-2- phenyl) ethyl chloromethyl ketone (TRTPCK trypsin) (Worthington Biochemical Corp.) and 7.5 ml Avicel suspension (4.8g Avicel (FMC Biopolymer) in 186 ml d.H2O). The overlay was mixed by inverting and 1 ml added per well. Plates were incubated for 2 days without moving or shaking. Overlay medium was aspirated and cells washed twice with PBS. Crystal violet solution was added (0.5 ml) (0.65g crystal violet (Sigma), 25 ml formaldehyde (Sigma), 25 ml 100% ethanol (Sigma), 450 ml 1 xPBS (5 DulA PBS tablets (Sigma) + 500 ml d.H20)) for 30 minutes at room temperature. Crystal violet was removed and plates washed in water. Plates were allowed to dry and plaques counted. Three independent experiments were conducted. Data from the first experiment and summary data are presented.
Data analysis
Data was analysed using Graphpad Prism software
Results
In this model flow cytometric analysis suggests that between 5 and 40% of cells are infected between 48 and 96 hours (Refer to Figures 1 a and 1 b) which is similar to the levels of productively infected cells reported in the clinical setting at peak infection (Baccam et al. (2006) Journal of Virology, 80: 7590-7599). Correspondingly this period is also associated with highest levels of virus shedding in our model (Figure 2a and 2b).
Treatment of cells with IFN-β at 48 hours post-infection, modelling treatment of established infection, caused a large reduction in the proportion of infected cells and viral shedding (Figures 1 a, 1 b, 2a and 2b). A reduction in viral load of greater than 10 fold is considered to be clinically relevant (Barnett et al. (2000) Antimicrob. Agents Chemother 44: 78-87). Conclusion
These results show that IFN-β treatment has the potential to alter the course of established influenza infection in the lungs and thus to reduce respiratory tract illness that develops during or following an established influenza-like illness (ILI). As elderly patients and those suffering from asthma and/or COPD are likely to have difficulty in making IFN-β when needed, the uses and methods described herein are of particular application to such vulnerable groups. In vivo study:
Introduction
The main complication of influenza-like illness is viral pneumonia that develops following the spread of the virus to the lower respiratory tract.
Human IFN-β is highly species specific in its biological activity. The cynomolgus macaque monkey is an appropriate species to study the effects of human IFN-β. Human IFN-β has been shown to upregulate the interferon- sensitive antiviral genes myxovirus resistance protein 1 (MxA) (Figure 3) and 2'-5' oligoadenylate synthetase (2'-5' OAS) (Figure 4) to the same extent in cynomolgus macaque blood cells and human blood cells. Furthermore in in vitro studies human IFN-β protects cynomolgus macaque cells from infection with influenza (Figure 5). Preclinical models of lower respiratory tract illness have been developed using the cynomolgus macaque monkey for highly pathogenic influenza A (H5N1 ) (Rimmelzwaan et al. (2001 ) J Virol. 75: 6687-91 ) and pandemic 2009 influenza A (H1 N1 ) (Herfst et al. (2010) Vet. Pathol. 47: 1040-7) viruses. These models have predicted the clinical efficacy of neuraminidase inhibitors against influenza when administered prior to infection.
This study investigates the effect of lung delivered IFN-β in a similar cynomolgus macaque monkey model of influenza induced lower respiratory tract illness. The challenge virus strains are A lndonesia/5/05 (H5N1 ) and A Netherlands/602/2009 (pandemic H1 N1 ) which have both been shown to replicate in the lower respiratory tract and cause significant lung pathology (Herfst et al. (2010) Vet. Pathol. 47: 1040-7; Rimmelzwaan et al. (2001 ) J Virol. 75: 6687-91 ).
The study comprises two stages. In the first stage successful delivery of inhaled IFN-β is established by showing upregulation of IFN^-dependent antiviral markers in lung cells obtained by broncho-alveolar lavage (BAL). Similar data in clinical studies have supported further development of inhaled IFN-β as an antiviral therapy. In the second stage the effect of prophylactic and therapeutic treatment with lung delivered IFN-β on influenza induced lung pathology and lung viral load is investigated. Stage 1 : Confirmation of successful delivery of IFN-β to the lung
In brief, IFN-β, in solution, is administered by nebuliser on two occasions to the lungs of cynomolgus macaque monkeys. BAL is collected prior to and following each dose. Gene expression of IFN- -dependent antiviral markers (MxA, 2'-5' OAS and Interferon gamma-induced protein 10 kDa (IP-10) in BAL cells is determined using quantitative PCR methods. Levels of upregulation are compared to the data generated in clinical studies in order to select the dose for the next stage. The study schedule and more details of the methods are described below.
Synairgen Interferon Beta Proof of Succesful Delivery Study j
Figure imgf000022_0001
Figure imgf000022_0002
Seven days prior to administration of inhaled IFN-β, five male cynomolgus macaques, aged approximately three years, are anaesthetized with ketamine- dormitor, have blood and broncho-alveolar lavage (BAL) collected and their weight measured. BAL involves flushing the lungs with 10 mL phosphate- buffered saline with an envisaged recovery of approximately 5 mL. On each occasion BAL samples are separated into supernatant and cellular fraction. The cellular fractions are lysed in RLT buffer (Qiagen) and analysed for the IFN- -dependent antiviral biomarkers MxA, 2'-5' OAS, and IP-10 using quantitative PCR methods. On day 0, under anaesthesia, blood is collected, weight measured and 3 ml inhaled IFN-β (Rentschler) delivered to each macaque. IFN-β nebuliser solution is delivered by inhalation using the l-neb device (Philips Respironics) coupled to a pediatric face mask (Philips Respironics, part number HS81 1 10EU-001 ). The l-neb device is programmed to generate a continuous stream of aerosol when switched on.
On day 1 , under anaesthesia, blood and BAL are collected and weight measured.
On day 8, under anaesthesia, blood is collected, weight measured and 1 .5 ml or 4.5 ml inhaled IFN-β delivered to each macaque. The dose of IFN-β selected will be dependent on the results obtained from dose 1 .
On day 9, under anaesthesia, blood and BAL is collected and weight measured.
Stage 2: Effect of prophylactic and therapeutic treatment with lung delivered IFN-β on influenza induced lung pathology and lung viral load
The second stage of the study includes three treatment groups: Groupl receives placebo; Group 2 receives inhaled IFN-β treatment 24hrs prior to being infected with challenge virus and subsequent daily treatment with IFN-β; Group 3 receives inhaled IFN-β treatment 4 hours after being infected with challenge virus and subsequent daily treatment with IFN-β. Therapeutic treatments are administered 4 hours post infection as in this model a relatively high dose of virus is administered directly to the lower respiratory tract to model established lower airways disease. A sample size calculation indicated that 9 animals per group is required to see a treatment effect of a 1 .5 log difference in viral titre between treated and control (untreated) animals. This is considered to be clinically significant on the basis of results of clinical studies with the neuraminidase inhibitors (Barnett et al. (2000) Antimicrob. Agents Chemother. 44: 78-87). The study schedule is shown below:
Figure imgf000024_0002
Figure imgf000024_0003
Figure imgf000024_0001
Due to the number of animals included in the study a block design is used. The study is conducted in three blocks, with each block consisting of nine animals, three animals from each treatment group. Fourteen days prior to virus challenge nine cynomolgus macaques confirmed as seronegative for circulating influenza viruses are equipped with a preprogrammed temperature sensor in the abdominal cavity and blood is collected under ketamine and dormitor anaesthesia.
On day -1 , after full recovery from surgery, all animals are anaesthetised, weighed and samples (whole blood for serum and nasal/throat swabs) collected. Animals belonging to Groups 1 and 2 are administered nebulised placebo or IFN-β (at the dose determined in stage 1 ) under anaesthesia.
On day 0 animals of Group 1 and 2 are treated with nebulised placebo or IFN-β as described above. All animals are then challenged intratracheally with influenza virus (105 TCID50 H5N1 or 107 TCID50 H1 N1 ). Four hours after intratracheal challenge animals of Group 3 are administered nebulised IFN-β as described above.
On days 1 , 2, 3, and 4 all animals are anaesthetised, weighed, samples collected (whole blood for serum and nasal/throat swabs) and treated with nebulised placebo or IFN-β as described above.
Five days after challenge all animals are anaesthetised, weighed and euthanized by exsanguinations. Full gross pathology is performed, all major organs inspected and lung lesions described. Samples are collected from all animals including: whole blood for serum, nasal/throat swabs and tissue from the right lung. Swabs collected throughout the experiment and sections of the right lung are subjected to quantitative PCR and virus titration to determine viral load. The left lung is inflated with 10% formalin for subsequent histopathological assessment.
The main endpoints are lung viral load and lung pathology score. Throat swabs taken daily will give an indication of the time course of the viral infection. Positive data, a reduction in lung viral load or lung pathology, particularly following therapeutic treatment with IFN-β, would support the development of inhaled IFN-β for the treatment of lower respiratory tract illness caused by influenza.

Claims

Claims
1 . An agent selected from:
(a) interferon-β (IFN-β);
(b) an agent that increases IFN-β expression; or
(c) a polynucleotide which is capable of expressing (a) or (b); for use in the treatment of individuals with lower respiratory tract illness that has developed during or following an established ILI , wherein said treatment is by airway delivery of said medicament.
2. An agent according to claim 1 wherein said agent is IFN-β.
3. An agent according to claim 2 wherein the IFN- β comprises the sequence of: (a) human IFN^I a (SEQ ID NO : 2); or (b) human IFN- 1 b (SEQ ID NO : 4).
4. An agent according to any one of the preceding claims in combination with a neuraminidase inhibitor.
5. An agent according to claim 4, wherein the neuraminidase inhibitor is topically active.
6. An agent according to claim 4 or claim 5, wherein the neuraminidase inhibitor is zanamivir.
7. An agent according to any one of claims 1 to 6, wherein the lower respiratory tract illness is asthma or COPD.
8. An agent according to any of claims 1 to 7, wherein the individual is elderly, for example, more than 60 years of age.
9. A method of treating an individual diagnosed with a lower respiratory tract illness that has developed during or following an established ILI comprising airway administration to the individual of an agent selected from the group consisting of :
(a) interferon-β (IFN-β);
(b) an agent that increases IFN-β expression;
(c) a polynucleotide which is capable of expressing (a) or (b).
10. A method according to claim 9 wherein said agent is IFN-β
1 1 . A method according to claim 10, wherein the IFN-β comprises the sequence of :
(a) human IFN- β (SEQ ID NO: 2); or (b) human IFN- pi b (SEQ ID NO: 4).
12. A method according to any one of claims 9 to 1 1 wherein said agent is combined with a neuraminidase inhibitor for simultaneous, separate or sequential administration.
13. A method according to any of claims 9 to 12, wherein the lower respiratory tract illness is asthma or COPD.
14. A method according to any one of claims 9 to 13, wherein the individual is elderly, for example, more than 60 years of age.
15. A product for treatment of an influenza like illness comprising for simultaneous, separate or sequential airway administration (i) a first agent selected from (a) to (c) as defined in claim 1 and (ii) an inhaled neuraminidase inhibitor
16. A product as claimed in claim 15 which is a pharmaceutical composition for aerosol delivery to the airways, especially the lower airways.
17. A product as claimed in claim 15 or claim 16 wherein said first agent is IFN- β-
18. An agent according to claims 1 to 8 or a method according to any one of claims 9 to 14, wherein the agent is delivered to the lower respiratory tract by aerosol at least 48 hours after ILI symptoms become apparent.
PCT/GB2011/050480 2010-03-12 2011-03-10 Therapy for influenza like illness WO2011110861A2 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
DK11730046.7T DK2544705T3 (en) 2010-03-12 2011-03-10 INTERFERON BETA TO USE IN THE LOWER AIR TRANSMISSION CAUSED BY INFLUENZA
EP11730046.7A EP2544705B1 (en) 2010-03-12 2011-03-10 Interferon beta for use in the treatment of lower respiratory tract illness caused by influenza
US13/583,976 US9421243B2 (en) 2010-03-12 2011-03-10 Therapy for influenza like illness
RS20160753A RS55154B1 (en) 2010-03-12 2011-03-10 Interferon beta for use in the treatment of lower respiratory tract illness caused by influenza
SI201130956A SI2544705T1 (en) 2010-03-12 2011-03-10 Interferon beta for use in the treatment of lower respiratory tract illness caused by influenza
LTEP11730046.7T LT2544705T (en) 2010-03-12 2011-03-10 Interferon beta for use in the treatment of lower respiratory tract illness caused by influenza
CN2011800135925A CN102883741A (en) 2010-03-12 2011-03-10 Therapy for influenza like illness
ES11730046.7T ES2592528T3 (en) 2010-03-12 2011-03-10 Interferon beta for use in the treatment of lower respiratory diseases caused by influenza
CA2792727A CA2792727C (en) 2010-03-12 2011-03-10 Therapy for influenza like illness
JP2012557607A JP5926200B2 (en) 2010-03-12 2011-03-10 Treatment of influenza-like diseases
HRP20161158TT HRP20161158T1 (en) 2010-03-12 2016-09-08 Interferon beta for use in the treatment of lower respiratory tract illness caused by influenza
CY20161100897T CY1117985T1 (en) 2010-03-12 2016-09-12 INTERFERON BETA FOR USE IN THE DISEASE TREATMENT OF THE BREATH-RESPIRATORY SYSTEM
SM201600334T SMT201600334B (en) 2010-03-12 2016-09-21 INTERFERONE BETA FOR THE USE IN THE TREATMENT OF THE LOWER RESPIRATORY TREATMENT DISEASE CAUSED BY INFLUENCE

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GBGB1004144.0A GB201004144D0 (en) 2010-03-12 2010-03-12 Therapy for influenza like illness
GB1004144.0 2010-03-12
GB1008114.9 2010-05-17
GBGB1008114.9A GB201008114D0 (en) 2010-05-17 2010-05-17 Therapy for influenza like illness

Publications (2)

Publication Number Publication Date
WO2011110861A2 true WO2011110861A2 (en) 2011-09-15
WO2011110861A3 WO2011110861A3 (en) 2011-11-24

Family

ID=44563925

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/050480 WO2011110861A2 (en) 2010-03-12 2011-03-10 Therapy for influenza like illness

Country Status (17)

Country Link
US (1) US9421243B2 (en)
EP (1) EP2544705B1 (en)
JP (1) JP5926200B2 (en)
CN (2) CN102883741A (en)
CA (1) CA2792727C (en)
CY (1) CY1117985T1 (en)
DK (1) DK2544705T3 (en)
ES (1) ES2592528T3 (en)
HR (1) HRP20161158T1 (en)
HU (1) HUE029250T2 (en)
LT (1) LT2544705T (en)
PL (1) PL2544705T3 (en)
PT (1) PT2544705T (en)
RS (1) RS55154B1 (en)
SI (1) SI2544705T1 (en)
SM (1) SMT201600334B (en)
WO (1) WO2011110861A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022018422A1 (en) * 2020-07-20 2022-01-27 Synairgen Research Limited Inhaled interferon-beta for improving outcome in sars-cov-2 infected patients

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB202014114D0 (en) 2020-09-08 2020-10-21 Synairgen Res Ltd Use of inhaled interferon-beta to treat virus-induced exacerbations in copd patients undergoing treatment with a systemic corticosteroid

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6030609A (en) 1995-05-19 2000-02-29 Case Western Reserve University Method and composition for treating paramyxovirus
WO2001091800A1 (en) 2000-05-30 2001-12-06 Isis Innovation Limited Ubiquitin promoter in vectors for gene therapy in respiratory tract

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20010540A1 (en) * 1999-07-30 2001-05-15 Procter & Gamble COMPOSITION OF STYLBENE PHYTOALEXINS USEFUL FOR PROPHYLAXIS AND TREATMENT OF SYMPTOMS ASSOCIATED WITH COLD AND INFLUENZA-SIMILAR DISEASES
TW200509961A (en) * 2003-05-23 2005-03-16 Pestka Biomedical Lab Inc Uses of interferons for the treatment of severe acute respiratory syndrome and other viral infections
WO2004108151A1 (en) 2003-06-09 2004-12-16 Genome Institute Of Singapore Inhibition of sars coronavirus infection with clinically approved antiviral drugs
CN101001644B (en) * 2003-08-28 2011-09-21 辉阳科技美国公司 Uses of spatial configuration to modulate protein function
GB0518425D0 (en) * 2005-09-09 2005-10-19 Imp College Innovations Ltd Methods
GB0405634D0 (en) * 2004-03-12 2004-04-21 Univ Southampton Anti-virus therapy for respiratory diseases
CN1927388B (en) * 2004-09-10 2011-02-02 北京金迪克生物技术研究所 Medicinal composition containing human interferon
CN1927389B (en) * 2004-09-10 2012-11-14 北京金迪克生物技术研究所 Application of human interferon containing pharmaceutical composition in preparation of medicine for preventing and treating virus infection of respiratory tract
EP1843782A1 (en) * 2005-02-04 2007-10-17 ViraNative AB Method and use of interferon compositions for the treatment of avian influenza
CA2622016A1 (en) 2005-09-09 2007-03-15 Imperial Innovations Limited Interferon lambda therapy for treatment of respiratory diseases
WO2007057436A2 (en) 2005-11-18 2007-05-24 Ares Trading S.A. Interferon in influenza
GB0609410D0 (en) * 2006-05-12 2006-06-21 Viragen Inc Method for the production of a type 1 interfemon in a transgenic avian
CA2589613A1 (en) * 2007-05-18 2008-11-18 Synairgen Plc Interferon-beta and/or lambda for use in treating rhinovirus infection in the elderly
WO2008143892A1 (en) * 2007-05-18 2008-11-27 New York University Method of treating tuberculosis with interferons

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6030609A (en) 1995-05-19 2000-02-29 Case Western Reserve University Method and composition for treating paramyxovirus
WO2001091800A1 (en) 2000-05-30 2001-12-06 Isis Innovation Limited Ubiquitin promoter in vectors for gene therapy in respiratory tract

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
"Remingtons Pharmaceutical Sciences", 1991, MACK PUB. CO.
ALTSCHUL S. F., J MOL EVOL, vol. 36, 1993, pages 290 - 300
ALTSCHUL, S.F ET AL., J MOL BIOL, vol. 215, 1990, pages 403 - 10
BACCAM ET AL., JOURNAL OF VIROLOGY, vol. 80, 2006, pages 7590 - 7599
BARNETT ET AL., ANTIMICROB. AGENTS CHEMOTHER, vol. 44, 2000, pages 78 - 87
CHOW ET AL., PROC. NATL. ACAD. SCI. USA, vol. 94, 1997, pages 14695 - 14700
DEVEREUX ET AL., NUCLEIC ACIDS RESEARCH, vol. 12, 1984, pages 387 - 395
HENIKOFF, HENIKOFF, PROC. NATL. ACAD. SCL USA, vol. 89, 1992, pages 10915 - 10919
HERFST ET AL., VET. PATHOL, vol. 47, 2010, pages 1040 - 7
HERFST ET AL., VET. PATHOL., vol. 47, 2010, pages 1040 - 7
KARLIN, ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 5787
RIMMELZWAAN ET AL., J VIROL., vol. 75, 2001, pages 6687 - 91

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022018422A1 (en) * 2020-07-20 2022-01-27 Synairgen Research Limited Inhaled interferon-beta for improving outcome in sars-cov-2 infected patients

Also Published As

Publication number Publication date
JP2013522282A (en) 2013-06-13
WO2011110861A3 (en) 2011-11-24
DK2544705T3 (en) 2016-09-19
CN102883741A (en) 2013-01-16
HUE029250T2 (en) 2017-04-28
SI2544705T1 (en) 2016-11-30
CN107050431A (en) 2017-08-18
PT2544705T (en) 2016-09-21
RS55154B1 (en) 2017-01-31
EP2544705A2 (en) 2013-01-16
JP5926200B2 (en) 2016-05-25
EP2544705B1 (en) 2016-08-31
CA2792727C (en) 2019-06-04
HRP20161158T1 (en) 2016-11-18
LT2544705T (en) 2016-10-10
PL2544705T3 (en) 2017-02-28
ES2592528T3 (en) 2016-11-30
US9421243B2 (en) 2016-08-23
SMT201600334B (en) 2016-11-10
CA2792727A1 (en) 2011-09-15
CY1117985T1 (en) 2017-05-17
US20130064792A1 (en) 2013-03-14

Similar Documents

Publication Publication Date Title
EP2206512A2 (en) Increase of interferon-beta for anti-virus therapy for respiratory diseases
CN111346219B (en) Use of interferon in preparing medicine for preventing coronavirus infection or preventing diseases caused by coronavirus infection
EP3022298B1 (en) Attenuated influenza vaccines and uses thereof
EP4117711A2 (en) Lactoferrin for oral use with antiviral action
KR101872218B1 (en) Composition comprising a peptide and an inhibitor of viral neuraminidase
US20230079150A1 (en) Methods for prevention or treatment of virus-induced organ injury or failure with il-22 dimer
KR20220147121A (en) Soluble ACE2 and fusion proteins, and their applications
AU2010306840A1 (en) Recombinant human CC10 protein for treatment of influenza
CN113425832A (en) Use of interferon lambda in the treatment of infections with novel coronaviruses (2019-nCoV)
CN111671886A (en) Pharmaceutical composition for preventing high-risk susceptible people from infecting coronavirus or generating coronavirus infection disease and application of pharmaceutical composition
US9421243B2 (en) Therapy for influenza like illness
CN112220913A (en) Use of TFF2 protein in combination with IFN-kappa protein for the treatment of novel coronavirus infections
CN115843267A (en) Treatment of respiratory viral infections
WO2021195883A1 (en) APPLICATION OF TFF2 PROTEIN AND IFN-κ PROTEIN COMBINATION IN TREATMENT OF A NOVEL CORONAVIRUS INFECTION
WO2005016247A2 (en) Dna sequences, peptides, antibodies and vaccines for prevention and treatment of sars
WO2023202711A1 (en) Mrna vaccine based on novel coronavirus
KR20120047780A (en) Vaccine composition for respiratory syncytial virus and manufacturing method thereof
WO2023104199A1 (en) Use of soluble receptor for advanced glycation end products protein for preventing or treating pulmonary infection diseases
TW202334430A (en) Alpha-1 antitrypsin produced from yeast for use in the treatment of viral infections
WO1995025539A1 (en) Novel remedy for respiratory-tract viral disease
EP4331571A1 (en) Formulations of ace2-igm fusion proteins
WO2022096708A1 (en) A composition comprising extra-cellular vesicles from mesenchymal stem cells and alpha-1 antitrypsin for the treatment of viral infections
WO2004096852A1 (en) A RECOMBINANT HUMAN INTERFERON ϖ, THE METHOD FOR EXPRESSING IT AND THE USES OF IT
CN116472054A (en) Inhaled interferon-beta for improving prognosis of SARS-CoV-2 infected patient

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180013592.5

Country of ref document: CN

ENP Entry into the national phase

Ref document number: 2792727

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2012557607

Country of ref document: JP

REEP Request for entry into the european phase

Ref document number: 2011730046

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011730046

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13583976

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: P-2016/0753

Country of ref document: RS