WO2011029931A1 - Composition for treatment of cxcl8-mediated lung inflammation - Google Patents

Composition for treatment of cxcl8-mediated lung inflammation Download PDF

Info

Publication number
WO2011029931A1
WO2011029931A1 PCT/EP2010/063389 EP2010063389W WO2011029931A1 WO 2011029931 A1 WO2011029931 A1 WO 2011029931A1 EP 2010063389 W EP2010063389 W EP 2010063389W WO 2011029931 A1 WO2011029931 A1 WO 2011029931A1
Authority
WO
WIPO (PCT)
Prior art keywords
modified
lung
amino acid
inflammation
asthma
Prior art date
Application number
PCT/EP2010/063389
Other languages
French (fr)
Inventor
Andreas Kungl
Jason Slingsby
Tiziana Adage
Angelika Rek
Original Assignee
Protaffin Biotechnologie Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Protaffin Biotechnologie Ag filed Critical Protaffin Biotechnologie Ag
Priority to JP2012528380A priority Critical patent/JP2013504545A/en
Priority to AU2010294225A priority patent/AU2010294225A1/en
Priority to CN2010800508273A priority patent/CN102596227A/en
Priority to CA2773664A priority patent/CA2773664A1/en
Priority to US13/395,128 priority patent/US20120288474A1/en
Priority to EA201200470A priority patent/EA201200470A1/en
Priority to EP10751694A priority patent/EP2475380A1/en
Publication of WO2011029931A1 publication Critical patent/WO2011029931A1/en
Priority to ZA2012/01802A priority patent/ZA201201802B/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
    • A61K38/20Interleukins [IL]
    • 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
    • C07K14/54Interleukins [IL]
    • C07K14/5421IL-8
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • composition for treatment of CXCL8-mediated lung inflammation Composition for treatment of CXCL8-mediated lung inflammation
  • the present invention relates to a new use of modified interleukin 8 (IL-8, CXCL8) having increased GAG binding affinity and further inhibited or down-regulated receptor binding activity compared to the respective wild type IL-8 for preventing or treating lung inflammation with neutrophilic infiltration, specifically for the prevention or treatment of CXCL8-mediated lung inflammation.
  • modified IL-8 as inhalant is provided.
  • Lung inflammatory diseases are of particular relevance in view of their predominance in the human population and the lack of efficacious therapy. Specifically, lung diseases shown to have increased infiltration of neutrophils are chronic
  • obstructive pulmonary disease cystic fibrosis, chronic severe asthma and acute lung injury with its more severe form, acute respiratory distress syndrome.
  • COPD chronic obstructive pulmonary disease
  • the inflammatory response observed in lungs of patients with COPD is complex and involves the activation of both innate and acquired immune responses; however it is clear that disease progression is dominated by leukocyte migration, the production of pro-inflammatory cytokines and chemokines and release of potentially destructive proteases (Kim et al. 2008).
  • neutrophils have been shown to be the most abundant inflammatory cells in lungs of COPD patients, both in sputum and bronchoalveolar lavage (BAL) samples (Nocker et al. 1996; Peleman et al. 1999).
  • CXCL8 levels are significantly elevated in sputum and BAL of COPD patients at different stage of disease progression (between 10-15 fold increase vs. healthy ) and correlate with disease severity and neutrophil presence (Yamamoto et al. 1997; Tanino et al. 2002), identifying CXCL8 as the key chemokine involved in neutrophil mobilization (Woolhouse et al. 2002).
  • elevated levels of CXCL8 are also present in sputum of COPD patients during exacerbations (Aaron et al. 2001 ; Spruit et al. 2003).
  • CF Cystic fibrosis
  • CXCL-8 Cystic fibrosis
  • Several studies have documented increased levels of CXCL-8 in BAL and sputum and increased expression of CXCL8 in bronchial glands of patients with CF (Nakamura et al. 1992; Tabary et al. 1998). Its potent neutrophil chemoattractant properties stimulate the influx of massive numbers of neutrophils in the airways (Chmiel et al. 2002).
  • Bacterial infection are further increasing CXCL8 levels, driving more neutrophils infiltration into the lungs and creating a vicious circle difficult to interrupt and resulting in chronic lung inflammation. Acting on this vicious circle with treatments acting on CXCL8-induced inflammation, such as PA401 , can result the most effective treatment for CF patients, which currently rely only on supportive therapy with bronchodilator and mucolytics or antibiotics.
  • asthmatics About one in 10 asthmatics patients present the severe form of the disease, which frequently requires progressively higher doses of steroids in an attempt to control symptoms. Severe asthma is also associated with a much higher risk of illness and death than milder forms.
  • corticosteroid resistant asthma Green et al 2002
  • nocturnal asthma Martin et al 1991
  • asthma in smokers Choalmers et al. 2001
  • occupational asthma Anees et al.2002
  • chronic neutrophilic severe asthma presents a clear different clinical phenotype, rather than an increased presence in asthma symptoms and share features with COPD (The ENFUMOSA study group).
  • epithelial cell-derived CXCL8 is the most likely candidate as the predominant neutrophil chemoattractant (Lamblin et al. 1998; Ordonez et al. 2000), and potential candidate for the development of new antiinflammatory therapies.
  • the aim of the present invention is therefore to provide a method for the prevention or treatment of lung inflammatory pathologies that are infiltrated with neutrophils.
  • the invention is based on the discovery that modified interleukin 8 (IL-8) having increased GAG binding affinity and inhibited or down-regulated GPCR (G-protein coupled receptor, i.e. CXCR1 and CXCR2) activity compared to the respective wild type IL-8 can be used for the prevention and treatment of lung inflammation with neutrophilic infiltration.
  • IL-8 modified interleukin 8
  • G-protein coupled receptor G-protein coupled receptor, i.e. CXCR1 and CXCR2
  • CXCR1 and CXCR2 G-protein coupled receptor
  • modified IL-8 Although the use of modified IL-8 was already described briefly for the treatment of "normal" asthma lacking high levels of neutrophil infiltration in the lung, the success- ful use of said modified IL-8 molecules for the treatment and prevention of lung inflammation with neutrophil infiltration has not been shown or indicated before.
  • the anti-asthma activity of said modified IL-8 molecules might result from non-specific or consecutive displacement of other, asthma-related, chemokines such as eotaxin.
  • the inhibited or down-regulated activity is at least a reduction or complete lack of neutrophil activation by GPCR activation.
  • Subject matter of the present invention is therefore to provide a modified IL-8 having increased GAG binding affinity and inhibited or down-regulated GPCR activity compared to the respective wild type IL-8 for use for the prevention or treatment of lung inflammatory diseases with neutrophilic infiltration in individuals.
  • Fig. 1 Dose-response effect of PA401 on total cell infiltrates in bronchoalveolar lavages of mice instilled with LPS.
  • Fig. 2 Dose-response effect of PA401 on neutrophils number in cytospin of bronchoalveolar lavages of mice instilled with LPS.
  • Fig. 3 Dose-response effect of PA401 on lymphocytes number in cytospin of bronchoalveolar lavages of mice instilled with LPS.
  • Fig. 4 Dose-response effect of PA401 on total cell infiltrates in bronchoalveolar lavages of mice aerosolized with LPS.
  • Fig. 5 Dose-response effect of PA401 on neutrophils number in cytospin of bronchoalveolar lavages of mice aerosolized with LPS.
  • Fig. 6 Dose-response effect of PA401 (Fig. 6a) and Roflumilast (Fig. 6b) on total cell infiltrates in bronchoalveolar lavages of mice exposed for 4 days to cigarette smoke.
  • Fig. 7 Dose-response effect of PA401 (Fig. 7a) and Roflumilast (Fig. 7b) on neutrophil infiltrates in bronchoalveolar lavages of mice exposed for 4 days to cigarette smoke.
  • Fig. 8 Dose-response effect of PA401 (Fig. 8a) and Roflumilast (Fig. 8b) on macrophage infiltrates in bronchoalveolar lavages of mice exposed for 4 days to cigarette smoke.
  • Fig. 9 Dose-response effect of PA401 (Fig. 9a) and Roflumilast (Fig. 9b) on epithelial cell infiltrates in bronchoalveolar lavages of mice exposed for 4 days to cigarette smoke.
  • Fig. 10 Dose-response effect of PA401 (Fig. 10a) and Roflumilast (Fig. 10b) on lymphocyte infiltrates in bronchoalveolar lavages of mice exposed for 4 days to cigarette smoke.
  • Fig. 1 1 Effect on total cell infiltrates after twice a day, daily and every other day administration of PA401 (Fig. 1 1 a) and and comparison with Roflumilast (Fig. 1 1 b).
  • Fig. 12 Reduction of neutrophils at all treatment frequencies by PA401 (Fig. 12a) and comparison with Roflumilast (Fig. 12b).
  • Fig. 13 Reduction of epithelial cells at all treatment frequencies by PA401 (Fig. 13a) and comparison with Roflumilast (Fig. 13b).
  • Fig. 14 Loss of significant reduction in lymphocyte obtained with b.i.d. and q.d. administration when PA401 was administered every other day (Fig. 14a) and comparison with Roflumilast (Fig. 14b).
  • Fig. 15 Loss of significant reduction in macrophage obtained with b.i.d. and q.d. administration when PA401 (Fig. 15a) was administered every other day. Comparison with Roflumilast (Fig. 15b).
  • Fig. 16 Effect of PA401 (Fig. 16a) on total cell infiltrates observed after 400 and 40 g/kg administration. Comparison with Roflumilast (Fig. 16b).
  • Fig. 17 Reduction in neutrophil numbers in BAL (PA401 , Fig. 17a and
  • Fig. 18 Reduction in epithelial cell numbers in BAL (PA401 , Fig. 18a and Roflumilast, Fig. 18b)
  • Fig. 19 Reduction in lymphocyte numbers in BAL (PA401 , Fig. 19a and
  • Fig. 20 Reduction in macrophage numbers in BAL (PA401 , Fig. 20a and Roflumilast, Fig. 20b)
  • Fig. 21 Dose-response activity on PA401 intra-tracheal administration 1 hour before (-1 h) and 1 hour after (+1 h) LPS aerosol exposure on total cell count in the bronchoalveolar lavage collected 8 hours post LPS. ANOVA followed by Dunnett ' s test: * p ⁇ 0.05; ** p ⁇ 0.01 versus vehicle treated animals.
  • Fig. 22 Dose-response activity on PA401 intra-tracheal administration 1 hour before (-1 h) and 1 hour after (+1 h) LPS aerosol exposure on neutrophils count in the bronchoalveolar lavage collected 8 hours post LPS. ANOVA followed by Dunnett ' s test: * p ⁇ 0.05; ** p ⁇ 0.01 versus vehicle treated animals.
  • the present invention covers a modified interleukin 8 (IL-8) having increased GAG binding affinity and further inhibited or down-regulated GPCR activity compared to the respective wild type IL-8 for the use for the prevention or treatment of lung inflammation with neutrophilic infiltration.
  • the modified IL-8 molecule as used in the present invention for the treatment of lung inflammation with neutrophil infiltration is modified in the GAG (glycosaminoglycan) binding region leading to increased affinity towards GAG, the IL-8-specific GAG ligand.
  • .Modification can be either in the naturally occurring GAG binding region or, alternatively, a new GAG binding region can be introduced in said molecule resulting in increased affinity towards GAG.
  • an artifical and/or improved GAG binding site is introduced in said protein.
  • an overall more electronegative molecular character can be introduced into the chemokine.
  • the main purpose is to increase the relative amount of basic or electron donating amino acids, preferably Arg, Lys, His, Asn and/or Gin, compared to the total amount of amino acids in said site, whereby the resulting GAG binding site should preferably comprise at least 3 basic amino acids, still preferred at least 4, most preferred at least 5 amino acids.
  • the GAG binding site is a C-terminal alpha helix which is modified to increase GAG binding affinity.
  • IL-8 refers to amino acids with long or sterically interfering side chains; these are in particular Trp, lie, Leu, Phe, Tyr.
  • the GAG binding site on the chemokine is free of bulky amino acids to allow optimal induced fit by the GAG ligand.
  • positions 17, 21 , 70, and/or 71 in IL-8 are substituted by Arg, Lys, His, Asn and/or Gin.
  • all four positions 17, 21 , 70 and 71 of IL-8 are substituted by Arg, Lys, His, Asn and/or Gin, preferably by Lys.
  • the modified IL-8 as used in the present invention comprises also an inhibited or down-regulated receptor binding region, specifically the GPCR (G-protein coupled receptor) binding.
  • GPCR G-protein coupled receptor
  • the GAG binding affinity is higher than in the wild-type GAG binding protein, so that the modified protein will to a large extent bind to the GAG instead of the wild-type protein.
  • the GPCR activity of the wild-type protein which mainly occurs when the protein is bound to GAG, is inhibited or down-regulated, since the modified protein will not carry out this specific activity or carries out this activity to a lesser extent.
  • the receptor binding region can be modified by deletion, insertion, and/or substitution, for example with alanine, a sterically and/or electrostatically similar residue. It is possible to either delete or insert or substitute at least one amino acid in a receptor binding region.
  • said GPCR binding region is located within the first 10 N-terminal amino acids.
  • the first N-terminal amino acids are involved in leukocyte activation, whereby in particular Glu-4, Leu-5 and Arg-6 were identified to be essential for receptor binding and activation. Therefore, either these three or even up to the first 10 N-terminal amino acids can be substituted or deleted in order to inhibit or down- regulate the receptor binding and activation.
  • the modified IL-8 can have the first 6 N- terminal amino acids deleted.
  • this mutant will not or to a lesser extent bind and activate leukocytes and/or promote neutrophil activation, so that it is particularly suitable for the treatment of organ transplant rejection.
  • the modified IL-8 is selected from the group consisting of
  • del6F17RE70KN71 R del6F17RE70RN71 K, del6E70KN71 K, del6F17RE70RN71 K, and del6F17KF21 KE70KN71 K.
  • amino acid sequence of the modified IL 8 molecule is preferably described by the general formula:
  • X2 is of amino acid sequence CQCI
  • X3 is selected of the group consisting of R, K, H, N and/or Q, preferably it is R,
  • X4 is selected of the group consisting of R, K, H, N and/or Q,
  • X5 is selected of the group consisting of R, K, H, N and/or Q, preferably it is K,
  • X6 is selected of the group consisting of R, K, H, N and/or Q, preferably it is K,
  • n and/or m can be either 0 or 1
  • sequence of the modified IL-8 molecule is as follows:
  • the modified IL-8 is similar or identical to modified IL-8 as disclosed in WO 05/054285.
  • the administration of the composition may be by intravenous, intramuscular or subcutaneous route.
  • Other routes of administration which may establish the desired blood levels of the respective ingredients such as systemic administration or inhalation, are also comprised.
  • the modified IL-8 can be formulated as inhalant and can be administered by an inhalation system as known in the art.
  • the modified IL-8 can be formulated as liquid, aerosol or powder.
  • the medicament comprising the composition according to the invention can be formulated together with a pharmaceutically acceptable carrier.
  • “Pharmaceutically acceptable” is meant to encompass any carrier, which does not interfere with the effectiveness of the biological activity of the active ingredient and that is not toxic to the host to which is administered.
  • the modified IL-8 may be formulated in unit dosage form for injection in vehicles such as saline, dextrose solution, serum albumin and Ringer's solution.
  • additives such as stabilisers, excipients, buffers and preservatives can be included.
  • the modified IL-8 comprising composition is used to prepare a medicament to prevent or treat any lung inflammatory diseases which are characterized by neutrophil infiltration. More specifically these diseases can be for example chronic obstructive pulmonary disease, cystic fibrosis, severe asthma, bronchitis, broncheolitis, acute lung injury and acute respiratory distress syndrome.
  • the modified IL-8 is used for therapy of COPD.
  • prevention or treatment of neutrophilic asthma or exacerbations is specifically covered using modified IL-8 according to the description.
  • any lung inflammation can be treated or prevented which is induced by LPS inhalation since LPS is one of the major factors inducing IL-8 expression (Chemokines and chemokine receptors in infectious diseases. (Mahalingam S, Karupiah G., Immunol Cell Biol. 1999 Dec;77(6):469-75). LPS is a component of the walls in Gram-negative bacteria and is therefore present when gram negative bacterial infections occur or is present in air pollutant and in the tobacco leaves (so in cigarette smoke as well).
  • a method for treatment of lung inflammation with neutrophilic infiltration in a subject in need thereof comprising administering to the subject a therapeutically effective amount of modified IL-8 is covered by the invention.
  • the administration is by inhalation or by intratracheal administration.
  • COPD Chronic obstructive pulmonary disease
  • neutrophils have been shown to be the most abundant neutrophils
  • Cigarette smoke is considered to be responsible for elevation of circulating neutrophils, probably due to increased mobilization from the bone marrow (Cowburn et al. 2008), and their sequestration to the lung capillaries were they exit the pulmonary circulation. This feature is peculiar for the pulmonary circulation, since in the systemic circulation neutrophils exit at the level of postcapillary venules.
  • Neutrophils are then mobilized to the bronchial walls and lung parenchyma (Peleman at al. 1999; Kim et al. 2008). Activation of neutrophils with the subsequent release of reactive oxygen species and elastase is considered the leading cause for the development of lung damage and chronic dysfunction. Indeed, blood neutrophilia has been since long correlated with the rate of decline in lung functions measured in term of forced expiratory volume (FEV; Sparrow et al. 1984).
  • FEV forced expiratory volume
  • CXCL8 levels are significantly elevated in sputum and BAL of COPD patients at different stage of disease progression (between 10-15 fold increase vs. healthy) and correlate with disease severity and neutrophil presence (Yamamoto et al. 1997; Tanino et al. 2002), identifying CXCL8 as the key chemokine involved in neutrophil
  • Cystic fibrosis is a severe monogenic disorder of ion transport in exocrine glands, with different mutations in the CF transmembrane conductance regulator (CFTR) gene leading to impaired epithelial chloride secretion (Riordan 1989; Ratjen 2009). Dehydration and plugging of mucous secretions in the ducts of exocrine glands predispose to multi-organ clinical manifestations, particularly in the gastrointestinal, hepatobiliary, reproductive and respiratory tracts.
  • CFTR CF transmembrane conductance regulator
  • CF patients Chronic bacterial infections and inflammation of the lung are the main causes of morbidity and mortality in CF patients (Ratjen 2006). With increasing age, CF patients develop airway obstruction and many of these patients also suffer from airway hyper- responsiveness and asthma-like symptoms.
  • neutrophils from CF children display a higher migratory responsiveness to CXCL8 in vitro compared to those of non CF, suggesting that persistent elevated CXCL8 levels can "prime" CF neutrophils (Brennan et al. 2001 ).
  • Eosinophil inflammation has for long been considered the most distinctive pathological hallmark of asthma (Bousquet 1990). However, eosinophil inflammation is present in the airways of only 50% of asthmatic patients (Douwes et al. 2002), and often not observed in asthma exacerbations.
  • eosinophils and neutrophils are coexisting (Wenzel et al 1999), but it is suggested that neutrophils are becoming the predominant cell population over time. Their presence in the airways is in proportion to disease severity and progression (Wenzel et al 1997) and is associated to airflow obstruction and reduced lung function (Shaw et al. 2007). Neutrophils are also the main leukocyte population observed in a very severe and often lethal form of asthma characterized by sudden-onset (sudden-onset asthma; Sur et al. 1993). Acute lung injury (ALI) and acute respiratory distress syndrome (ARSD)
  • ALI acute lung injury
  • ARSD acute respiratory distress syndrome
  • the pathology results in a very high mortality rate (about 40%) with no trend in decreasing in the last decade, despite improved intensive care intervention (Puha et al. 2009).
  • glycosaminoglycans are the main component of the non- fibrillar compartment of the interstitium, and are located in the sub-epithelial tissue and in the bronchial walls, as well as in the airways secretions. Their presence is essential to regulate hydration and water homeostasis, maintain tissue structure, and modulate inflammatory responses (e.g. rev. Souza-Fernandes et al. 2006).
  • glycosaminoglycans including heparin/heparan sulphate, chondroitin/dermatan sulfate, keratin sulfate, and hyaluronan are present in normal lungs. Heparan sulfate has been reported as the predominant form (-40%), followed by chondroitin/dermatan sulphate (-31 %) and to minor extent to hyaluronan (-14%) and keratin sulfate (Frevert et al. 2003).
  • PA401 del6F17KF21 KE70KN71 K IL-8 mutant
  • LPS lipopolysaccharide
  • LPS instilled intranasal, or aerosolized induces a dose and time dependent neutrophil infiltration in the lung vasculature, interstitium and BAL (Reutershan et al. 2005), with peak levels reached between 4-8 hours post challenge, and remaining significantly above baseline for up to 24 hours in mice.
  • the LPS dose administered varies based on the LPS serotype, the method of application and the strain of the mice used, with significant BAL neutrophilia reported for doses of LPS as low as 0.1 pg/mouse and up to 800Mg/mouse.
  • LPS inhalation is able to induce lung neutrophil infiltration across species (e.g. mice and rats, Chapman et al.2007; guinea pigs; Wu et al. 2002; rabbits, Smith et al. 2008; sheep, Waerhaug et al. 2009; horses, van den Hoven et al. 2006; dogs, Koshika et al. 2001 ).
  • Inhalation of 1 to 100 g of LPS in healthy volunteers is regarded as robust and reliable model for acute lung inflammation (Maris et al. 2005, Kitz et al. 2008) as well as chronic obstructive lung disease exacerbation (Kharitonov et al, 2007).
  • PA401 induced a dose-dependent reduction in the total number of cells infiltrated in lung as assessed in the BAL fluid (Fig.1 ).
  • the effect was due to a significant reduction in the number of neutrophils (Fig. 2) and lymphocytes (Fig.3), with significant effect for dose of PA401 as low as 4 g/kg.
  • the inhibition of cell infiltration in the BAL obtained with PA401 at the doses of 200 and 400 g/kg was comparable to that obtained with the treatment with a high dose of Dexamethasone (3mg/kg). This study was conducted at Argenta Discovery Ltd, UK.
  • PA401 induced a highly significant reduction in the number of total cells in the BAL (Fig. 4), due to reduction in neutrophils count (Fig.5).
  • the activity of PA401 was more significant when administered by intravenous than by
  • PA401 effects in an acute model of cigarette smoke induced lung inflammation Acute exposure of mice to cigarette smoke leads to lung responses that, at least in part, mimic the lung inflammation observed in COPD patients.
  • Different mouse strains present variable degree of lung inflammation following acute cigarette smoke exposure (Guerrassimov et al. 2004, Vlahos et al. 2006).
  • This genetic variability in the response in mice appears quite representative of the variable susceptibility to develop COPD among human smokers, and therefore this model is considered the most relevant to model the human pathology.
  • Lung inflammation was induced in C57BL/6J female mice (a susceptible strain) by exposure to cigarette smoke of 4 to 6 cigarette over a four-day period.
  • This study demonstrates activity of PA401 on mixed cell infiltration induced by 4-day repeated exposure to cigarette smoke, and animal model predictive of antiinflammatory activities in COPD patients.
  • PA401 effects in a sub-chronic model of cigarette smoke induced lung
  • PA401 effects in a sub-chronic model of cigarette smoke induced lung inflammation.
  • lung inflammation was induced in C57BL/6J female mice by exposure to cigarette smoke of 4 to 6 cigarettes over an eleven-day period.
  • PA401 at the dose of 400Mg/kg daily subcutaneous was compared to the dose of 40pg/kg daily subcutaneous.
  • Treatment was performed at t+3h and effects on cell infiltrates on bronchoalveolar lavages were evaluated 24h after last cigarette smoke exposure.
  • PA401 effects in lung inflammation after local delivery to the lung.
  • lung inflammation was induced by the delivery of LPS by aerosol (Salmonella enterica, 3.5mg/7mL over 30min) in male Balb/c mice.
  • PA401 was administered intra-tracheally (i.t.) using a MicroSprayer (FJM-250 syringe;
  • PennCentury a device that allows delivery of a plume of aerosol (mass median diameter 16-22 ⁇ ) directly into the lungs.
  • PA401 i.t. administration was performed at the doses of 100, 40, 10 and 4 g/kg either 1 h before LPS exposure, or 1 h after the end of LPS exposure.
  • Bronchoalveolar lavage (BAL) was performed 8h post LPS exposure and total cell and neutrophil numbers were measured.
  • Dexamethasone 20 g/20 l mouse i.t. was used as reference compound.
  • PA401 induced a significant dose dependent reduction of total cells in BAL (Fig. 21 ), mainly due to neutrophil number reduction (Fig. 22).
  • Fig.21 shows the dose-response activity on PA401 intra-tracheal administration 1 hour before (-1 h) and 1 hour after (+1 h) LPS aerosol exposure on total cell count in the bronchoalveolar lavage collected 8 hours post LPS.
  • Fig.22 shows the dose-response activity on PA401 intra-tracheal administration 1 hour before (-1 h) and 1 hour after (+1 h) LPS aerosol exposure on neutrophils count in the bronchoalveolar lavage collected 8 hours post LPS. ANOVA followed by
  • Cowburn SC Condliffe AM, Farhai N et al. Advances in neutrophil biology. Clinical Implications. Chest 2008; 134: 606-612.
  • Kharitonov SA Sjobring U. Lypopolysaccharide challenge in humans as a model for chronic obstructive lung disease exacerbations. Contrib Microbiol 2007; 14: 83-100.
  • Lamblin C Gosset P, Tillie-Leblond I et al. Bronchial neutrophilia in patients with non infectious status asthmaticus. Am J Respir Crit Care Med 1998; 157: 394-402.
  • McDougall CM Helmes PJ. Neutrophil airway inflammation in childhood asthma. Thorax 2006; 61 (9): 739-741 .
  • Maris NA de VOS AF, Dessing MC et al. Antiinflammatory effects of salmeterol after inhalation of lipopolysaccharide by healthy volunteers. Am J Respir Crit Care Med 2005; 172(7): 878-884.
  • Ratjen FA Diagnosing and managing infection in CF. Paediatr Respir rev 2006; 7 (suppM ): S151 -S153.
  • Ratjen FA Cystic fibrosis: pathogenesis and future treatment strategies. Respir
  • Tanino M Betsuyaku T, Takeyabu K, et al. Increased levels of interleukin-8 in BAL fluid from smokers susceptible to pulmonary emphysema. Thorax 2002; 57: 405- 41 1 .
  • the ENFUMOSA study group The ENFUMOSA cross-sectional European multicentre study of the clinical phenotype of severe asthma. Eur Respir J 2003; 22- 470-477.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Zoology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pulmonology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Toxicology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Rheumatology (AREA)
  • Epidemiology (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Pain & Pain Management (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention provides a composition comprising a modified interleukin 8 (IL-8) having increased GAG binding affinity and further inhibited or down-regulated GPCR activity compared to the respective wild type IL-8 for use in preventing or treating lung inflammation with neutrophilic infiltration, for example for the prevention or treatment of chronic obstructive pulmonary disease, cystic fibrosis, severe asthma, bronchitis, broncheolitis, acute lung injury and acute respiratory distress syndrome.

Description

Composition for treatment of CXCL8-mediated lung inflammation
The present invention relates to a new use of modified interleukin 8 (IL-8, CXCL8) having increased GAG binding affinity and further inhibited or down-regulated receptor binding activity compared to the respective wild type IL-8 for preventing or treating lung inflammation with neutrophilic infiltration, specifically for the prevention or treatment of CXCL8-mediated lung inflammation. Specifically the use of modified IL-8 as inhalant is provided.
Background of the invention:
Lung inflammatory diseases are of particular relevance in view of their predominance in the human population and the lack of efficacious therapy. Specifically, lung diseases shown to have increased infiltration of neutrophils are chronic
obstructive pulmonary disease, cystic fibrosis, chronic severe asthma and acute lung injury with its more severe form, acute respiratory distress syndrome.
Chronic obstructive pulmonary disease (COPD) is a progressive debilitating disease which is predicted to become the third leading cause of death worldwide by 2020 (Lopez et al. 1998). Cigarette smoke has been established as the most important etiological factor for its development, however only 15 to 20% of smokers develop
COPD, suggesting that genetic component and other environmental factors play a role in the pathogenesis of the disease.
The inflammatory response observed in lungs of patients with COPD is complex and involves the activation of both innate and acquired immune responses; however it is clear that disease progression is dominated by leukocyte migration, the production of pro-inflammatory cytokines and chemokines and release of potentially destructive proteases (Kim et al. 2008).
In particular, neutrophils have been shown to be the most abundant inflammatory cells in lungs of COPD patients, both in sputum and bronchoalveolar lavage (BAL) samples (Nocker et al. 1996; Peleman et al. 1999). CXCL8 levels are significantly elevated in sputum and BAL of COPD patients at different stage of disease progression (between 10-15 fold increase vs. healthy ) and correlate with disease severity and neutrophil presence (Yamamoto et al. 1997; Tanino et al. 2002), identifying CXCL8 as the key chemokine involved in neutrophil mobilization (Woolhouse et al. 2002). Further, elevated levels of CXCL8 are also present in sputum of COPD patients during exacerbations (Aaron et al. 2001 ; Spruit et al. 2003).
However, current therapies are acting more as supportive and symptomatic care, than having an anti-inflammatory activity. The drugs used for the management of COPD based on the recommendation of the World Health Organization and GOLD include short and long acting β2^οηί5ί; short and long acting anticholinergic agents; methylxanthine, inhaled or systemic glucocosteroids (Pauwels et al., 2005; Lenfant and Khaltaev 2005). These therapies have some effects on controlling acute exacerbation, but treatment with traditional glucocorticosteroids results largely ineffective and fails in attenuate inflammation in patients with COPD (Culpitt et al. 1999; Fitzgerald et al. 2007), highlighting the need of the development of new antiinflammatory strategies (Fabbri et al 2004).
Another lung disease characterized by neutrophilic inflammation is Cystic fibrosis (CF). Several studies have documented increased levels of CXCL-8 in BAL and sputum and increased expression of CXCL8 in bronchial glands of patients with CF (Nakamura et al. 1992; Tabary et al. 1998). Its potent neutrophil chemoattractant properties stimulate the influx of massive numbers of neutrophils in the airways (Chmiel et al. 2002). Bacterial infection are further increasing CXCL8 levels, driving more neutrophils infiltration into the lungs and creating a vicious circle difficult to interrupt and resulting in chronic lung inflammation. Acting on this vicious circle with treatments acting on CXCL8-induced inflammation, such as PA401 , can result the most effective treatment for CF patients, which currently rely only on supportive therapy with bronchodilator and mucolytics or antibiotics.
About one in 10 asthmatics patients present the severe form of the disease, which frequently requires progressively higher doses of steroids in an attempt to control symptoms. Severe asthma is also associated with a much higher risk of illness and death than milder forms.
A strong association has now been established between neutrophilic
inflammation and chronic severe asthma (Little et al. 2002; Wenzel et al. 1997, Jatakanon et al 1999, Ordonez at al. 2000, Kamath et al. 2005; Fahy 2009), childhood asthma (McDougall et al. 2006), asthma exacerbations (Fahy et al. 1995),
corticosteroid resistant asthma (Green et al 2002), nocturnal asthma (Martin et al 1991 ), asthma in smokers (Chalmers et al. 2001 ) and occupational asthma (Anees et al.2002). It is now more and more recognised that chronic neutrophilic severe asthma presents a clear different clinical phenotype, rather than an increased presence in asthma symptoms and share features with COPD (The ENFUMOSA study group). Also in the case of chronic severe asthma, epithelial cell-derived CXCL8 is the most likely candidate as the predominant neutrophil chemoattractant (Lamblin et al. 1998; Ordonez et al. 2000), and potential candidate for the development of new antiinflammatory therapies. The aim of the present invention is therefore to provide a method for the prevention or treatment of lung inflammatory pathologies that are infiltrated with neutrophils.
SUMMARY OF THE INVENTION
The invention is based on the discovery that modified interleukin 8 (IL-8) having increased GAG binding affinity and inhibited or down-regulated GPCR (G-protein coupled receptor, i.e. CXCR1 and CXCR2) activity compared to the respective wild type IL-8 can be used for the prevention and treatment of lung inflammation with neutrophilic infiltration. Especially in COPD and in COPD exacerbations, where in- creased levels of IL-8 are present and correlate with disease progression and severity (Yamamoto et al. 1997; Tanino et al. 2002), a therapeutic intervention targeting the key chemokine involved in neutrophil mobilization (Woolhouse et al. 2002) should provide beneficial anti-inflammatory activity. Moreover, current treatments for these patients rely on supportive and symptomatic care, while application of traditional glucocortico- steroids proved to be largely ineffective (Culpitt et al. 1999; Fitzgerald et al. 2007), highlighting the need of the development of new anti-inflammatory strategies (Fabbri et al. 2004, de Boer et al. 2007).
Although the use of modified IL-8 was already described briefly for the treatment of "normal" asthma lacking high levels of neutrophil infiltration in the lung, the success- ful use of said modified IL-8 molecules for the treatment and prevention of lung inflammation with neutrophil infiltration has not been shown or indicated before. The anti-asthma activity of said modified IL-8 molecules might result from non-specific or consecutive displacement of other, asthma-related, chemokines such as eotaxin. The inhibited or down-regulated activity is at least a reduction or complete lack of neutrophil activation by GPCR activation. Although it has long been established that CXCL8-mediated lung inflammation or IL-8 induced neutrophil infiltration is significant in specific lung diseases, it has not been reported or suggested that said modified IL-8, a "protein-based GAG antagonist" would have such efficacy in the prevention or treatment of lung inflammatory diseases with neutrophilic infiltration.
Subject matter of the present invention is therefore to provide a modified IL-8 having increased GAG binding affinity and inhibited or down-regulated GPCR activity compared to the respective wild type IL-8 for use for the prevention or treatment of lung inflammatory diseases with neutrophilic infiltration in individuals.
Figures
Fig. 1 Dose-response effect of PA401 on total cell infiltrates in bronchoalveolar lavages of mice instilled with LPS.
Fig. 2 Dose-response effect of PA401 on neutrophils number in cytospin of bronchoalveolar lavages of mice instilled with LPS.
Fig. 3 Dose-response effect of PA401 on lymphocytes number in cytospin of bronchoalveolar lavages of mice instilled with LPS.
Fig. 4 Dose-response effect of PA401 on total cell infiltrates in bronchoalveolar lavages of mice aerosolized with LPS.
Fig. 5 Dose-response effect of PA401 on neutrophils number in cytospin of bronchoalveolar lavages of mice aerosolized with LPS.
Fig. 6 Dose-response effect of PA401 (Fig. 6a) and Roflumilast (Fig. 6b) on total cell infiltrates in bronchoalveolar lavages of mice exposed for 4 days to cigarette smoke.
Fig. 7 Dose-response effect of PA401 (Fig. 7a) and Roflumilast (Fig. 7b) on neutrophil infiltrates in bronchoalveolar lavages of mice exposed for 4 days to cigarette smoke.
Fig. 8 Dose-response effect of PA401 (Fig. 8a) and Roflumilast (Fig. 8b) on macrophage infiltrates in bronchoalveolar lavages of mice exposed for 4 days to cigarette smoke. Fig. 9 Dose-response effect of PA401 (Fig. 9a) and Roflumilast (Fig. 9b) on epithelial cell infiltrates in bronchoalveolar lavages of mice exposed for 4 days to cigarette smoke.
Fig. 10 Dose-response effect of PA401 (Fig. 10a) and Roflumilast (Fig. 10b) on lymphocyte infiltrates in bronchoalveolar lavages of mice exposed for 4 days to cigarette smoke.
Fig. 1 1 : Effect on total cell infiltrates after twice a day, daily and every other day administration of PA401 (Fig. 1 1 a) and and comparison with Roflumilast (Fig. 1 1 b).
Fig. 12: Reduction of neutrophils at all treatment frequencies by PA401 (Fig. 12a) and comparison with Roflumilast (Fig. 12b).
Fig. 13: Reduction of epithelial cells at all treatment frequencies by PA401 (Fig. 13a) and comparison with Roflumilast (Fig. 13b).
Fig. 14: Loss of significant reduction in lymphocyte obtained with b.i.d. and q.d. administration when PA401 was administered every other day (Fig. 14a) and comparison with Roflumilast (Fig. 14b).
Fig. 15: Loss of significant reduction in macrophage obtained with b.i.d. and q.d. administration when PA401 (Fig. 15a) was administered every other day. Comparison with Roflumilast (Fig. 15b).
Fig. 16: Effect of PA401 (Fig. 16a) on total cell infiltrates observed after 400 and 40 g/kg administration. Comparison with Roflumilast (Fig. 16b).
Fig. 17: Reduction in neutrophil numbers in BAL (PA401 , Fig. 17a and
Roflumilast, Fig. 17b)
Fig. 18: Reduction in epithelial cell numbers in BAL (PA401 , Fig. 18a and Roflumilast, Fig. 18b)
Fig. 19: Reduction in lymphocyte numbers in BAL (PA401 , Fig. 19a and
Roflumilast, Fig. 19b)
Fig. 20: Reduction in macrophage numbers in BAL (PA401 , Fig. 20a and Roflumilast, Fig. 20b)
Fig. 21 Dose-response activity on PA401 intra-tracheal administration 1 hour before (-1 h) and 1 hour after (+1 h) LPS aerosol exposure on total cell count in the bronchoalveolar lavage collected 8 hours post LPS. ANOVA followed by Dunnett's test: *p<0.05; **p<0.01 versus vehicle treated animals.
Fig. 22 Dose-response activity on PA401 intra-tracheal administration 1 hour before (-1 h) and 1 hour after (+1 h) LPS aerosol exposure on neutrophils count in the bronchoalveolar lavage collected 8 hours post LPS. ANOVA followed by Dunnett's test: *p<0.05; **p<0.01 versus vehicle treated animals.
DETAILED DESCRIPTION OF THE INVENTION CXCL8-mediated lung inflammation can lead to neutrophil infiltration in the lung of patients.
The present invention covers a modified interleukin 8 (IL-8) having increased GAG binding affinity and further inhibited or down-regulated GPCR activity compared to the respective wild type IL-8 for the use for the prevention or treatment of lung inflammation with neutrophilic infiltration. The modified IL-8 molecule as used in the present invention for the treatment of lung inflammation with neutrophil infiltration is modified in the GAG (glycosaminoglycan) binding region leading to increased affinity towards GAG, the IL-8-specific GAG ligand. .Modification can be either in the naturally occurring GAG binding region or, alternatively, a new GAG binding region can be introduced in said molecule resulting in increased affinity towards GAG. By substituting at least one naturally occurring amino acid against an amino acid, preferably a basic or electron donating amino acid, and/or substituting at least one bulky and/or acidic amino acid in the GAG binding region, an artifical and/or improved GAG binding site is introduced in said protein. By this means, an overall more electronegative molecular character can be introduced into the chemokine.
The main purpose is to increase the relative amount of basic or electron donating amino acids, preferably Arg, Lys, His, Asn and/or Gin, compared to the total amount of amino acids in said site, whereby the resulting GAG binding site should preferably comprise at least 3 basic amino acids, still preferred at least 4, most preferred at least 5 amino acids. This leads to a chemokine-based GAG antagonist competing wtlL-8 off from its HSPG co-receptor.
According to a specific embodiment the GAG binding site is a C-terminal alpha helix which is modified to increase GAG binding affinity.
The term "bulky amino acid" refers to amino acids with long or sterically interfering side chains; these are in particular Trp, lie, Leu, Phe, Tyr. Preferably, the GAG binding site on the chemokine is free of bulky amino acids to allow optimal induced fit by the GAG ligand. Advantageously, positions 17, 21 , 70, and/or 71 in IL-8 are substituted by Arg, Lys, His, Asn and/or Gin. Most preferred, all four positions 17, 21 , 70 and 71 of IL-8 are substituted by Arg, Lys, His, Asn and/or Gin, preferably by Lys.
The modified IL-8 as used in the present invention comprises also an inhibited or down-regulated receptor binding region, specifically the GPCR (G-protein coupled receptor) binding. Inactivation by protein engineering according to the present invention therefore leads to an IL-8 molecule which is reduced in promoting neutrophil activation or incapable of promoting neutrophil activation.
This means for the entire approach, that on the one hand the GAG binding affinity is higher than in the wild-type GAG binding protein, so that the modified protein will to a large extent bind to the GAG instead of the wild-type protein. On the other hand, the GPCR activity of the wild-type protein which mainly occurs when the protein is bound to GAG, is inhibited or down-regulated, since the modified protein will not carry out this specific activity or carries out this activity to a lesser extent.
The receptor binding region can be modified by deletion, insertion, and/or substitution, for example with alanine, a sterically and/or electrostatically similar residue. It is possible to either delete or insert or substitute at least one amino acid in a receptor binding region.
In the used modified IL-8 said GPCR binding region is located within the first 10 N-terminal amino acids. The first N-terminal amino acids are involved in leukocyte activation, whereby in particular Glu-4, Leu-5 and Arg-6 were identified to be essential for receptor binding and activation. Therefore, either these three or even up to the first 10 N-terminal amino acids can be substituted or deleted in order to inhibit or down- regulate the receptor binding and activation.
For example, the modified IL-8 can have the first 6 N- terminal amino acids deleted. As mentioned above, this mutant will not or to a lesser extent bind and activate leukocytes and/or promote neutrophil activation, so that it is particularly suitable for the treatment of organ transplant rejection.
Preferably, the modified IL-8 is selected from the group consisting of
del6F17RE70KN71 R, del6F17RE70RN71 K, del6E70KN71 K, del6F17RE70RN71 K, and del6F17KF21 KE70KN71 K.
The amino acid sequence of the modified IL 8 molecule is preferably described by the general formula:
(X1 )n(X2)m KTYSKP(X3)HPK (X4)IKELRVIES GPHCANTEII VKLSDGRELC LDPKENWVQR WEKFLKRA(X5) (X6)S wherein X1 is of amino acid sequence SAKELR,
wherein X2 is of amino acid sequence CQCI,
wherein X3 is selected of the group consisting of R, K, H, N and/or Q, preferably it is R,
wherein X4 is selected of the group consisting of R, K, H, N and/or Q,
wherein X5 is selected of the group consisting of R, K, H, N and/or Q, preferably it is K,
wherein X6 is selected of the group consisting of R, K, H, N and/or Q, preferably it is K,
and wherein n and/or m can be either 0 or 1
In a preferred embodiment the sequence of the modified IL-8 molecule is as follows:
SAKELRCQCI KTYSKPFHPK FIKELRVIES GPHCANTEII VKLSDGRELC LDPKENWVQR WEKFLKRAE NS wherein the first 6 amino acids (SAKELR) are deleted. Preferably, the modified IL-8 is similar or identical to modified IL-8 as disclosed in WO 05/054285.
The administration of the composition may be by intravenous, intramuscular or subcutaneous route. Other routes of administration, which may establish the desired blood levels of the respective ingredients such as systemic administration or inhalation, are also comprised.
Specifically it has been shown that local delivery to the lung, preferably inhalation or intratracheal administration is an advantageous administration mode. Therefore the modified IL-8 can be formulated as inhalant and can be administered by an inhalation system as known in the art. The modified IL-8 can be formulated as liquid, aerosol or powder.
The medicament comprising the composition according to the invention can be formulated together with a pharmaceutically acceptable carrier.
"Pharmaceutically acceptable" is meant to encompass any carrier, which does not interfere with the effectiveness of the biological activity of the active ingredient and that is not toxic to the host to which is administered. For example, for parenteral administration, the modified IL-8 may be formulated in unit dosage form for injection in vehicles such as saline, dextrose solution, serum albumin and Ringer's solution.
Besides the pharmaceutically acceptable carrier also minor amounts of additives, such as stabilisers, excipients, buffers and preservatives can be included.
The modified IL-8 comprising composition is used to prepare a medicament to prevent or treat any lung inflammatory diseases which are characterized by neutrophil infiltration. More specifically these diseases can be for example chronic obstructive pulmonary disease, cystic fibrosis, severe asthma, bronchitis, broncheolitis, acute lung injury and acute respiratory distress syndrome.
Specifically, the modified IL-8 is used for therapy of COPD.
As an alternative embodiment, prevention or treatment of neutrophilic asthma or exacerbations is specifically covered using modified IL-8 according to the description.
According to the present invention also any lung inflammation can be treated or prevented which is induced by LPS inhalation since LPS is one of the major factors inducing IL-8 expression (Chemokines and chemokine receptors in infectious diseases. (Mahalingam S, Karupiah G., Immunol Cell Biol. 1999 Dec;77(6):469-75). LPS is a component of the walls in Gram-negative bacteria and is therefore present when gram negative bacterial infections occur or is present in air pollutant and in the tobacco leaves (so in cigarette smoke as well).
Alternatively, a method for treatment of lung inflammation with neutrophilic infiltration in a subject in need thereof comprising administering to the subject a therapeutically effective amount of modified IL-8 is covered by the invention. Specifically, the administration is by inhalation or by intratracheal administration.
Chronic obstructive pulmonary disease (COPD).
In particular, neutrophils have been shown to be the most abundant
inflammatory cell in lungs of COPD patients, both in sputum and bronchoalveolar lavage (BAL) samples (Nocker et al. 1996; Peleman et al. 1999). Cigarette smoke is considered to be responsible for elevation of circulating neutrophils, probably due to increased mobilization from the bone marrow (Cowburn et al. 2008), and their sequestration to the lung capillaries were they exit the pulmonary circulation. This feature is peculiar for the pulmonary circulation, since in the systemic circulation neutrophils exit at the level of postcapillary venules.
Neutrophils are then mobilized to the bronchial walls and lung parenchyma (Peleman at al. 1999; Kim et al. 2008). Activation of neutrophils with the subsequent release of reactive oxygen species and elastase is considered the leading cause for the development of lung damage and chronic dysfunction. Indeed, blood neutrophilia has been since long correlated with the rate of decline in lung functions measured in term of forced expiratory volume (FEV; Sparrow et al. 1984).
CXCL8 levels are significantly elevated in sputum and BAL of COPD patients at different stage of disease progression (between 10-15 fold increase vs. healthy) and correlate with disease severity and neutrophil presence (Yamamoto et al. 1997; Tanino et al. 2002), identifying CXCL8 as the key chemokine involved in neutrophil
mobilization (Woolhouse et al. 2002). Further, elevated levels of CXCL8 are also present in sputum of COPD patients during exacerbations (Aaron et al. 2001 ; Spruit et al. 2003).
Cystic fibrosis
Cystic fibrosis (CF) is a severe monogenic disorder of ion transport in exocrine glands, with different mutations in the CF transmembrane conductance regulator (CFTR) gene leading to impaired epithelial chloride secretion (Riordan 1989; Ratjen 2009). Dehydration and plugging of mucous secretions in the ducts of exocrine glands predispose to multi-organ clinical manifestations, particularly in the gastrointestinal, hepatobiliary, reproductive and respiratory tracts.
Chronic bacterial infections and inflammation of the lung are the main causes of morbidity and mortality in CF patients (Ratjen 2006). With increasing age, CF patients develop airway obstruction and many of these patients also suffer from airway hyper- responsiveness and asthma-like symptoms.
Many inflammatory cytokines are produced in the airways in CF patients (Sagel et al. 2002). Several studies have documented increased levels of CXCL-8 in BAL and sputum and increased expression of CXCL8 in bronchial glands of patients with CF (Nakamura et al. 1992; Tabary et al. 1998). Its potent neutrophil chemoattractant properties stimulate the influx of massive numbers of neutrophils in the airways (Chmiel et al. 2002). Moreover neutrophils from CF children display a higher migratory responsiveness to CXCL8 in vitro compared to those of non CF, suggesting that persistent elevated CXCL8 levels can "prime" CF neutrophils (Brennan et al. 2001 ).
In a recent study on in vitro co-culture of CF neutrophils and bronchial epithelial cells bearing the CFTR mutation, it is suggested that in CF patients a high number of non-apoptotic neutrophils adherent on airway epithelium and associated with elevated CXCL8 levels may contribute to sustained and exaggerated inflammatory response in the airways (Tabary et al. 2006).
Indeed, the Na-CI imbalance seems to be the first cause of CXCL8 increased production and subsequent neutrophil infiltration. Bacterial infection are further in- creasing CXCL8 levels, driving more neutrophils infiltration into the lungs and creating a vicious circle difficult to interrupt and resulting in chronic lung inflammation. Acting on this vicious circle can result the most effective treatment for CF patients, which currently rely only on supportive therapy or antibiotics. Severe Asthma
Eosinophil inflammation has for long been considered the most distinctive pathological hallmark of asthma (Bousquet 1990). However, eosinophil inflammation is present in the airways of only 50% of asthmatic patients (Douwes et al. 2002), and often not observed in asthma exacerbations.
A strong association has now been established between neutrophilic inflammation and severe asthma (Little et al. 2002; Wenzel et al. 1997, Jatakanon et al 1999, Ordonez at al. 2000, Kamath et al. 2005; Fahy 2009), childhood asthma
(McDouglas et al. 2006), asthma exacerbations (Fahy et al. 1995), corticosteroid resistant asthma (Green et al 2002), nocturnal asthma (Martin et al 1991 ), asthma in smokers (Chalmers et al. 2001 ) and occupational asthma (Anees et al.2002).
During an acute asthma attack, eosinophils and neutrophils are coexisting (Wenzel et al 1999), but it is suggested that neutrophils are becoming the predominant cell population over time. Their presence in the airways is in proportion to disease severity and progression (Wenzel et al 1997) and is associated to airflow obstruction and reduced lung function (Shaw et al. 2007). Neutrophils are also the main leukocyte population observed in a very severe and often lethal form of asthma characterized by sudden-onset (sudden-onset asthma; Sur et al. 1993). Acute lung injury (ALI) and acute respiratory distress syndrome (ARSD)
Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS), represent two different stages of the same disease, characterized by acute lung inflammation with enhanced vascular permeability and lung oedema formation of increasing severity (Bernard et al. 1994). The pathology results in a very high mortality rate (about 40%) with no trend in decreasing in the last decade, despite improved intensive care intervention (Puha et al. 2009).
The involvement of neutrophils in ALI and ARDS has been documented by several groups (Ware et al 2000; Abraham 2003) and increased level of neutrophils in BAL and its correlation with increased CXCL8 levels has been reported (Aggarwal et al. 2000). Moreover CXCL8 elevated levels in BAL have been correlated with ARDS development on at-risk patients (Donnelly et al. 1993; Reid et al. 1995).
In the lung, glycosaminoglycans (GAGs) are the main component of the non- fibrillar compartment of the interstitium, and are located in the sub-epithelial tissue and in the bronchial walls, as well as in the airways secretions. Their presence is essential to regulate hydration and water homeostasis, maintain tissue structure, and modulate inflammatory responses (e.g. rev. Souza-Fernandes et al. 2006).
Compared to other human pathologies not a lot of literature is available supporting the involvement of GAGs in mediating chemokine actions in pulmonary diseases.
All four classes of glycosaminoglycans, including heparin/heparan sulphate, chondroitin/dermatan sulfate, keratin sulfate, and hyaluronan are present in normal lungs. Heparan sulfate has been reported as the predominant form (-40%), followed by chondroitin/dermatan sulphate (-31 %) and to minor extent to hyaluronan (-14%) and keratin sulfate (Frevert et al. 2003).
The foregoing description will be more fully understood with reference to the following examples. Such examples are, however, merely representative of methods of practicing one or more embodiments of the present invention and should not be read as limiting the scope of invention.
EXAMPLES
Considering that in most, if not all lung pathologies an alteration of the lung vascular permeability and of the matrix is observed, systemic administration of PA401 (del6F17KF21 KE70KN71 K IL-8 mutant) should be appropriate to reach adequate levels of PA401 at both lung venule and epithelial level.
Example 1 :
PA401 effects on LPS-induced acute lung inflammation models in mice.
A variety of stimuli induce neutrophil migration into the lung. Among the most frequently used and best characterized inflammatory inducer is the endotoxin of Gram- negative bacteria (lipopolysaccharide: LPS).
LPS instilled intranasal, or aerosolized induces a dose and time dependent neutrophil infiltration in the lung vasculature, interstitium and BAL (Reutershan et al. 2005), with peak levels reached between 4-8 hours post challenge, and remaining significantly above baseline for up to 24 hours in mice.
The LPS dose administered varies based on the LPS serotype, the method of application and the strain of the mice used, with significant BAL neutrophilia reported for doses of LPS as low as 0.1 pg/mouse and up to 800Mg/mouse.
LPS inhalation is able to induce lung neutrophil infiltration across species (e.g. mice and rats, Chapman et al.2007; guinea pigs; Wu et al. 2002; rabbits, Smith et al. 2008; sheep, Waerhaug et al. 2009; horses, van den Hoven et al. 2006; dogs, Koshika et al. 2001 ). Inhalation of 1 to 100 g of LPS in healthy volunteers is regarded as robust and reliable model for acute lung inflammation (Maris et al. 2005, Kitz et al. 2008) as well as chronic obstructive lung disease exacerbation (Kharitonov et al, 2007).
To assess potential of PA401 as anti-inflammatory in models of acute lung neutrophilia, a dose-response study with subcutaneous administration of PA401 at doses of 4, 40, 200 and 400 g/Kg in C57BL/6J female mice intranasal instilled with LPS (0.3Mg/mouse, serotype pseudomonas aeruginosa) was performed. Sham instilled (saline) and Dexamethasone 3mg/kg s.c. (administered at t=-1 h before LPS instillation) treated animals were used as controls. Bronchoalveolar lavage (BAL) was performed 4h post LPS instillation and total cell count on BAL samples, as well as differential cell count on BAL cytospin were measured.
PA401 induced a dose-dependent reduction in the total number of cells infiltrated in lung as assessed in the BAL fluid (Fig.1 ). The effect was due to a significant reduction in the number of neutrophils (Fig. 2) and lymphocytes (Fig.3), with significant effect for dose of PA401 as low as 4 g/kg. Surprisingly, the inhibition of cell infiltration in the BAL obtained with PA401 at the doses of 200 and 400 g/kg was comparable to that obtained with the treatment with a high dose of Dexamethasone (3mg/kg). This study was conducted at Argenta Discovery Ltd, UK.
Example 2
A second study was performed using a slightly different model, which imply a different mouse strain and gender (male Balb/c instead of C57BL/6) a different LPS strain and serotype (Salmonella enterica instead of E. Coli) and a different LPS administration (aerosol - 3.5mg/7mL over 30min - instead of intranasal). PA401 doses of 4, 40 and 400 g/kg were administered either by s.c. or i.v. route at t=-5 and t=+3h from LPS exposure. BAL total and differential cell count were evaluated at t=8h, a later time point compared to the previous study.
Saline aerosolized mice and mice receiving an intra tracheal administration of Dexamethasone (20 g/20 l/mouse at t=-1 h) were used as control.
Also in this case PA401 induced a highly significant reduction in the number of total cells in the BAL (Fig. 4), due to reduction in neutrophils count (Fig.5). The activity of PA401 was more significant when administered by intravenous than by
subcutaneous route, reaching the same inhibitory effects of intra tracheal
administration of dexamethasone. The study was performed at Pneumolabs Ltd, UK. These studies demonstrate strong activity of PA401 administered by
subcutaneous or intravenous route in 2 acute lung neutrophilic inflammatory animal models resembling human ALI/ARDS and COPD exacerbations. The effect was obtained independently of gender or genetic background of the animals; the LPS serotype and after both intranasal instillation and aerosol exposure.
Example 3
PA401 effects in an acute model of cigarette smoke induced lung inflammation. Acute exposure of mice to cigarette smoke leads to lung responses that, at least in part, mimic the lung inflammation observed in COPD patients. Different mouse strains present variable degree of lung inflammation following acute cigarette smoke exposure (Guerrassimov et al. 2004, Vlahos et al. 2006). This genetic variability in the response in mice appears quite representative of the variable susceptibility to develop COPD among human smokers, and therefore this model is considered the most relevant to model the human pathology. Lung inflammation was induced in C57BL/6J female mice (a susceptible strain) by exposure to cigarette smoke of 4 to 6 cigarette over a four-day period. Dose response activity of subcutaneous PA401 treatment at the doses of 4, 40 and
400 g/kg administered at t=+30min and t=+6h from smoke exposure, on cell infiltrates on bronchoalveolar lavages was evaluated 24h after last cigarette smoke exposure. Air exposed, and Roflumilast (5mg/kg oral) treated animals serve as controls.
Also in this animal model of COPD, PA401 dose dependently inhibited the cigarette smoke induced increased in total cell number recovered in the BAL.
Highly significant effect on total cell infiltrates was observed at 40 and 400 g/kg. (Fig.6). At the highest dose used, PA401 significantly reduced neutrophil (Fig.7), macrophage (Fig.8), epithelial cells (Fig.9) as well as lymphocyte (Fig.10) numbers in BAL. Significant effect on most of these cell subtypes were observed also for the other two doses used in the study. The inhibitory effect of PA401 400 g/kg was comparable of that obtained with Roflumilast 5mg/kg. This study was performed at Argenta
Discovery Ltd, UK.
PA401
This study demonstrates activity of PA401 on mixed cell infiltration induced by 4-day repeated exposure to cigarette smoke, and animal model predictive of antiinflammatory activities in COPD patients.
Example 4:
PA401 effects in a sub-chronic model of cigarette smoke induced lung
inflammation.
In this study lung inflammation was induced in C57BL/6J female mice by exposure to cigarette smoke of 4 to 6 cigarettes over an eleven-day period. Dose frequency activity of subcutaneous PA401 treatment at the optimal dose of 400 g/kg, based on the study in example 3, administered at t=+30min and t=+6h from smoke exposure (twice a day: b.i.d.), once daily at +3h (q.d) and every other day (q.o.d) at +3h, on cell infiltrates on bronchoalveolar lavages was evaluated 24h after last cigarette smoke exposure. Air exposed, and Roflumilast (5mg/kg oral) treated animals serve as controls.
Significant effect on total cell infiltrates was observed after twice a day, daily and every other day administration (Fig.1 1 ). At all the treatment frequency used, PA401 significantly reduced neutrophil (Fig.12) and epithelial cells (Fig.13) numbers in BAL, while the significant reduction in lymphocyte (Fig 14) and macrophage (Fig.15) obtained with b.i.d. and q.d. administration was lost when PA401 was administered every other day. The inhibitory effect of PA401 administered twice and once a day were comparable of that obtained with Roflumilast 5mg/kg. This study was performed at Argenta Discovery Ltd, UK.
PA401
This study demonstrates that PA401 administration at the dose of 400 g/kg subcutaneously twice a day and once a day have comparable activity on mixed cell infiltration induced by 1 1 -day repeated exposure to cigarette smoke.
Example 5:
PA401 effects in a sub-chronic model of cigarette smoke induced lung inflammation.
In this study lung inflammation was induced in C57BL/6J female mice by exposure to cigarette smoke of 4 to 6 cigarettes over an eleven-day period. PA401 at the dose of 400Mg/kg daily subcutaneous was compared to the dose of 40pg/kg daily subcutaneous. Treatment was performed at t+3h and effects on cell infiltrates on bronchoalveolar lavages were evaluated 24h after last cigarette smoke exposure. Air exposed, Roflumilast (5mg/kg oral) treated animals as well as animals treated with the CXCR2 antagonist SCH527123 (10 mg/kg twice a day oral- total daily dose 20mg/kg) serve as controls.
Significant effect of PA401 on total cell infiltrates was observed after 400 and 40 g/kg administration (Fig.16). This was due to a significant reduction in neutrophil (Fig.17), epithelial cells (Fig.18), lymphocyte (Fig 19) and macrophage (Fig.20) numbers in BAL. The inhibitory effect of PA401 administrered at the dose of 400 g/kg are comparable of that obtained with Roflumilast 5mg/kg and the CXCR2 antagonist SCH527123 20mg (10mg/kg twice a day). This study was performed at Argenta Discovery Ltd, UK.
PA401
This study demonstrates activity of PA401 administered once a day
subcutaneously at the doses of 400 g/kg and 40 g/kg on mixed cell infiltration induced by 1 1 -day repeated exposure to cigarette smoke. Example 6:
PA401 effects in lung inflammation after local delivery to the lung.
An experiment was performed to verify activity of PA401 after local delivery to the lung in an animal model of LPS induced lung inflammation.
In this experiment lung inflammation was induced by the delivery of LPS by aerosol (Salmonella enterica, 3.5mg/7mL over 30min) in male Balb/c mice. PA401 was administered intra-tracheally (i.t.) using a MicroSprayer (FJM-250 syringe;
PennCentury), a device that allows delivery of a plume of aerosol (mass median diameter 16-22μηη) directly into the lungs. PA401 i.t. administration was performed at the doses of 100, 40, 10 and 4 g/kg either 1 h before LPS exposure, or 1 h after the end of LPS exposure. Bronchoalveolar lavage (BAL) was performed 8h post LPS exposure and total cell and neutrophil numbers were measured. Dexamethasone 20 g/20 l mouse i.t. was used as reference compound.
PA401 induced a significant dose dependent reduction of total cells in BAL (Fig. 21 ), mainly due to neutrophil number reduction (Fig. 22). The therapeutic treatment (t=+1 h) resulted in about 10% improved activity, compared to the prophylactic treatment (-1 h) with doses of 100, 40 and 10 g/kg being significantly active.
Fig.21 shows the dose-response activity on PA401 intra-tracheal administration 1 hour before (-1 h) and 1 hour after (+1 h) LPS aerosol exposure on total cell count in the bronchoalveolar lavage collected 8 hours post LPS. ANOVA followed by Dunnett 's test: *p<0.05; **p<0.01 versus vehicle treated animals.
Fig.22 shows the dose-response activity on PA401 intra-tracheal administration 1 hour before (-1 h) and 1 hour after (+1 h) LPS aerosol exposure on neutrophils count in the bronchoalveolar lavage collected 8 hours post LPS. ANOVA followed by
Dunnett's test: *p<0.05; **p<0.01 versus vehicle treated animals.
These data demonstrate activity of PA401 following local delivery to the lung and open the possibility to the use of this administration route, which has normally good patient compliance, as alternative to intravenous or subcutaneous administration for chronic lung indications References
Aaron SD, Angel JB, Lunau M, et al. Granulocyte inflammatory markers and airway infection during acute exacerbation of chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2001 ; 163: 349-55.
Abraham E. Neutrophils and acute lung injury, Crit Cre Med 2003; Suppl: S195-
S199.
Aggarwal A, baker CS, Evans TW et al. G-Csf and II-8 but not GM-CSF correlate with disease severity of pulmonary neutrophilia in acute respiratory distress syndrome. Eur Resp J 2000; 15: 895-901 .
Anees W, Huggins V, Pavord ID, et al. Occupational asthma due to low molecular weight agents: eosinophilic and non-eosinophilic variants. Thorax 2002; 57:231- 6.
Bernard GR, Artigas A, Brigham KL et al. The American-European Consensus Conference on ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination. Am j Respir Crit Care Med 1994; 149: 818-824.
Brennan S, Cooper D, Sly PD. Directed neutrophil migration to IL-8 is increased in cystic fibrosis: a study of the effect of erythromycin. Thorax 2001 ; 56: 62-64.
Bousquet J, Chanez P, Lacoste JY, et al. Eosinophilic inflammation in asthma. N Engl J Med 1990; 323: 1033-9.
Chalmers GW, Macleod KJ, Thomson L, et al. Smoking and airway
inflammation in patients with mild asthma. Chest 2001 ; 120: 1917-22.
Chapman RW, Minnicozzi M, Celly CS, et al. A novel, orally active CXCR1/2 receptor antagonist, Sch527123, inhibits neutrophil recruitment, mucus production, and goblet cell hyperplasia in animal models of pulmonary inflammation. J Pharmacol Exp Ther 2007; 22(2): 486-93.
Chmiel JF, Berger M, Konstan MW. The role of inflammation in the pathophysiology of CF lung disease. Clin Rev Allergy Immunol 2002; 23(1 ): 5-27.
Cowburn SC, Condliffe AM, Farhai N et al. Advances in neutrophil biology. Clinical Implications. Chest 2008; 134: 606-612.
Culpitt SV, Maziak W, Luokidis S et al. Effects of high dose inhaled steroids on cells, cytokines, and proteases in induced sputum in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 1999; 160: 1635-1639. De Boer Wl, Yao H, Rahman I. Future therapeutic treatment of COPD: struggle between antioxidants and cytokines. I J COPD 2007; 2(3): 205-228.
Donnelly SC, Strieter RM, Kunkel SL et al. lnterleukine-8 and development of adult distress syndrome in at-risk patients groups. Lancet 1993; 341 : 643-647.
Douwes J, Gibson P, Pekkanen J, et al. Non-eosinophilic asthma: importance and possible mechanisms. Thorax 2002; 57: 643-648
Fabbri L, Pauwels RA, Hurds SS. Global strategy for the diagnosis,
management and prevention of chronic obstructive pulmonary disease: GOLD executive summary updated 2003. J COPD 2004; 1 : 105-141 .
Fahy JV, Kim KW, Liu J, et al. Prominent neutrophilic inflammation in sputum from subjects with asthma exacerbation. J Allergy Clin Immunol 1995; 95: 843-52.
Fahy JV. Eosinophilic and neutrophilic inflammation in asthma. Insight from Clinical studies. Proc Am Thorac Soc 2009; 6: 256-259.
Fitzgerald MF, Fox JC. Emerging trends in the therapy of COPD: novel anti- inflammatory agents in clinical development. Drug Discov Today 2007; 12(1 1 12): 479- 485.
Frevert CW, Kinsella MG, Vathanaprida C et al. Binding of interleikin-8 to heparin and chondroitin sulfate in lung tissue. Am J Respir Cell Mol Biol 2003; 28: 464- 472.
Green RH, Brightling CE, Woltmann G, et al. Analysis of induced sputum in adults with asthma: identification of subgroup with isolated sputum neutrophilia and poor response to inhaled corticosteroids. Thorax 2002; 57: 875-9.
Guerassimov A, Hoshino Y, Takubo Y et al. The development of emphysema in cigarette smoke-exposed mice is strain dependent. Am J Respir Crit Care Med 2004; 170. 974-980.
Kamath AV, Pavord ID, Ruparelia PR et al. Is the neutrophil the key effector cell in severe asthma? Thorax 2005; 60: 529-530.
Kharitonov SA, Sjobring U. Lypopolysaccharide challenge in humans as a model for chronic obstructive lung disease exacerbations. Contrib Microbiol 2007; 14: 83-100.
Kim V, Rogers TJ, Criner Gj. New concepts in the pathology of Chronic
Obstructive Pulmonary Disese. Proc Am Thorac Soc 2008; 5: 478-485. Kitz R, Rose MA, Placzek K et al. LPS inhalation challenge: a new tool to characterize the inflammatory response in humans. Med Microbiol Immunol 2008; 197: 13-19.
Koshika T, Ishizaka A, Nagatomi I et al. Pretreatment with FK506 improves survival rate and gas exchange in canine model of acute lung injury. Am J Respir Crit Care Med. 2001 ; 163 (1 ): 79-84.
Jatakanon A, Uasuf C, Maziak W, et al. Neutrophilic inflammation in severe persistent asthma. Am J Respir Crit Care Med 1999; 160: 1532-9.
Lamblin C, Gosset P, Tillie-Leblond I et al. Bronchial neutrophilia in patients with non infectious status asthmaticus. Am J Respir Crit Care Med 1998; 157: 394-402.
Lefant C, Khaltaev N. Workshop report: Global strategy for the diagnosis, management and prevention of COPD. URL: http://aoldcopd.org/Guidelineitem.asp Little SA, Macleod KJ, Chalmers GW, et al. Association of forced expiratory volume with disease duration and sputum neutrophils in chronic asthma. Am J Med 2002; 1 12: 446-452.
Lopez AD, Murray CC. The global burden of disease, 1990-220. Nat Med 1998; 4: 1241 -1243.
McDougall CM, Helmes PJ. Neutrophil airway inflammation in childhood asthma. Thorax 2006; 61 (9): 739-741 .
Mahadeva R, Shapiro SD. Chronic obstructive pulmonary disease-3:
experimental animal models of pulmonary emphysema. Thorax 2002; 57: 908-914.
Maris NA, de VOS AF, Dessing MC et al. Antiinflammatory effects of salmeterol after inhalation of lipopolysaccharide by healthy volunteers. Am J Respir Crit Care Med 2005; 172(7): 878-884.
Martin RJ, Cicutto LC, Smith HR, et al. Airways inflammation in nocturnal asthma. Am Rev Respir Dis 1991 ; 143: 351-357.
Nakamura H, Yoshimura K, McElvaney NG, et al. Neutrophil elastase in respiratory epithelial lining fluid of individuals with cystic fibrosis induces interleukin-8 gene expression in a human bronchial epithelial cell line. J Clin Inves. 1992; 89: 1478- 1484
Nocker RE, Schoonbrood DF, van de Graaf EA, et al. Interleukin-8 in airway inflammation in patients with asthma and chronic obstructive pulmonary disease. Int Arch Allergy Immunol 1996; 109: 183-191 Ordonez CL, Shaughnessy TE, Matthay MA, et al. Increased neutrophil numbers and IL-8 levels in airway secretions in acute severe asthma: clinical and biological significance. Am J Respir Crit Care Med 2000; 161 : 1 185-1 190.
Pauwels RA, Buist As, Calverley PM, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. NHLBI/WHO Global Initiative for Chronic Obstructive Pulmonary Disease (GOLD) Workshop summary. Am J Respir Crit Care Med. 2005; 5: 1256-1276.
Peleman RA, Rytila PH, Kips JC et al. The cellular composition of induced sputum in chronic obstructive pulmonary disease. Eur Respir J 1999; 13: 839-843.
Puha J, Badia JR, Adhikari et al. Has mortality from acute respiratory distress syndrome decreased over time?: A systematic review. Am J Respir Crit Care Med 2009; 179(3): 220-227.
Ratjen FA. Diagnosing and managing infection in CF. Paediatr Respir rev 2006; 7 (suppM ): S151 -S153.
Ratjen FA. Cystic fibrosis: pathogenesis and future treatment strategies. Respir
Care 2009; 54(5): 595-602.
Reid PT, Donnelly Sc, Haslett C. Inflammatory predictors for the development of adult respiratory distress syndrome. Thorax 1995; 50: 10023-10026.
Reutershan J, Basit A, Galkina EV et al. Sequential recruitment of neutrophils into lung and bronchoalveolar lavage fluid in LPS-induced acute lung injury. Am J Physiol Cell Mol Physiol Lung Cell Mol Physiol 2005; 289 (5): L807-L815.
Riordan JR, Rommens JM, Kerem B, et al. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science. 1989; 245 :1066- 1073.
Sagel SD, Accurso FJ. Monitoring inflammation in CF. Cytokines. Clin Rev
Allergy Immunol. 2002; 23:41 -57.
Shaw DE, Berry MA, Hargadon B et al. Association between neutrophilic airway inflammation and airflow limitation in adults with asthma. Chest 2007; 132: 1871 -1875.
Smith LS, Kajikawa O, Elson G et al. effect of Toll-like receptor 4 blockade on pulmonary inflammation caused by mechanical ventilation and bacterial endotoxin. Exp Lung Res 2008; 34: 225-245.
Souza-Fernandes AB, Pelosi P, Rocco PRM. Bench-to-bedside review: the role of glycosaminoglycans in respiratory disease. Crit care 2006; 10(6): 237-252. Sparrow D, Glynn RJ, Cohen M et al. The relationship of the peripheral leukocyte count and cigarette smiking to pulmonary function among adult men. Chest 1984. 86: 383-386.
Spruit MA, Gosselink R, Troosters T, et al. Muscle force during an acute exacerbation in hospitalized patients with COPD and its relationship with CXCL8 and IGF-I. Thorax 2003; 58: 752-756.
Sur S, Crotty TB, Kephart GM, et al. Sudden-onset fatal asthma: a distinct entity with few eosinophils and relatively more neutrophils in the airway submucosa? Am Rev Respir Dis 1993; 148: 713-719.
Tabary O, Zahm JM, Hinnrasky J, et al. Selective up-regulation of chemokine IL-
8 expression in cystic fibrosis bronchial gland cells in vivo and in vitro. Am J Pathol 1998; 153: 921-930.
Tabary O, Corvol H, Boncoeur E et al. Adherence of airway neuthrophils and inflammatory response are increased in Cf airway epithelial cell-neutrophil interactions. Am J Physiol Lung Cell Mol Physiol 2006; 290 (3): L588-596.
Tanino M, Betsuyaku T, Takeyabu K, et al. Increased levels of interleukin-8 in BAL fluid from smokers susceptible to pulmonary emphysema. Thorax 2002; 57: 405- 41 1 .
Van den Hoven R, Duvigneau JC, Hertl RT et al. Clenbuterol affects the expression of messenger RNA for interleukin 10 in peripheral leukocytes from horses challenged intrabronchially with lipopolysaccharides. Vet Res Commun 2006; 30(8): 921 -928.
Vlahos R, Bozinovski S, Jones JE et al. Differential protease, innate immunity, and NK-kB induction profiles during lung inflammation induced by subchronic cigarette smoke exposure in mice. Am J Physiol Lung Cell Mol Physiol 2006; 290: L931 -L945.
Wenzel SE, Schwartz LB, Langmack EL, et al. Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics. Am J Respir Crit Care Med 1999; 160: 1001-8.
Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med 2000; 342: 1334-1349.
Waerhaug K, Kuzkov W, Kulin VN et al. Inhaled aerosolized recombinant human protein C ameliorates endotoxin-induced lung injury in anesthetized sheep. Crit Care 2009; 13(2) R51 . Wenzel SE, Szefler SJ, Leung DYM, et al.Bronchoscopic evaluation of severe asthma: persistent inflammation associated with high dose glucocorticoids. Am J Respir Crit Care Med 1997; 156: 737-473.
Woolhouse IS, Bayley DL, Stockley RA. Sputum chemotactic activity in chronic obstructive pulmonary disease. Effects of a1 -antitrypsin deficiency and the role of leukotriene B4 and interleukine 8. Thorax 2002; 57: 709-714.
Wu Y, Singer M, Thouron F et al. Effect of surfactant on pulmonary expression of type IIA PLA(2) in an animal model of acute lung injury. Am J Physiol Lung Cell Mol Physiol 2002; 282(4): L743-750.
Yamamoto C, Yoneda T, YoshikawaM, et al. Airway inflammation in COPD assessed by sputum levels of interleukin-8. Chest 1997; 1 12: 505-510.
The ENFUMOSA study group. The ENFUMOSA cross-sectional European multicentre study of the clinical phenotype of severe asthma. Eur Respir J 2003; 22- 470-477.

Claims

Claims
1 . Modified interleukin 8 (IL-8) having increased glyosaminoglycan (GAG) binding affinity and further inhibited or down-regulated G-protein coupled receptor (GPCR) activity compared to the respective wild type IL-8 for use in the prevention or treatment of lung inflammation with neutrophilic infiltration.
2. Modified IL-8 according to claim 1 , wherein the modified IL-8 comprises a GAG binding region which is modified by substitution, insertion, and/or deletion of at least one amino acid in order to increase the relative amount of basic amino acids in said GAG binding region, and/or reduce the amount of bulky and/or acidic amino acids in said GAG binding region preferably at a solvent exposed position.
3. Modified IL-8 according to claim 2, wherein at least one amino acid selected from the group consisting of Arg, Lys, and His is inserted into said GAG binding region.
4. Modified IL-8 according to any one of claims 1 to 3, wherein positions 17, 21 , 70, and/or 71 of IL-8 are substituted by Arg, Lys, His, Asn and/or Gin.
5. Modified IL-8 according to any one of claims 1 to 4, wherein the GPCR binding region of IL-8 is modified by deletion, insertion, and/or substitution, preferably with alanine, a sterically and/or electrostatically similar residue.
6. Modified IL-8 according to any one of claims 1 to 5 wherein the amino acid sequence of the modified IL-8 molecule is
(X1 )n(X2)m KTYSKP(X3)HPK (X4JKELRVIES GPHCANTEII VKLSDGRELC
LDPKENWVQR WEKFLKRA(X5) (X6)S (SEQ ID No. 2)
wherein X1 is of amino acid sequence SAKELR,
wherein X2 is of amino acid sequence CQCI,
wherein X3 is selected of the group consisting of F, R, K, H, N and/or Q, preferably X3is K,
wherein X4 is selected of the group consisting of F, R, K, H, N and/or Q, preferably X4 is K wherein X5 is selected of the group consisting of E, R, K, H, N and/or Q, preferably X5 is K,
wherein X6 is selected of the group consisting of R, K, H, N and/or Q, preferably X6 is K,
and wherein n and/or m is 0 or 1 .
7. Modified IL-8 according to any one of claims 1 to 6, wherein said modified IL- 8 molecule is selected from the group consisting of del6F17RE70KN71 R,
del6F17RE70RN71 K, del6E70KN71 K, and del6F17KF21 KE70KN71 K.
8. Modified IL-8 according to any one of claims 1 to 7, wherein the amino acid sequence of the modified IL-8 molecule is CQCI KTY SKPKHPKKIK ELRVIESGPH CANTEIIVKL SDGRELCLDP KENWVQRVVE KFLKRAKKS (SEQ ID No. 1 ).
9. Modified IL-8 according to any one of claims 1 to 8, wherein the lung inflammation with neutrophilic infiltration is selected from chronic obstructive
pulmonary disease, cystic fibrosis, severe asthma, bronchitis, broncheolitis, acute lung injury and acute respiratory distress syndrome.
10. Modified IL-8 according to any one of claims 1 to 9 formulated as inhalant.
1 1 . Method for treatment of lung inflammation with neutrophilic infiltration in a subject in need thereof comprising administering to the subject a therapeutically effective amount of modified IL-8.
12. Method according to claim 1 1 wherein the administration is by inhalation or by intratracheal administration.
PCT/EP2010/063389 2009-09-11 2010-09-13 Composition for treatment of cxcl8-mediated lung inflammation WO2011029931A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
JP2012528380A JP2013504545A (en) 2009-09-11 2010-09-13 Composition for the treatment of CXCL8-mediated pulmonary inflammation
AU2010294225A AU2010294225A1 (en) 2009-09-11 2010-09-13 Composition for treatment of CXCL8-mediated lung inflammation
CN2010800508273A CN102596227A (en) 2009-09-11 2010-09-13 Composition for treatment of CXCL8-mediated lung inflammation
CA2773664A CA2773664A1 (en) 2009-09-11 2010-09-13 Composition for treatment of cxcl8-mediated lung inflammation
US13/395,128 US20120288474A1 (en) 2009-11-06 2010-09-13 Composition for treatment of cxcl8-mediated lung inflammation
EA201200470A EA201200470A1 (en) 2009-09-11 2010-09-13 COMPOSITION FOR THE TREATMENT OF THE MEDIATED CXCL8 LUNG INFLAMMATION
EP10751694A EP2475380A1 (en) 2009-09-11 2010-09-13 Composition for treatment of cxcl8-mediated lung inflammation
ZA2012/01802A ZA201201802B (en) 2009-09-11 2012-03-12 Composition for treatment of cxcl8-mediated lung inflammation

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US24177109P 2009-09-11 2009-09-11
US61/241,771 2009-09-11
EP09175203 2009-11-06
EP09175203.0 2009-11-06
EP10155086 2010-03-01
EP10155086.1 2010-03-01

Publications (1)

Publication Number Publication Date
WO2011029931A1 true WO2011029931A1 (en) 2011-03-17

Family

ID=42830226

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2010/063389 WO2011029931A1 (en) 2009-09-11 2010-09-13 Composition for treatment of cxcl8-mediated lung inflammation

Country Status (10)

Country Link
US (1) US20120288474A1 (en)
EP (1) EP2475380A1 (en)
JP (1) JP2013504545A (en)
KR (1) KR20120080196A (en)
CN (1) CN102596227A (en)
AU (1) AU2010294225A1 (en)
CA (1) CA2773664A1 (en)
EA (1) EA201200470A1 (en)
WO (1) WO2011029931A1 (en)
ZA (1) ZA201201802B (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018131724A1 (en) * 2017-01-13 2018-07-19 Tobishi Pharmaceutical Co., Ltd. Neutrophil activation regulator
WO2019243272A1 (en) * 2018-06-18 2019-12-26 College De France Ligand controling interaction between gags with their effector molecules and use thereof
KR20230141227A (en) 2022-03-31 2023-10-10 (주)케어젠 Peptide Having Activity of Anti-Inflammation and Anti-Fibrosis and Uses Thereof
US11840565B2 (en) 2016-12-29 2023-12-12 University Of Miami Methods and compositions for treating virus-associated inflammation

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103399151A (en) * 2013-06-24 2013-11-20 上海交通大学医学院附属瑞金医院 Application of CXCL8 cytokine detection reagent in preparation of diagnostic reagent for papillary thyroid carcinoma
CN106279399B (en) * 2015-06-03 2021-01-12 北京锐瑟生物医药科技发展有限公司 Chemokine modified peptide
WO2016192067A1 (en) * 2015-06-03 2016-12-08 翔升科技股份有限公司 Modified chemokine peptide
WO2018067938A1 (en) * 2016-10-06 2018-04-12 The Trustees Of Columbia University In The City Of New York Cell-seeded porous lung hydrogel sealant
US20190336598A1 (en) * 2016-12-29 2019-11-07 University Of Miami Methods for modulating inflammasome activity and inflammation in the lung
SG10201902000YA (en) * 2019-03-06 2020-10-29 Nat Univ Singapore Isthmin 1 for treatment of lung inflammation
KR20220128382A (en) * 2020-01-17 2022-09-20 새미-사빈사 그룹 리미티드 Composition for managing chronic obstructive pulmonary disease

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5399341A (en) * 1990-06-21 1995-03-21 Huland; Edith Use of cytokin-containing aerosols and the cytokin-containing aerosols
US5665346A (en) * 1991-12-04 1997-09-09 Research Corporation Technologies, Inc. Human interleukin-8 analogs
WO2005054285A1 (en) 2003-12-04 2005-06-16 Protaffin Biotechnologie Ag Gag binding proteins
EP1894571A1 (en) * 2006-08-29 2008-03-05 Protaffin Biotechnologie AG Use of modified interleukin-8 proteins for treating reperfusion injury or transplant rejection
WO2008074047A1 (en) * 2006-12-20 2008-06-26 Protaffin Biotechnologie Ag Chemokine-chaperone fusion proteins
WO2009015884A1 (en) * 2007-07-31 2009-02-05 Protaffin Biotechnologie Ag Glycosaminoglycan-antagonising mcp-1 mutants and methods of using same
EP2053060A1 (en) * 2007-10-24 2009-04-29 Protaffin Biotechnologie AG SDF-1-based glyocosaminoglycan antagonists and methods of using same

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5399341A (en) * 1990-06-21 1995-03-21 Huland; Edith Use of cytokin-containing aerosols and the cytokin-containing aerosols
US5665346A (en) * 1991-12-04 1997-09-09 Research Corporation Technologies, Inc. Human interleukin-8 analogs
WO2005054285A1 (en) 2003-12-04 2005-06-16 Protaffin Biotechnologie Ag Gag binding proteins
EP1894571A1 (en) * 2006-08-29 2008-03-05 Protaffin Biotechnologie AG Use of modified interleukin-8 proteins for treating reperfusion injury or transplant rejection
WO2008074047A1 (en) * 2006-12-20 2008-06-26 Protaffin Biotechnologie Ag Chemokine-chaperone fusion proteins
WO2009015884A1 (en) * 2007-07-31 2009-02-05 Protaffin Biotechnologie Ag Glycosaminoglycan-antagonising mcp-1 mutants and methods of using same
EP2053060A1 (en) * 2007-10-24 2009-04-29 Protaffin Biotechnologie AG SDF-1-based glyocosaminoglycan antagonists and methods of using same

Non-Patent Citations (67)

* Cited by examiner, † Cited by third party
Title
"The ENFUMOSA study group. The ENFUMOSA cross-sectional European multicentre study of the clinical phenotype of severe asthma", EUR RESPIR J, vol. 22, 2003, pages 470 - 477
AARON SD; ANGEL JB; LUNAU M ET AL.: "Granulocyte inflammatory markers and airway infection during acute exacerbation of chronic obstructive pulmonary disease", AM J RESPIR CRIT CARE MED, vol. 163, 2001, pages 349 - 55
ABRAHAM E.: "Neutrophils and acute lung injury", CRIT CRE MED, 2003, pages S195 - S199
AGGARWAL A; BAKER CS; EVANS TW ET AL.: "G-Csf and 11-8 but not GM-CSF correlate with disease severity of pulmonary neutrophilia in acute respiratory distress syndrome", EUR RESP J, vol. 15, 2000, pages 895 - 901
ANEES W; HUGGINS V; PAVORD ID ET AL.: "Occupational asthma due to low molecular weight agents: eosinophilic and non-eosinophilic variants", THORAX, vol. 57, 2002, pages 231 - 6
BERNARD GR; ARTIGAS A; BRIGHAM KL ET AL.: "The American-European Consensus Conference on ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination", AM J RESPIR CRIT CARE MED, vol. 149, 1994, pages 818 - 824
BOUSQUET J; CHANEZ P; LACOSTE JY ET AL.: "Eosinophilic inflammation in asthma", N ENGL J MED, vol. 323, 1990, pages 1033 - 9
BRENNAN S; COOPER D; SLY PD: "Directed neutrophil migration to IL-8 is increased in cystic fibrosis: a study of the effect of erythromycin", THORAX, vol. 56, 2001, pages 62 - 64
CHALMERS GW; MACLEOD KJ; THOMSON L ET AL.: "Smoking and airway inflammation in patients with mild asthma", CHEST, vol. 120, 2001, pages 1917 - 22
CHAPMAN RW; MINNICOZZI M; CELLY CS ET AL.: "A novel, orally active CXCR1/2 receptor antagonist, Sch527123, inhibits neutrophil recruitment, mucus production, and goblet cell hyperplasia in animal models of pulmonary inflammation", J PHARMACOL EXP THER, vol. 22, no. 2, 2007, pages 486 - 93
CHMIEL JF; BERGER M; KONSTAN MW: "The role of inflammation in the patho- physiology of CF lung disease", CLIN REV ALLERGY IMMUNOL, vol. 23, no. 1, 2002, pages 5 - 27
COWBURN SC; CONDLIFFE AM; FARHAI N ET AL.: "Advances in neutrophil biology", CLINICAL IMPLICATIONS. CHEST, vol. 134, 2008, pages 606 - 612
CULPITT SV; MAZIAK W; LUOKIDIS S ET AL.: "Effects of high dose inhaled steroids on cells, cytokines, and proteases in induced sputum in chronic obstructive pulmonary disease", AM J RESPIR CRIT CARE MED, vol. 160, 1999, pages 1635 - 1639
DE BOER WI; YAO H; RAHMAN I: "Future therapeutic treatment of COPD: struggle between antioxidants and cytokines", I J COPD, vol. 2, no. 3, 2007, pages 205 - 228
DONNELLY SC; STRIETER RM; KUNKEL SL ET AL.: "Interleukine-8 and development of adult distress syndrome in at-risk patients groups", LANCET, vol. 341, 1993, pages 643 - 647
DOUWES J; GIBSON P; PEKKANEN J ET AL.: "Non-eosinophilic asthma: importance and possible mechanisms", THORAX, vol. 57, 2002, pages 643 - 648
FABBRI L; PAUWELS RA; HURDS SS: "Global strategy for the diagnosis, management and prevention of chronic obstructive pulmonary disease: GOLD executive summary updated 2003", J COPD, vol. 1, 2004, pages 105 - 141
FAHY JV: "Eosinophilic and neutrophilic inflammation in asthma. Insight from Clinical studies", PROC AM THORAC SOC, vol. 6, 2009, pages 256 - 259, XP055255848, DOI: doi:10.1513/pats.200808-087RM
FAHY JV; KIM KW; LIU J ET AL.: "Prominent neutrophilic inflammation in sputum from subjects with asthma exacerbation", J ALLERGY CLIN !MMUNO!, vol. 95, 1995, pages 843 - 52, XP005128455, DOI: doi:10.1016/S0091-6749(95)70128-1
FITZGERALD MF; FOX JC: "Emerging trends in the therapy of COPD: novel anti- inflammatory agents in clinical development", DRUG DISCOV TODAY, vol. 12, no. 11/12, 2007, pages 479 - 485, XP007911674, DOI: doi:10.1016/j.drudis.2007.04.005
FREVERT CW; KINSELLA MG; VATHANAPRIDA C ET AL.: "Binding of interleikin-8 to heparin and chondroitin sulfate in lung tissue", AM J RESPIR CELL MOL BIOL, vol. 28, 2003, pages 464 - 472
GREEN RH; BRIGHTLING CE; WOLTMANN G ET AL.: "Analysis of induced sputum in adults with asthma: identification of subgroup with isolated sputum neutrophilia and poor response to inhaled corticosteroids", THORAX, vol. 57, 2002, pages 875 - 9
GUERASSIMOV A; HOSHINO Y; TAKUBO Y ET AL.: "The development of emphysema in cigarette smoke-exposed mice is strain dependent", AM J RESPIR CRIT CARE MED, vol. 170, 2004, pages 974 - 980
JATAKANON A; UASUF C; MAZIAK W ET AL.: "Neutrophilic inflammation in severe persistent asthma", AM J RESPIR CRIT CARE MED, vol. 160, 1999, pages 1532 - 9
KAMATH AV; PAVORD ID; RUPARELIA PR ET AL.: "Is the neutrophil the key effector cell in severe asthma?", THORAX, vol. 60, 2005, pages 529 - 530
KHARITONOV SA; SJOBRING U: "Lypopolysaccharide challenge in humans as a model for chronic obstructive lung disease exacerbations", CONTRIB MICROBIOL, vol. 14, 2007, pages 83 - 100
KIM V; ROGERS TJ; CRINER GJ: "New concepts in the pathology of Chronic Obstructive Pulmonary Disese", PROC AM THORAC SOC, vol. 5, 2008, pages 478 - 485
KITZ R; ROSE MA; PLACZEK K ET AL.: "LPS inhalation challenge: a new tool to characterize the inflammatory response in humans", MED MICROBIOL LMMUNOL, vol. 197, 2008, pages 13 - 19, XP019563718, DOI: doi:10.1007/s00430-007-0053-2
KOSHIKA T; ISHIZAKA A; NAGATOMI I ET AL.: "Pretreatment with FK506 improves survival rate and gas exchange in canine model of acute lung injury", AM J RESPIR CRIT CARE MED., vol. 163, no. 1, 2001, pages 79 - 84
LAMBLIN C; GOSSET P; TILLIE-LEBLOND I ET AL.: "Bronchial neutrophilia in patients with non infectious status asthmaticus", AM J RESPIR CRIT CARE MED, vol. 157, 1998, pages 394 - 402
LITTLE SA; MACLEOD KJ; CHALMERS GW ET AL.: "Association of forced expiratory volume with disease duration and sputum neutrophils in chronic asthma", AM J MED, vol. 112, 2002, pages 446 - 452
LOPEZ AD; MURRAY CC: "The global burden of disease", NAT MED, vol. 4, 1998, pages 1241 - 1243
MAHADEVA R; SHAPIRO SD: "Chronic obstructive pulmonary disease-3: experimental animal models of pulmonary emphysema", THORAX, vol. 57, 2002, pages 908 - 914
MAHALINGAM S; KARUPIAH G., IMMUNOL CELL BIOL., vol. 77, no. 6, December 1999 (1999-12-01), pages 469 - 75
MARIS NA; DE VOS AF; DESSING MC ET AL.: "Antiinflammatory effects of salmeterol after inhalation of lipopolysaccharide by healthy volunteers", AM J RESPIR CRIT CARE MED, vol. 172, no. 7, 2005, pages 878 - 884
MARTIN RJ; CICUTTO LC; SMITH HR ET AL.: "Airways inflammation in nocturnal asthma", AM REV RESPIR DIS, vol. 143, 1991, pages 351 - 357
MCDOUGALL CM; HELMES PJ.: "Neutrophil airway inflammation in childhood asthma", THORAX, vol. 61, no. 9, 2006, pages 739 - 741
NAKAMURA H; YOSHIMURA K; MCELVANEY NG ET AL.: "Neutrophil elastase in respiratory epithelial lining fluid of individuals with cystic fibrosis induces interleukin-8 gene expression in a human bronchial epithelial cell line", J CLIN INVES., vol. 89, 1992, pages 1478 - 1484
NOCKER RE; SCHOONBROOD DF; VAN DE GRAAF EA ET AL.: "Interleukin-8 in airway inflammation in patients with asthma and chronic obstructive pulmonary disease", INT ARCH ALLERGY !MMUNO!, vol. 109, 1996, pages 183 - 191
ORDONEZ CL; SHAUGHNESSY TE; MATTHAY MA ET AL.: "Increased neutrophil numbers and IL-8 levels in airway secretions in acute severe asthma: clinical and biological significance", AM J RESPIR CRIT CARE MED, vol. 161, 2000, pages 1185 - 1190
PAUWELS RA; BUIST AS; CALVERLEY PM ET AL.: "Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. NHLBI/WHO Global Initiative for Chronic Obstructive Pulmonary Disease (GOLD) Workshop summary", AM J RESPIR CRIT CARE MED., vol. 5, 2005, pages 1256 - 1276
PELEMAN RA; RYTILA PH; KIPS JC ET AL.: "The cellular composition of induced sputum in chronic obstructive pulmonary disease", EUR RESPIR J, vol. 13, 1999, pages 839 - 843
PUHA J; BADIA JR; ADHIKARI ET AL.: "Has mortality from acute respiratory distress syndrome decreased over time?: A systematic review", AM J RESPIR CRIT CARE MED, vol. 179, no. 3, 2009, pages 220 - 227
RATJEN FA: "Cystic fibrosis: pathogenesis and future treatment strategies", RESPIR CARE, vol. 54, no. 5, 2009, pages 595 - 602
RATJEN FA: "Diagnosing and managing infection in CF", PAEDIATR RESPIR REV, vol. 7, no. 1, 2006, pages S151 - S153, XP005513221, DOI: doi:10.1016/j.prrv.2006.04.217
REID PT; DONNELLY SC; HASLETT C: "Inflammatory predictors for the development of adult respiratory distress syndrome", THORAX, vol. 50, 1995, pages 10023 - 10026
REUTERSHAN J; BASIT A; GALKINA EV ET AL.: "Sequential recruitment of neutrophils into lung and bronchoalveolar lavage fluid in LPS-induced acute lung injury", AM J PHYSIOL CELL MOL PHYSIOL LUNG CELL MOL PHYSIOL, vol. 289, no. 5, 2005, pages L807 - L815
RIORDAN JR; ROMMENS JM; KEREM B ET AL.: "Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA", SCIENCE, vol. 245, 1989, pages 1066 - 1073
SAGEL SD; ACCURSO FJ: "Monitoring inflammation in CF. Cytokines", CLIN REV ALLERGY IMMUNOL., vol. 23, 2002, pages 41 - 57
SHAW DE; BERRY MA; HARGADON B ET AL.: "Association between neutrophilic airway inflammation and airflow limitation in adults with asthma", CHEST, vol. 132, 2007, pages 1871 - 1875
SMITH LS; KAJIKAWA O; ELSON G ET AL.: "effect of Toll-like receptor 4 blockade on pulmonary inflammation caused by mechanical ventilation and bacterial endotoxin", EXP LUNG RES, vol. 34, 2008, pages 225 - 245
SOUZA-FERNANDES AB; PELOSI P; ROCCO PRM: "Bench-to-bedside review: the role of glycosaminoglycans in respiratory disease", CRIT CARE, vol. 10, no. 6, 2006, pages 237 - 252, XP002584379, DOI: doi:10.1186/CC5069
SPARROW D; GLYNN RJ; COHEN M ET AL.: "The relationship of the peripheral leukocyte count and cigarette smiking to pulmonary function among adult men", CHEST, vol. 86, 1984, pages 383 - 386
SPRUIT MA; GOSSELINK R; TROOSTERS T ET AL.: "Muscle force during an acute exacerbation in hospitalized patients with COPD and its relationship with CXCL8 and IGF-I", THORAX, vol. 58, 2003, pages 752 - 756
SUR S; CROTTY TB; KEPHART GM ET AL.: "Sudden-onset fatal asthma: a distinct entity with few eosinophils and relatively more neutrophils in the airway submucosa?", AM REV RESPIR DIS, vol. 148, 1993, pages 713 - 719
TABARY O; CORVOL H; BONCOEUR E ET AL.: "Adherence of airway neuthrophils and inflammatory response are increased in Cf airway epithelial cell-neutrophil interactions", AM J PHYSIOL LUNG CELL MOL PHYSIOL, vol. 290, no. 3, 2006, pages L588 - 596
TABARY O; ZAHM JM; HINNRASKY J ET AL.: "Selective up-regulation of chemokine IL-8 expression in cystic fibrosis bronchial gland cells in vivo and in vitro", AM J PATHOL, vol. 153, 1998, pages 921 - 930, XP008158825, DOI: doi:10.1016/S0002-9440(10)65633-7
TANINO M; BETSUYAKU T; TAKEYABU K ET AL.: "Increased levels of interleukin-8 in BAL fluid from smokers susceptible to pulmonary emphysema", THORAX, vol. 57, 2002, pages 405 - 411
VAN DEN HOVEN R; DUVIGNEAU JC; HERTL RT ET AL.: "Clenbuterol affects the expression of messenger RNA for interleukin 10 in peripheral leukocytes from horses challenged intrabronchially with lipopolysaccharides", VET RES COMMUN, vol. 30, no. 8, 2006, pages 921 - 928, XP019455092, DOI: doi:10.1007/s11259-006-3383-4
VLAHOS R; BOZINOVSKI S; JONES JE ET AL.: "Differential protease, innate immunity, and NK-kB induction profiles during lung inflammation induced by subchronic cigarette smoke exposure in mice", AM J PHYSIOL LUNG CELL MOL PHYSIOL, vol. 290, 2006, pages L931 - L945, XP002439921, DOI: doi:10.1152/ajplung.00201.2005
WAERHAUG K; KUZKOV VV; KULIN VN ET AL.: "Inhaled aerosolized recombinant human protein C ameliorates endotoxin-induced lung injury in anesthetized sheep", CRIT CARE, vol. 13, no. 2, 2009, pages R51
WARE LB; MATTHAY MA: "The acute respiratory distress syndrome", N ENGL J MED, vol. 342, 2000, pages 1334 - 1349
WENZEL SE; SCHWARTZ LB; LANGMACK EL ET AL.: "Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics", AM J RESPIR CRIT CARE MED, vol. 160, 1999, pages 1001 - 8
WENZEL SE; SZEFLER SJ; LEUNG DYM ET AL.: "Bronchoscopic evaluation of severe asthma: persistent inflammation associated with high dose glucocorticoids", AM J RESPIR CRIT CARE MED, vol. 156, 1997, pages 737 - 473
WOOLHOUSE IS; BAYLEY DL; STOCKLEY RA: "Sputum chemotactic activity in chronic obstructive pulmonary disease. Effects of a1-antitrypsin deficiency and the role of leukotriene B4 and interleukine 8", THORAX, vol. 57, 2002, pages 709 - 714
WU Y; SINGER M; THOURON F ET AL.: "Effect of surfactant on pulmonary expression of type IIA PLA(2) in an animal model of acute lung injury", AM J PHYSIOL LUNG CELL MOL PHYSIOL, vol. 282, no. 4, 2002, pages L743 - 750
YAMAMOTO C; YONEDA T; YOSHIKAWAM ET AL.: "Airway inflammation in COPD assessed by sputum levels of interleukin-8", CHEST, vol. 112, 1997, pages 505 - 510, XP003023162, DOI: doi:10.1378/chest.112.2.505

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11840565B2 (en) 2016-12-29 2023-12-12 University Of Miami Methods and compositions for treating virus-associated inflammation
WO2018131724A1 (en) * 2017-01-13 2018-07-19 Tobishi Pharmaceutical Co., Ltd. Neutrophil activation regulator
WO2019243272A1 (en) * 2018-06-18 2019-12-26 College De France Ligand controling interaction between gags with their effector molecules and use thereof
CN112566921A (en) * 2018-06-18 2021-03-26 国家科学研究中心 Ligands that control the interaction between GAG and its effector molecules and uses thereof
US11357816B2 (en) 2018-06-18 2022-06-14 Centre National De La Recherche Scientifique (Cnrs) Ligand controling interaction between gags with their effector molecules and use thereof
KR20230141227A (en) 2022-03-31 2023-10-10 (주)케어젠 Peptide Having Activity of Anti-Inflammation and Anti-Fibrosis and Uses Thereof

Also Published As

Publication number Publication date
US20120288474A1 (en) 2012-11-15
EA201200470A1 (en) 2012-08-30
CN102596227A (en) 2012-07-18
EP2475380A1 (en) 2012-07-18
KR20120080196A (en) 2012-07-16
JP2013504545A (en) 2013-02-07
CA2773664A1 (en) 2011-03-17
AU2010294225A1 (en) 2012-04-05
ZA201201802B (en) 2013-05-29

Similar Documents

Publication Publication Date Title
EP2475380A1 (en) Composition for treatment of cxcl8-mediated lung inflammation
Borthwick The IL-1 cytokine family and its role in inflammation and fibrosis in the lung
Russo et al. Role of the chemokine receptor CXCR2 in bleomycin-induced pulmonary inflammation and fibrosis
Frevert et al. Functional characterization of the rat chemokine KC and its importance in neutrophil recruitment in a rat model of pulmonary inflammation.
Cameron et al. Airway epithelium expresses interleukin-18
US20230021168A1 (en) Methods for preventing or treating certain disorders by inhibiting binding of il-4 and/or il-13 to their respective receptors
Durham Mechanisms of mucosal inflammation in the nose and lungs.
ES2663379T3 (en) Treatment of inflammation and / or endotoxic shock
Yuksel et al. E-cadherin: an important functional molecule at respiratory barrier between defence and dysfunction
Garcia et al. Toxicological effects of fine particulate matter (PM2. 5): health risks and associated systemic injuries—systematic review
CN101484579B (en) High-affinity antagonists of elr-cxc chemokines
KR100699509B1 (en) Treatment with small peptides to effect antifibrotic activity
Luzina et al. Full-length IL-33 augments pulmonary fibrosis in an ST2-and Th2-independent, non-transcriptomic fashion
Massion et al. Staphylococcus aureus stimulates neutrophil recruitment by stimulating interleukin-8 production in dog trachea
JP2002515459A (en) Method for treating asthma and other allergic conditions and use of a composition comprising a TNF-RII (P75) agonist
US20070123464A1 (en) Methods and reagents for treating inflammation and fibrosis
KR20240086733A (en) Systemic sclerosis model platform with coronavirus infectious disease through the induction of pathogenic vimentin expression
Mitchell et al. The role of eotaxin and related CC-chemokines in asthma and allergy
Holgate Macrophage and Dendritic Cell Populations in Airway Tissues
JP2024525669A (en) IL1RA-derived peptides for the treatment of diabetic nephropathy - Patents.com

Legal Events

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

Ref document number: 201080050827.3

Country of ref document: CN

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

Ref document number: 10751694

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2773664

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2010294225

Country of ref document: AU

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2012528380

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2010751694

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2010294225

Country of ref document: AU

Date of ref document: 20100913

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20127008952

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 201200470

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 13395128

Country of ref document: US