US20150258133A1 - Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases - Google Patents

Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases Download PDF

Info

Publication number
US20150258133A1
US20150258133A1 US14/699,563 US201514699563A US2015258133A1 US 20150258133 A1 US20150258133 A1 US 20150258133A1 US 201514699563 A US201514699563 A US 201514699563A US 2015258133 A1 US2015258133 A1 US 2015258133A1
Authority
US
United States
Prior art keywords
cyclodextrin
inflammation
treatment
bronchial
mice
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/699,563
Inventor
Didier Cataldo
Brigitte Evrard
Agnes Noel
Jean-Michel Foldart
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Universite de Liege ULG
Original Assignee
Universite de Liege ULG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from EP04104957A external-priority patent/EP1655034A1/en
Application filed by Universite de Liege ULG filed Critical Universite de Liege ULG
Priority to US14/699,563 priority Critical patent/US20150258133A1/en
Assigned to UNIVERSITE DE LIEGE reassignment UNIVERSITE DE LIEGE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOEL, AGNES, EVRARD, BRIGITTE, CATALDO, DIDIER, FOIDART, JEAN-MICHEL
Publication of US20150258133A1 publication Critical patent/US20150258133A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/716Glucans
    • A61K31/724Cyclodextrins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0078Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a nebulizer such as a jet nebulizer, ultrasonic nebulizer, e.g. in the form of aqueous drug solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system

Definitions

  • the present invention relates to the use of cyclodextrin compound for the treatment and prevention of bronchial inflammatory diseases, including chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • bronchodilator also called reliever medications or nonbronchodilators antinflammatory agents referred to as controller agents, on the basis of their pharmacodynamic effects.
  • Short-acting bronchodilators such as inhaled beta agonist or anticholinergics are considered reliever medications.
  • Corticosteroids, cromolyn sodium, nedocromil sodium, sustained-release theophylline and long-acting beta agonist are considered controller medications, since they are used to achieve and maintain control of symptoms and are used daily on a long-term basis.
  • inhaled ⁇ 2-adrenergic agonists are drugs for relief of symptoms due to acute airway obstruction. They have a rapid onset of action and a 3-6 h duration of activity. Unfortunately they have side effects such as tachycardia, palpitations and tremor that often disappear during chronic administration.
  • Anticholinergic agents induce airway smooth muscle relaxation. Their activity is not as effective as beta agonists in asthma but is more prolonged (6 to 8 hours).
  • glucocorticosteroids are effective agents with anti-inflammatory effects.
  • side effects include adrenal suppression, osteoporosis, growth suppression, weight gain, hypertension, diabetes, dermal thinning, cataracts, myopathy and psychotic actions. These effects are dose related and are usually seen with systemic administration. Local side effects, including oral candidiasis and dysphonia may occur at lower doses of inhaled glucocorticoids.
  • Cromolyn sodium and nedocromil sodium are also classified as controller agents, because of their similar clinical profile. They inhibit bronchoconstriction induced by neurally mediated events.
  • Theophylline is generally considered as a bronchodilator but has weak bronchodilator activity in therapeutic doses. It may also have anti-inflammatory properties.
  • the dose-related adverse effects of theophylline are nausea, nervousness, anxiety and tachycardia.
  • Lipoxygenase inhibitors and leukotriene receptor agonists are also controller agents. They alter the pathological effects of leukotrienes derived from the 5-lipoxygenation of arachidonic acid. They can inhibit the bronchospastic effects of allergens, exercise, cold dry air, and aspirin allergy. Both are efficacious in alleviating symptoms and improving pulmonary function during 4-6 weeks of therapy in patients with moderate asthma.
  • cyclodextrin is useful as active component for the treatment or prevention of bronchial inflammatory diseases, including COPD.
  • the invention therefore provides the use of cyclodextrin compound for the treatment or prevention of bronchial inflammatory disease including COPD in a host mammal in need of such treatment.
  • cyclodextrin compound By cyclodextrin compound, one means cyclodextrin as well as their pharmaceutically acceptables salts, enantiomeric forms, diastereoisomers and racemates.
  • Cyclodextrin is also represented by CD hereafter.
  • Cyclodextrin compound according to the invention is cyclodextrin per se, alkyl-cyclodextrin (R-CD) wherein R is methyl, ethyl, propyl and butyl; carboxyalkyl-cyclodextrin(CR-CD), etherified-cyclodextrin (RO-CD), sulfoalkyl-cyclodextrin (SR-CD), hydroxyalkyl-cyclodextrin(HR-CD), glucosyl-cyclodextrin, di and triglycerides-cyclodextrin or a combination thereof and their pharmaceutically acceptable salts which are at least water soluble in an amount of 0.5 gr/100 ml at 25° C.
  • the water-soluble cyclodextrin compound preferably used in the present invention refers to a cyclodextrin compound having water solubility of at least that of ⁇ -cyclodextrin (1.85 g/100 ml).
  • water-soluble cyclodextrin compound examples include sulfobutylcyclodextrin, hydroxypropylcyclodextrin, maltosylcyclodextrin, and salts thereof.
  • sulfobutyl- ⁇ -cyclodextrin, hydroxypropyl- ⁇ -cyclodextrin, maltosyl- ⁇ -cyclodextrin, and salts thereof are examples of such water-soluble cyclodextrin compound.
  • methylcyclodextrins products of the cyclodextrins methylation
  • 2-O-methyl ⁇ -cyclodextrin such as 2-O-methyl ⁇ -cyclodextrin; dimethylcyclodextrin (DIMEB) (preferably substituted in 2 and in 6); trimethylcyclodextrin (preferably substituted in 2, 3 and 6);
  • Random methylated cyclodextrins (preferably substituted at random in 2, 3 and 6, but with a number of 1.7 to 1.9 methyl by unit glucopyrannose), hydroxypropylcyclodextrins (HPCD), hydroxypropylated cyclodextrins preferably substituted randomly mainly in position 2 and 3 (HP- ⁇ CD, HP- ⁇ CD)), sulfobutylethercyclodextrins (SBECD), hydroxyethyl-cyclodextrins, carboxymethylethylcyclodextrins, ethylcyclodextrins, cyclodextrins amphiphiles obtained by grafting hydrocarbonated chains in the hydroxyl groups and being able to form nanoparticles, cholesterol cyclodextrins and triglycerides-cyclodextrins obtained by grafting cyclodextrins monoaminated (with
  • cyclodextrin compounds according to the invention are produced by the well-known enzymatic degradation of starch such as the method described in “Cyclodextrin Technology, J Szejtli, Kluwer Academic Publishers 1998, pp 1-78, followed by grafting of an appropriate chemical group.
  • the invention further provides the use of such cyclodextrin compound for the manufacturing of a medicament for the treatment or prevention of bronchial inflammatory diseases to a patient in need of such treatment.
  • the cyclodextrin compound has to be administered to the patient over several months or years (especially in case of prevention).
  • the cyclodextrin compound is administered preferably as aerosol, with non-toxic doses ranging between nanomolar and molar concentrations.
  • the present invention relates to a method used for treating bronchial inflammatory diseases, preferably asthma and chronic obstructive pulmonary disease (COPD) in a host mammal in need of such treatment, e.g., a patient suffering from such a disease, by the application of a cyclodextrin compound according to the invention in a pharmaceutically effective amount.
  • COPD chronic obstructive pulmonary disease
  • the present invention provides cyclodextrins for controlling inflammation in COPD and COPD-related diseases.
  • Asthma is an inflammatory disease of the bronchial tree related or not to an allergen exposure. This inflammation provokes symptoms in patients by stimulating the bronchial smooth muscles to contract, enhancing the mucus secretion, and inducing bronchial morphological changes thought to be an aggravating factor regarding the course of the disease. Airway hyperresponsiveness is a hallmark of the disease and is responsible for most of symptoms.
  • the invention therefore further relates to a method for treating bronchial inflammatory diseases in a patient suffering from such a disease, using a cyclodextrin compound according to the invention in a therapeutically effective amount.
  • the invention preferably further relates to a method for treating emphysema in a patient suffering from such a disease, using cyclodextrin compounds according to the invention.
  • a disease the alveolar walls are destroyed by proteolytic processes and this destruction impairs the transfer of oxygen to the blood.
  • physiological problems also occurs because of the derived hyperinflation which causes abnormalities in the ventilation by causing a dysfunction of respiratory muscles and because of a hypertension in pulmonary arteries leading to cardiac failure in advanced stages.
  • the invention preferably further relates to a method for treating chronic obstructive pulmonary disease (COPD) in a patient suffering from such a disease, using cyclodextrin compounds according to the invention.
  • COPD chronic obstructive pulmonary disease
  • the bronchial walls of small airways are remodelled by proteolytic processes and this remodelling and fibrosis induce an airway obstruction which can be measured by spirometry.
  • physiological problems also occurs because of the derived hyperinflation which causes abnormalities in the ventilation/perfusion ratio and causes hypoventilation and eventually CO2 accumulation.
  • the cyclodextrin compound has to be administered over several months or years, to the patient in need of such a therapy.
  • the cyclodextrin compounds are administered preferably by the aerosolization of a liquid or powder composition, with non-toxic doses ranging between micro and molar concentrations per kg and day.
  • a further preferred object of the invention is a pharmaceutical composition of cyclodextrin compound according to the invention for the treatment of bronchial inflammatory diseases, and its use, containing a cyclodextrin or a salt thereof and preferably a water-soluble cyclodextrin derivative (water soluble being defined as a solubility of at least 0.5 g/100 ml water at 25° C.).
  • the pharmaceutical compositions are aqueous compositions having physiological compatibility.
  • the compositions include, in addition to cyclodextrin or a salt thereof, auxiliary substances, buffers, preservatives, solvents and/or viscosity modulating agents.
  • Appropriate buffer systems are based on sodium phosphate, sodium acetate or sodium borate.
  • Preservatives are required to prevent microbial contamination of the pharmaceutical composition during use. Suitable preservatives are, for example, benzalkonium chloride, chlorobutanol, methylparabene, propylparabene, phenylethyl alcohol, sorbic acid. Such preservatives are used typically in an amount of 0.01 to 1% weight/volume.
  • the cyclodextrin compound of the present invention exhibits its effects through either oral administration, parenteral administration or topical administration, and it is preferably formed into a composition for parenteral administration, particularly an injection composition or topical administration, particularly an aerosol composition.
  • a composition for parenteral administration particularly an injection composition or topical administration, particularly an aerosol composition.
  • aerosol composition is for example a solution, a suspension, a micronised powder mixture and the like.
  • the composition is administered by using a nebulizer, a metered dose inhaler or a dry powder inhaler or any device designed for such an administration.
  • galenic compositions include tablets, capsules, powders, granules and the like. These may be produced through well known technique and with use of typical additives such as excipients, lubricants, and binders.
  • Suitable auxiliary substances and pharmaceutical compositions are described in Remington's Pharmaceutical Sciences, 16th ed., 1980, Mack Publishing Co., edited by Oslo et al.
  • an appropriate amount of a pharmaceutically-acceptable salt is used in the composition to render the composition isotonic.
  • pharmaceutically acceptable substances include saline, Ringer's solution and dextrose solution. pH of the solution is preferably from about 5 to about 8, and more preferably from about 7 to about 7.5.
  • a preferred pharmaceutical composition for nebulization comprises cyclodextrin (CD), NaCl and water.
  • the solution is prepared by dissolving CD in 100 ml of purified water, adding NaCl by stirring so as to dissolve them and complete with water so as to obtain 200 ml of solution.
  • the solution is sterilized by filtration through a 0.22 ⁇ m polypropylene membrane or by a steam sterilization process.
  • composition is a combination of (for 200 ml of solution):
  • the present invention provides a method for the treatment of COPD (Chronic obstruction pulmonary disease) in a host mammal in need of such treatment, comprising the step of administering an effective amount of cyclodextrin compound to the mammal, wherein the cyclodextrin compound is selected from the group consisting of b-cyclodextrin, hydroxypropyl-bcyclodextrin, sulfolbutylether-bcyclodextrin, random methylated-bcyclodextrin, dimethyl-bcyclodextrin, trimethyl-bcyclodextrin, hydroxypropyl b-cyclodextrin, hydroxybutyl bcyclodextrin, glucosyl-bcyclodextrin, maltosyl-bcyclodextrin, 2-O-methyl-bcyclodextrin or a combination thereof and their pharmaceutically acceptable salts.
  • the cyclodextrin compound is selected from the group
  • FIGS. 1-2 Effects of inhalation of cyclodextrin compound on BAL eosinophil percentage ( FIG. 1 ) and peribronchial inflammation score ( FIG. 2 ).
  • Controls are mice exposed to ova by inhalation and placebo by inhalation (OVA)
  • FIG. 3 Effects of inhalation of cyclodextrin compound on peribronchial eosinophils reported here as a number/mm of epithelial basement membrane.
  • FIG. 4 Airway responsiveness measurements: Enhanced Pause (Penh) was measured in OVA exposed mice during 5 minutes after a 2 minutes inhalation of cyclodextrin or Placebo (OVA) and increasing doses of methacholine (Mch).
  • Penh Enhanced Pause
  • OVA cyclodextrin or Placebo
  • Mch methacholine
  • FIG. 6 Measurement of allergen specific IgE levels in serum.
  • FIGS. 7A-B Measurement of eotaxin and IL-13 in Bal and lung protein extracts.
  • IL-13 was measured by incubation in a wheel coated with a primary antibody specifically dedicated to the recognition of the protein and after rinsing, a second antibody against IL-13, coupled with horse radish peroxydase was used to quantify the amounts of eotaxin in the solution.
  • FIG. 11 Levels of IL-13 measured by Elisa in lung protein extracts.
  • PBS aerosolized placebo
  • HPBeta hydroxypropyl- ⁇ -cyclodextrin
  • compositions Containing HP- ⁇ -Cyclodextrin for Therapy of Allergen-Induced Airway Inflammation and Bronchial Hyperresponsiveness in a Mouse Model of Asthma
  • ⁇ - and HP- ⁇ -CD were obtained from Wacker Chemie Gmbh (Germany).
  • Apyrogenic phosphate buffered saline (PBS) was purchased from Bio-Wittaker (Verviers, Belgium).
  • Methacholine was from Sigma-Aldrich (Germany).
  • Aerosol was produced by using an ultrasonic nebuliser SYSTAM, the vibration frequency of which is 2.4 MHz with variable vibration intensity and ventilation levels. Vibration intensity was fixed in position 6 and the ventilation level was 25 (t1 ⁇ 2) Umin.
  • SYSTAM ultrasonic nebuliser
  • Aerosol size distribution emitted from CDs solutions was determined with a laser size analyzer Mastersizer (Malvern, Orsay, France). Ten milliliters of each solution were directly nebulized in the laser beam. The mouth piece was held at 1 cm from the center of the laser beam. The resulting aerosol was aspirated on the opposite side of the beam. Environmental temperature and relative humidity were maintained constant, that is to say at 20° C. and 40-45%. Triplicates of each measurement were performed and compared to controls of PBS. The results are expressed as the percentage of droplets comprised in the range 0.5 to 5.79 ⁇ m and the median diameter. The concentration of droplets in the air evaluated by the obscuration percentage of the laser beam was in the same range for each experiment (15-25%).
  • OVA ovalbumin
  • AlumInject Perbio, Erembodegem, Belgium
  • Mice were subsequently exposed to allergens by daily inhalation of an aerosol of OVA 1%, for 30 minutes, generated by ultrasonic nebulizer (Devilbiss 2000), from day 21 to 27.
  • Mice were subjected to inhalation of ⁇ -CD, HP- ⁇ -CD 1, 7.5, 50 mM and HP- ⁇ -CD 50 mM 30 minutes before OVA inhalation. Mice were sacrificed performed on day 28 as previously reported by Cataldo and al in Am. J. Pathol 2002; 161(2):491-498.
  • Mices were sacrificed and a bronchoalveolar lavage was performed using 4 ⁇ 1 ml PBS-EDTA 0.05 mM (Calbiochem, Darmstadt, Germany) as previously described by Cataldo D D, Tournoy K G, Vermaelen K et al. in Am J Pathol 2002; 161(2):491-498. Cells were recovered by gentle manual aspiration. After centrifugation of bronchoalveolar fluid (BALF) (1200 rpm for 10 minutes, at 4° C.), the supernatant was frozen at ⁇ 80° C. for protein assessment and the cell pellet was resuspended in 1 ml PBS-EDTA 0.05 mM. The differential cell counts were performed on cytocentrifuged preparations (Cytospin) after staining with Diff-Quick (Dade, Belgium).
  • BALF bronchoalveolar fluid
  • a value of 0 was adjudged when no inflammation was detectable, a value of 1 when there was occasionally inflammatory cells, a value of 2 when most bronchi were surrounded by a thin layer (1 to 5 cells) of inflammatory cells and a value of 3 when most bronchi were surrounded by a thick layer (>5 cells) of inflammatory cells. Since 5-7 randomly selected tissue sections per mouse were scored, inflammation scores are expressed as a mean value and can be compared between groups. After Congo Red staining, the eosinophilic infiltration in the airway walls was quantified by manual count and reported to the perimeter of epithelial basement membrane defining an eosinophilic inflammatory score.
  • the left lung was crushed using a Mikro-Dismembrator (Braun Biotech International, Gmbh Melsungen, Germany).
  • the crushed lung tissue was incubated overnight at 4° C. in a solution containing 2M urea, 1M NaCl and 50 mM Tris (pH 7.5) and subsequently centrifuged 15 minutes at 16.000 ⁇ g. The supernatant was stored at ⁇ 80° C.
  • Bronchial responsiveness measurement Twenty-four hours after the last allergen exposure, the bronchial hyper responsiveness was determined by measuring the Penh (Enhanced Pause) using a barometric plethysmograph (Emka technologies, Paris) as proposed by Hamelmann, E., et al., Am. J Respir. Crit. Care Med. 156 (1997) 766-775). The Penh was measured at baseline and 5 min after the inhalation of increasing doses (25, 50, 75 and 100 mM) of methacholine (Mch).
  • Eotaxin and IL-13 levels were assessed using commercial ELISAs (R&D systems, Abingdon, UK). Eotaxin was measured by incubation in a wheel coated with a primary antibody specifically dedicated to the recognition of the protein and after rinsing, a second antibody against eotaxin, coupled with horse radish peroxydase was used to quantify the amounts of eotaxin in the solution
  • IL-13 was measured by incubation in a wheel coated with a primary antibody specifically dedicated to the recognition of the protein and after rinsing, a second antibody against IL-13, coupled with horse radish peroxydase was used to quantify the amounts of eotaxin in the solution.
  • Results of BAL cell count, pulmonary histology, cytokines and mRNA levels were expressed as mean+/ ⁇ SEM and the comparison between the groups was performed using Mann-Whitney test.
  • Mann-Whitney test was performed using GRAPHPAD INSTAT version 3.00 for WINDOWS 95 (GRAPHPAD SOFTWARE, San Diego, Calif., USA, WWW.GRAPHPAD.Com.). P values ⁇ 0.05 were considered as significant.
  • mice treated with placebo displayed a significant increase in peribronchial inflammation as quantified by the peribronchial inflammation score.
  • Mice treated with HP- ⁇ -CD 1, 7.5, and 50 mM and HP- ⁇ -CD 50 mM were shown to have decreased inflammation score when compared to placebo treated mice.
  • ⁇ -CD inhalation did not reduce the peribronchial inflammation score ( FIG. 2 ).
  • HP- ⁇ -CD 50 mM reduced the methacholine-induced Penh increase ( FIG. 4 ).
  • HP- ⁇ -CD 50 mM was the only to show a significant decrease ( FIG. 5 ).
  • compositions Comprising 2-O-Methyl-Cyclodextrin for Therapy of Allergen-Induced Airway Inflammation and Bronchial Hyperresponsiveness in a Mouse Model of Asthma
  • cyclodextrin compound which is here 2-O-methyl-cyclodextrin, KLEPTOSE CRYSMEB®, a product commercialised by Roquette. It has, on average, 4 methyl groups per native cyclodextrin molecule and is characterized by an average molecular weight of 1135 and a average molar degree of substitution of 0.57.
  • mice Same methods are used as in example 1 but in the present example we did expose mice to aerosolized CRYSMEB (10, 20, 50, 100 or 200 mM) in a standard exposure box (20 ⁇ 30 ⁇ 15 cm) for 30 min/day during 7 days.
  • the cellular composition of the bronchoalveolar lavage was not significantly altered by the exposure to CRYSMEB. In particular, there were no differences regarding eosinophil and neutrophil counts (see table 1).
  • Bronchoalveolar lavage eosinophilia was significantly decreased in the groups treated by CRYSMEB.
  • the decrease in lavage eosinophilia was comparable with that obtained with different concentrations of HP-beta-cyclodextrins or fluticasone, a commonly used inhalation steroid used as a reference therapy (table 2)
  • mice treated with placebo displayed a significant increase in peribronchial inflammation as quantified by the peribronchial inflammation score.
  • Mice treated with CRYSMEB 20 mM were shown to have decreased inflammation score when compared to placebo treated mice ( FIG. 9A ).
  • Peribronchial inflammation score was measured and was significantly decreased in every treatment group as compared to placebo ( FIG. 9B )
  • IL-13 a major Th2 cytokine implicated in the airway hyperresponsiveness and inflammation.
  • levels of IL-13 measured by ELISA in whole lung protein extracts were significantly decreased by the exposure to CRYSMEB as well as fluticasone and HP-beta-CD 50 mM. (see FIG. 11 )
  • composition to be Administered in an Aerosol to a Patient in Need of Treatment for Bronchial Inflammatory Disease
  • HP betaCD 75 mM Solution osmolality is 308 mOs/kg. pH is 7.2.
  • the concentration of CD compound can be modified in function of the requirements. It is preferred to adjust the tonicity by addition of NaCl.
  • a preferred composition for nebulization is:
  • OVA ovalbumin
  • mice were immunized by intraperitoneal injection of OVA (10 ⁇ g) (Sigma Aldrich, Schnelldorf, Germany) and aluminium hydroxyde on days 0 and 7. From days 21 to 25, mice were exposed to inhalation of 1% OVA or PBS (phosphate buffer saline) for 30 minutes. Airway hyperresponsiveness was measured on day 26 before sacrifice. Mice were either exposed to aerosolized hydroxypropyl-beta-cyclodextrin (15 mM) or PBS for 30 min 6 hours before allergen exposure.
  • thorax was opened and the right lungs were excised and snap frozen in liquid nitrogen for protein extraction.
  • the left lung was insufflated at constant pressure with 4% paraformaldehyde and embedded in paraffin for further histological analysis.
  • a peribronchial inflammation score was applied on each hematoxylin-eosin stained slide as previously reported (Cataldo et al Am J Pathol 2002).
  • a value from 0 to 2 was adjudged to each bronchus. Score of 0 corresponded to bronchi without inflammation; score 1 to occasional mononuclear cells observed around bronchi, and score 2 to 1 to 5 layer(s) of inflammatory cells around bronchi.
  • bronchi per mice Six bronchi per mice were counted and statistical analysis was performed by using GraphPad Program. Lung tissues were crushed and total protein extracts were prepared by incubating crushed lung tissues in a 2M urea solution. Tissue lysates were centrifuged for 15 min at 16100 ⁇ g. ELISA for CXCL-1 on lung protein extracts were assessed using antibodies from R&D Systems and the R&D Duoset® Elisa Development kit (R&D Systems, Minneapolis, Minn., USA).
  • mice were exposed to 3 ⁇ g LPS diluted in 100 ⁇ l of PBS or PBS (phosphate buffer saline) only administered by direct tracheal instillations at day 1 and 5 of the protocol. Mice were daily exposed either to aerosolized hydroxypropyl-beta-cyclodextrin (15 mM) or PBS for 30 min from day 0 to 5 (6 hours after LPS or sham exposure at days 1 and 5). Mice were sacrificed at day 6.
  • PBS lipopolysaccharide
  • thorax was opened and the right lungs were excised and snap frozen in liquid nitrogen for protein extraction.
  • the left lung was insufflated at constant pressure with 4% paraformaldehyde and embedded in paraffin for further histological analysis.
  • a peribronchial inflammation score was applied on each hematoxylin-eosin stained slide as previously reported (Cataldo et al Am J Pathol 2002).
  • a value from 0 to 2 was adjudged to each bronchus. Score of 0 corresponded to bronchi without inflammation; score 1 to occasional mononuclear cells observed around bronchi, and score 2 to 1 to 5 layer(s) of inflammatory cells around bronchi.
  • Six bronchi per mice were counted and statistical analysis was performed by using GraphPad Program.
  • Lung tissues were crushed and total protein extracts were prepared by incubating crushed lung tissues in a 2M urea solution. Tissue lysates were centrifuged for 15 min at 16100 ⁇ g. ELISA for CXCL-1 on lung protein extracts were assessed using antibodies from R&D Systems and the R&D Duoset® Elisa Development kit (R&D Systems, Minneapolis, Minn., USA).
  • CXCL-1 a chemotactic agent for neutrophils

Landscapes

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

Abstract

The invention provides the use of a cyclodextrin compound for the manufacturing of a medicament for the treatment or prevention of bronchial inflammatory diseases, particularly for asthma and chronic obstructive pulmonary disease (COPD).

Description

    FIELD OF THE INVENTION
  • The present invention relates to the use of cyclodextrin compound for the treatment and prevention of bronchial inflammatory diseases, including chronic obstructive pulmonary disease (COPD).
  • BACKGROUND OF THE INVENTION
  • Compounds for the treatment and prevention of bronchial inflammatory disesases including COPD are classified in the art as bronchodilator also called reliever medications or nonbronchodilators antinflammatory agents referred to as controller agents, on the basis of their pharmacodynamic effects.
  • Short-acting bronchodilators such as inhaled beta agonist or anticholinergics are considered reliever medications. Corticosteroids, cromolyn sodium, nedocromil sodium, sustained-release theophylline and long-acting beta agonist are considered controller medications, since they are used to achieve and maintain control of symptoms and are used daily on a long-term basis.
  • Among reliever medication, inhaled β2-adrenergic agonists are drugs for relief of symptoms due to acute airway obstruction. They have a rapid onset of action and a 3-6 h duration of activity. Unfortunately they have side effects such as tachycardia, palpitations and tremor that often disappear during chronic administration.
  • Anticholinergic agents induce airway smooth muscle relaxation. Their activity is not as effective as beta agonists in asthma but is more prolonged (6 to 8 hours).
  • Among controller medications, glucocorticosteroids are effective agents with anti-inflammatory effects. Unfortunately, their side effects include adrenal suppression, osteoporosis, growth suppression, weight gain, hypertension, diabetes, dermal thinning, cataracts, myopathy and psychotic actions. These effects are dose related and are usually seen with systemic administration. Local side effects, including oral candidiasis and dysphonia may occur at lower doses of inhaled glucocorticoids.
  • Cromolyn sodium and nedocromil sodium are also classified as controller agents, because of their similar clinical profile. They inhibit bronchoconstriction induced by neurally mediated events.
  • Theophylline is generally considered as a bronchodilator but has weak bronchodilator activity in therapeutic doses. It may also have anti-inflammatory properties. The dose-related adverse effects of theophylline are nausea, nervousness, anxiety and tachycardia.
  • Lipoxygenase inhibitors and leukotriene receptor agonists are also controller agents. They alter the pathological effects of leukotrienes derived from the 5-lipoxygenation of arachidonic acid. They can inhibit the bronchospastic effects of allergens, exercise, cold dry air, and aspirin allergy. Both are efficacious in alleviating symptoms and improving pulmonary function during 4-6 weeks of therapy in patients with moderate asthma.
  • There is therefore a need for improved compounds which can be used for the treatment or prevention of bronchial inflammatory diseases including COPD.
  • BRIEF SUMMARY OF THE INVENTION
  • It is now surprisingly found that cyclodextrin is useful as active component for the treatment or prevention of bronchial inflammatory diseases, including COPD.
  • The invention therefore provides the use of cyclodextrin compound for the treatment or prevention of bronchial inflammatory disease including COPD in a host mammal in need of such treatment.
  • By cyclodextrin compound, one means cyclodextrin as well as their pharmaceutically acceptables salts, enantiomeric forms, diastereoisomers and racemates.
  • By cyclodextrin, one means cyclic oligosaccharides produced by enzymatic degradation of starch such as described in “Cyclodextrin Technology, J Szejtli, Kluwer Academic Publishers 1998, pp 1-78”, and which are composed of a variable number of glucopyrannose units (n), mostly 6, 7 or 8. These cyclodextrins are respectively named α, β and γ cyclodextrins (α CD, β CD, γCD).
  • Figure US20150258133A1-20150917-C00001
  • Cyclodextrin is also represented by CD hereafter.
  • Cyclodextrin compound according to the invention is cyclodextrin per se, alkyl-cyclodextrin (R-CD) wherein R is methyl, ethyl, propyl and butyl; carboxyalkyl-cyclodextrin(CR-CD), etherified-cyclodextrin (RO-CD), sulfoalkyl-cyclodextrin (SR-CD), hydroxyalkyl-cyclodextrin(HR-CD), glucosyl-cyclodextrin, di and triglycerides-cyclodextrin or a combination thereof and their pharmaceutically acceptable salts which are at least water soluble in an amount of 0.5 gr/100 ml at 25° C.
  • The water-soluble cyclodextrin compound preferably used in the present invention refers to a cyclodextrin compound having water solubility of at least that of β-cyclodextrin (1.85 g/100 ml).
  • Examples of such water-soluble cyclodextrin compound are sulfobutylcyclodextrin, hydroxypropylcyclodextrin, maltosylcyclodextrin, and salts thereof. In particular, sulfobutyl-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, maltosyl-β-cyclodextrin, and salts thereof
  • Other preferred cyclodextrin compound according to the invention are methylcyclodextrins (products of the cyclodextrins methylation) such as 2-O-methylβ-cyclodextrin; dimethylcyclodextrin (DIMEB) (preferably substituted in 2 and in 6); trimethylcyclodextrin (preferably substituted in 2, 3 and 6);
  • “random methylated” cyclodextrins (RAMEB or RM) (preferably substituted at random in 2, 3 and 6, but with a number of 1.7 to 1.9 methyl by unit glucopyrannose), hydroxypropylcyclodextrins (HPCD), hydroxypropylated cyclodextrins preferably substituted randomly mainly in position 2 and 3 (HP-βCD, HP-γ CD)), sulfobutylethercyclodextrins (SBECD), hydroxyethyl-cyclodextrins, carboxymethylethylcyclodextrins, ethylcyclodextrins, cyclodextrins amphiphiles obtained by grafting hydrocarbonated chains in the hydroxyl groups and being able to form nanoparticles, cholesterol cyclodextrins and triglycerides-cyclodextrins obtained by grafting cyclodextrins monoaminated (with a spacer arm) as described in Critical Review in Therapeutic drug Carrier Systems, Stephen D. Bruck Ed, Cyclodextrin-Enabling Excipient; their present and future use in Pharmaceuticals, D. Thomson, Volume 14, Issue 1 p 1-114 (1997)
  • Most preferred cyclodextrins compounds are
      • β-cyclodextrin with optionally a chemical function grafted on the glucopyrannose units such as hydroxypropyl-βcyclodextrin(HPβCD), sulfonylbutylether-βcyclodextrin(SBEβCD), random methylated-βcyclodextrin(RMβCD), dimethyl-βcyclodextrin(DIMEβCD), trimethyl-βcyclodextrin(TRIMEβCD), hydroxybutyl βcyclodextrin(HBβCD), glucosyl βcyclodextrin, maltosyl βcyclodextrin and 2-O-methyl βcyclodextrin(Crysmeb), or a combination thereof and their pharmaceutically acceptable salts.
  • The cyclodextrin compounds according to the invention are produced by the well-known enzymatic degradation of starch such as the method described in “Cyclodextrin Technology, J Szejtli, Kluwer Academic Publishers 1998, pp 1-78, followed by grafting of an appropriate chemical group.
  • The invention further provides the use of such cyclodextrin compound for the manufacturing of a medicament for the treatment or prevention of bronchial inflammatory diseases to a patient in need of such treatment.
  • According to the invention the cyclodextrin compound has to be administered to the patient over several months or years (especially in case of prevention). The cyclodextrin compound is administered preferably as aerosol, with non-toxic doses ranging between nanomolar and molar concentrations.
  • The present invention relates to a method used for treating bronchial inflammatory diseases, preferably asthma and chronic obstructive pulmonary disease (COPD) in a host mammal in need of such treatment, e.g., a patient suffering from such a disease, by the application of a cyclodextrin compound according to the invention in a pharmaceutically effective amount. The present invention provides cyclodextrins for controlling inflammation in COPD and COPD-related diseases.
  • In COPD patients, there is a significant neutrophilic inflammation in the bronchial walls leading to progressive destruction of airways structures by the repeated productions of proteases and oxidants (oxygen reactive species). To date, marketed therapies are not able to adequately decrease or prevent this neutrophilic inflammation in COPD patients. In particular, it is well know that inhaled or oral steroids display no efficiency against neutrophilic inflammation. For example, a study of S. Culpitt et al (Am J Respir Crit Care Med (1999) 160: 1635-1639) performed in COPD patients reports the lack of efficacy of high doses inhaled steroids in COPD-related neutrophilic inflammation and chemotactic agents for neutrophils (mainly IL-8 in humans).
  • It is well established that neutrophils are the major inflammatory cells in COPD and that these cells are attracted in the airways by potent chemotactic agents such as IL-8 in human (V. Murugan et al, Exp Lung Res. 2009 August; 35(6):439-85). In rodents, the cytokine system is similar but chemotactic agents responsible for neutrophils chemotaxis are slightly different and include CXCL-1 (also named KC) as a major chemoattractant playing a similar role to human IL-8 especially documented in the context of tobacco smoke exposure (V. Lagente Clin Exp Pharmacol Physiol. 2008 May; 35(5-6):601-5). Lipopolysaccharide (LPS), a component of external membrane of gram-negative bacteria, is able to induce a neutrophilic inflammation with characteristics similar to that observed in COPD and is therefore considered as a suitable model of COPD. Pathways activated by LPS that lead to neutrophilic inflammation are similar to those observed in COPD and include CXCL-1 production (A. Roos Biochem Biophys Res Commun. 2012 Jun. 22; 423(1):134-9).
  • Asthma is an inflammatory disease of the bronchial tree related or not to an allergen exposure. This inflammation provokes symptoms in patients by stimulating the bronchial smooth muscles to contract, enhancing the mucus secretion, and inducing bronchial morphological changes thought to be an aggravating factor regarding the course of the disease. Airway hyperresponsiveness is a hallmark of the disease and is responsible for most of symptoms. Bronchial tree is a very complex tissue with many cell types (as for example epithelial cells, smooth muscle cells, inflammatory cells, nerves, mucus producing cells, fibroblasts) and the bronchial remodelling events which comprise many aspects mainly consist in a deposition of extracellular matrix components in the bronchial walls, a smooth muscle hyperplasia and a hyperplasia of the mucus producing cells. The use of cyclodextrin compounds according to the invention inhibits the inflammatory cells influx in the compartments of bronchoalveolar lavage and peribronchial tissue and inhibits the hyperresponsiveness defined as an abnormal response to stimulating agents such as methacholine. The disease and current treatments are reviewed in, e.g., GINA Workshop Report, Global Strategy for Asthma Management and Prevention (NIH Publication No. 02-3659) and Fabbri, L. M., and Hurd, S. S., Eur. Respir. J. 22 (2003) 1-2.
  • The invention therefore further relates to a method for treating bronchial inflammatory diseases in a patient suffering from such a disease, using a cyclodextrin compound according to the invention in a therapeutically effective amount.
  • The invention preferably further relates to a method for treating emphysema in a patient suffering from such a disease, using cyclodextrin compounds according to the invention. In such a disease, the alveolar walls are destroyed by proteolytic processes and this destruction impairs the transfer of oxygen to the blood. In such a disease, physiological problems also occurs because of the derived hyperinflation which causes abnormalities in the ventilation by causing a dysfunction of respiratory muscles and because of a hypertension in pulmonary arteries leading to cardiac failure in advanced stages.
  • The invention preferably further relates to a method for treating chronic obstructive pulmonary disease (COPD) in a patient suffering from such a disease, using cyclodextrin compounds according to the invention. In such a disease, the bronchial walls of small airways are remodelled by proteolytic processes and this remodelling and fibrosis induce an airway obstruction which can be measured by spirometry. In such a disease, physiological problems also occurs because of the derived hyperinflation which causes abnormalities in the ventilation/perfusion ratio and causes hypoventilation and eventually CO2 accumulation.
  • According to the invention the cyclodextrin compound has to be administered over several months or years, to the patient in need of such a therapy. The cyclodextrin compounds are administered preferably by the aerosolization of a liquid or powder composition, with non-toxic doses ranging between micro and molar concentrations per kg and day.
  • A further preferred object of the invention is a pharmaceutical composition of cyclodextrin compound according to the invention for the treatment of bronchial inflammatory diseases, and its use, containing a cyclodextrin or a salt thereof and preferably a water-soluble cyclodextrin derivative (water soluble being defined as a solubility of at least 0.5 g/100 ml water at 25° C.).
  • The pharmaceutical compositions are aqueous compositions having physiological compatibility. The compositions include, in addition to cyclodextrin or a salt thereof, auxiliary substances, buffers, preservatives, solvents and/or viscosity modulating agents. Appropriate buffer systems are based on sodium phosphate, sodium acetate or sodium borate. Preservatives are required to prevent microbial contamination of the pharmaceutical composition during use. Suitable preservatives are, for example, benzalkonium chloride, chlorobutanol, methylparabene, propylparabene, phenylethyl alcohol, sorbic acid. Such preservatives are used typically in an amount of 0.01 to 1% weight/volume.
  • The cyclodextrin compound of the present invention exhibits its effects through either oral administration, parenteral administration or topical administration, and it is preferably formed into a composition for parenteral administration, particularly an injection composition or topical administration, particularly an aerosol composition. Such aerosol composition is for example a solution, a suspension, a micronised powder mixture and the like. The composition is administered by using a nebulizer, a metered dose inhaler or a dry powder inhaler or any device designed for such an administration.
  • Examples of galenic compositions include tablets, capsules, powders, granules and the like. These may be produced through well known technique and with use of typical additives such as excipients, lubricants, and binders.
  • Suitable auxiliary substances and pharmaceutical compositions are described in Remington's Pharmaceutical Sciences, 16th ed., 1980, Mack Publishing Co., edited by Oslo et al. Typically, an appropriate amount of a pharmaceutically-acceptable salt is used in the composition to render the composition isotonic. Examples of pharmaceutically acceptable substances include saline, Ringer's solution and dextrose solution. pH of the solution is preferably from about 5 to about 8, and more preferably from about 7 to about 7.5.
  • A preferred pharmaceutical composition for nebulization comprises cyclodextrin (CD), NaCl and water. The solution is prepared by dissolving CD in 100 ml of purified water, adding NaCl by stirring so as to dissolve them and complete with water so as to obtain 200 ml of solution. Preferably the solution is sterilized by filtration through a 0.22 μm polypropylene membrane or by a steam sterilization process.
  • Especially preferred composition is a combination of (for 200 ml of solution):
      • 10-50 g CD, preferably 20 gCD, preferably HPβCD; sodium chloride 1.2-1.5 g, preferably 1.42 g (isotonicity) and water, preferably pyrogen-free, sterile, purified water ad 200 ml.
  • Such a composition is useful for the treatment of bronchial inflammatory diseases.
  • Most preferred composition is a combination of 2-O-methylβCD with sodium chloride 1.2-1.5 g, preferably 1.42 g (isotonicity) and water, preferably pyrogen-free, sterile, purified water ad 200 ml.
  • In summary, the present invention provides a method for the treatment of COPD (Chronic obstruction pulmonary disease) in a host mammal in need of such treatment, comprising the step of administering an effective amount of cyclodextrin compound to the mammal, wherein the cyclodextrin compound is selected from the group consisting of b-cyclodextrin, hydroxypropyl-bcyclodextrin, sulfolbutylether-bcyclodextrin, random methylated-bcyclodextrin, dimethyl-bcyclodextrin, trimethyl-bcyclodextrin, hydroxypropyl b-cyclodextrin, hydroxybutyl bcyclodextrin, glucosyl-bcyclodextrin, maltosyl-bcyclodextrin, 2-O-methyl-bcyclodextrin or a combination thereof and their pharmaceutically acceptable salts. Preferably, the cyclodextrin compound is hydroxypropyle-beta-cyclodextrine and its pharmaceutically acceptable salts. Preferably, the mode of administration is inhalation.
  • The following examples, references, and figures are provided to aid the understanding of the present invention. It is understood that modifications can be made in the procedures set forth without departing from the invention.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1-2 Effects of inhalation of cyclodextrin compound on BAL eosinophil percentage (FIG. 1) and peribronchial inflammation score (FIG. 2). Controls are mice exposed to ova by inhalation and placebo by inhalation (OVA)
  • FIG. 3 Effects of inhalation of cyclodextrin compound on peribronchial eosinophils reported here as a number/mm of epithelial basement membrane.
  • FIG. 4 Airway responsiveness measurements: Enhanced Pause (Penh) was measured in OVA exposed mice during 5 minutes after a 2 minutes inhalation of cyclodextrin or Placebo (OVA) and increasing doses of methacholine (Mch).
  • FIG. 5 Measurement of cytokines by Elisa. Eotaxin was measured by incubation in a wheel coated with a primary antibody specifically dedicated to the recognition of the protein and after rinsing, a second antibody against eotaxin, coupled with horse radish peroxydase was used to quantify the amounts of eotaxin in the solution.
  • FIG. 6 Measurement of allergen specific IgE levels in serum.
  • FIGS. 7A-B Measurement of eotaxin and IL-13 in Bal and lung protein extracts. IL-13 was measured by incubation in a wheel coated with a primary antibody specifically dedicated to the recognition of the protein and after rinsing, a second antibody against IL-13, coupled with horse radish peroxydase was used to quantify the amounts of eotaxin in the solution.
  • FIG. 8 Airway responsiveness measurements: Enhanced Ponse (Penh) was measured in mouses after receiving crysmeb or placebo treatment FIGS. 9A-B Peribronchial inflammation score measured in histology when treated with various Crysmeb concentration compared to placebo (FIG. 9A) and to other cyclodextrin and fluticasone (FIG. 9B)
  • FIG. 10 Airway responsiveness measurements: comparison of cyclodextrin compounds with placebo and Fluticasone. Measurements of methacholine-induced airway response in mice exposed 7 days to allergens and receiving an inhaled therapy 30 min before the allergen exposition.
  • FIG. 11 Levels of IL-13 measured by Elisa in lung protein extracts.
  • FIG. 12 Measurement of CXCL-1 by ELISA in whole lung tissue extracts (crushed in liquid nitrogen) ***p<0.01 (n=8/group).
  • FIG. 13 Measurements of CXCL-1 levels in lung protein extracts of mice exposed to aerosolized placebo (PBS) or hydroxypropyl-β-cyclodextrin (HPBeta) at 15 mM (n=6/group).
  • FIG. 14 Measurements of peribronchial inflammation by histology (see <<experimental protocol>> section for description of the scoring system) (n=6/group).
  • DETAILED DESCRIPTIONS OF THE INVENTION Example 1 Use of Compositions Containing HP-β-Cyclodextrin for Therapy of Allergen-Induced Airway Inflammation and Bronchial Hyperresponsiveness in a Mouse Model of Asthma Materials
  • HP-β-CD (degree of substitution=0.64) was obtained from Roquette (France). α- and HP-γ-CD were obtained from Wacker Chemie Gmbh (Germany). Apyrogenic phosphate buffered saline (PBS) was purchased from Bio-Wittaker (Verviers, Belgium). Methacholine was from Sigma-Aldrich (Germany).
  • All other materials were of analytical grade. Sterile water for injection was used throughout this study. Sterile, apyrogenic and isotonic CD solutions were prepared at 1, 7.5 and 50 mM for HP-β-CD and α-CD and at 50 mM for HP-γ-CD. Cyclodextrins were tested following the Bacterial Endotoxin Test described in USP XXVI using Limulus Amebocyte Lysate (LAL). Osmolalities of all the solutions were measured by a Knauer Automatic semi-micro Osmometer and adjusted to the value of 300 mOsm/kg by the addition of an adequate amount of NaCl. A terminal sterilization of the solutions was performed by steam sterilization process.
  • Methods
  • Aerosol was produced by using an ultrasonic nebuliser SYSTAM, the vibration frequency of which is 2.4 MHz with variable vibration intensity and ventilation levels. Vibration intensity was fixed in position 6 and the ventilation level was 25 (t½) Umin.
  • Characterization of Nebulized Aerosol
  • Aerosol size distribution emitted from CDs solutions was determined with a laser size analyzer Mastersizer (Malvern, Orsay, France). Ten milliliters of each solution were directly nebulized in the laser beam. The mouth piece was held at 1 cm from the center of the laser beam. The resulting aerosol was aspirated on the opposite side of the beam. Environmental temperature and relative humidity were maintained constant, that is to say at 20° C. and 40-45%. Triplicates of each measurement were performed and compared to controls of PBS. The results are expressed as the percentage of droplets comprised in the range 0.5 to 5.79 μm and the median diameter. The concentration of droplets in the air evaluated by the obscuration percentage of the laser beam was in the same range for each experiment (15-25%). A comparison of the MMAD, the GSD and the percentage of droplets comprised in the range of 0.5 to 5.79 μM of all the CDs solutions with the corresponding values for PBS demonstrated that the presence of CDs in the solution did not influence the droplet size distribution in the aerosols. A fraction of droplets comprised in the range of 0.5 to 5.79 μm close to 65% was obtained in each experiment.
  • Sensitisation, Allergen Exposure and Therapeutic Protocols.
  • In order to study modulation of airway inflammation, males BALB/c mice of 6 to 8 weeks old were sensitized by intraperitoneal injection of 10 μg ovalbumin (OVA) (Sigma Aldrich, Schnelldorf, Germany) emulsified in aluminum hydroxyde (AlumInject; Perbio, Erembodegem, Belgium) on days 1 and 8. Mice were subsequently exposed to allergens by daily inhalation of an aerosol of OVA 1%, for 30 minutes, generated by ultrasonic nebulizer (Devilbiss 2000), from day 21 to 27. Mice were subjected to inhalation of α-CD, HP-β-CD 1, 7.5, 50 mM and HP-γ-CD 50 mM 30 minutes before OVA inhalation. Mice were sacrificed performed on day 28 as previously reported by Cataldo and al in Am. J. Pathol 2002; 161(2):491-498.
  • Bronchoalveolar Lavage Fluid (BAL)
  • Immediately after assessment of airway responsiveness, and 24 hours after the last allergen exposure.
  • Mices were sacrificed and a bronchoalveolar lavage was performed using 4×1 ml PBS-EDTA 0.05 mM (Calbiochem, Darmstadt, Germany) as previously described by Cataldo D D, Tournoy K G, Vermaelen K et al. in Am J Pathol 2002; 161(2):491-498. Cells were recovered by gentle manual aspiration. After centrifugation of bronchoalveolar fluid (BALF) (1200 rpm for 10 minutes, at 4° C.), the supernatant was frozen at −80° C. for protein assessment and the cell pellet was resuspended in 1 ml PBS-EDTA 0.05 mM. The differential cell counts were performed on cytocentrifuged preparations (Cytospin) after staining with Diff-Quick (Dade, Belgium).
  • Pulmonary histology and tissue processing After BAL, the thorax was opened and the left main bronchus was clamped. The left lung was excised and frozen immediately at −80° C. for protein and mRNA extraction. The right lung was infused with 4 ml paraformaldehyde 4%, embedded in paraffin and used for histology. Sections of 5 μm thickness were cut off from paraffin and were stained with haematoxylin-eosin. The extent of peribronchial inflammation was estimated by a score calculated by quantification of peribronchial inflammatory cells, as previously described by Cataldo D D, Tournoy K G, Vermaelen K et al. in Am J Pathol 2002; 161(2):491-498. A value of 0 was adjudged when no inflammation was detectable, a value of 1 when there was occasionally inflammatory cells, a value of 2 when most bronchi were surrounded by a thin layer (1 to 5 cells) of inflammatory cells and a value of 3 when most bronchi were surrounded by a thick layer (>5 cells) of inflammatory cells. Since 5-7 randomly selected tissue sections per mouse were scored, inflammation scores are expressed as a mean value and can be compared between groups. After Congo Red staining, the eosinophilic infiltration in the airway walls was quantified by manual count and reported to the perimeter of epithelial basement membrane defining an eosinophilic inflammatory score.
  • The left lung was crushed using a Mikro-Dismembrator (Braun Biotech International, Gmbh Melsungen, Germany). For proteins extraction, the crushed lung tissue was incubated overnight at 4° C. in a solution containing 2M urea, 1M NaCl and 50 mM Tris (pH 7.5) and subsequently centrifuged 15 minutes at 16.000×g. The supernatant was stored at −80° C.
  • Bronchial responsiveness measurement Twenty-four hours after the last allergen exposure, the bronchial hyper responsiveness was determined by measuring the Penh (Enhanced Pause) using a barometric plethysmograph (Emka technologies, Paris) as proposed by Hamelmann, E., et al., Am. J Respir. Crit. Care Med. 156 (1997) 766-775). The Penh was measured at baseline and 5 min after the inhalation of increasing doses (25, 50, 75 and 100 mM) of methacholine (Mch).
  • Measurements of Cytokines by ELISA
  • Eotaxin and IL-13 levels were assessed using commercial ELISAs (R&D systems, Abingdon, UK). Eotaxin was measured by incubation in a wheel coated with a primary antibody specifically dedicated to the recognition of the protein and after rinsing, a second antibody against eotaxin, coupled with horse radish peroxydase was used to quantify the amounts of eotaxin in the solution
  • Measurement of Allergen Specific Serum IgE
  • At the end of the experiment, blood was drawn from the heart for measurement of OVA specific serum IgE. Microtiter plates were coated with OVA. Serum was added followed by a biotinylated polyclonal rabbit anti-mouse IgE (S. Florquin, ULB, Brussels, Belgium). A serum pool from OVA-sensitized animals was used as internal laboratory standard; 1 unit was arbitrarily defined as 1/100 dilution of this pool.
  • Measurement of Eotaxin and IL-13 in Bal and Lung Protein Extracts.
  • IL-13 was measured by incubation in a wheel coated with a primary antibody specifically dedicated to the recognition of the protein and after rinsing, a second antibody against IL-13, coupled with horse radish peroxydase was used to quantify the amounts of eotaxin in the solution.
  • Statistical Analysis
  • Results of BAL cell count, pulmonary histology, cytokines and mRNA levels were expressed as mean+/−SEM and the comparison between the groups was performed using Mann-Whitney test. Mann-Whitney test was performed using GRAPHPAD INSTAT version 3.00 for WINDOWS 95 (GRAPHPAD SOFTWARE, San Diego, Calif., USA, WWW.GRAPHPAD.Com.). P values <0.05 were considered as significant.
  • Pharmacological Results:
  • Inflammatory cells in the BAL.
  • After allergen exposure, eosinophil counts were significantly decreased after the inhalation of HP-β-CD and HP-γ-CD at the dose of 50 mM. There was a dose dependent decrease in BAL eosinophils with the HP-β-CD 1, 7.5 and 50 mM inhalation. Other inflammatory cells were not present in different amounts in the BAL after HP-β-CD inhalation when compared to placebo. On the contrary, α-CD inhalation led to a tendency to increase the number of eosinophils in BAL after allergen exposure (FIG. 1).
  • Peribronchial Inflammation
  • After allergen exposure, mice treated with placebo displayed a significant increase in peribronchial inflammation as quantified by the peribronchial inflammation score. Mice treated with HP-β-CD 1, 7.5, and 50 mM and HP-γ-CD 50 mM were shown to have decreased inflammation score when compared to placebo treated mice. α-CD inhalation did not reduce the peribronchial inflammation score (FIG. 2).
  • Peribronchial Eosinophil Infiltration
  • As demonstrated previously, the allergen exposure did induce a significant increase in the number of eosinophils detectable in the peribronchial area. All CD tested induced a decrease of this infiltration and this decrease reached statistical significance for α-CD, HP-β-CD 1, 7.5, and HP-γ-CD 50 mM (FIG. 3).
  • Bronchial Responsiveness
  • The inhalation of HP-β-CD 50 mM reduced the methacholine-induced Penh increase (FIG. 4). When measuring the area under the methacholine dose-response curve (A.U.C) for different CDs, HP-β-CD 50 mM was the only to show a significant decrease (FIG. 5).
  • Cytokine Measurements in BAL and Lung Protein Extracts
  • When compared to placebo exposed mice, all doses of HP-β-CD tested induced a decrease in levels of eotaxin measured by ELISA in lung protein extracts (FIG. 7 a). IL-13 levels were decreased in BAL after HP-β-CD exposure and, on the contrary, were increased after α-CD exposure (FIG. 7 b).
  • Measurements of Allergen-Specific IgE in Serum
  • There were no significant differences between the groups for the allergen sensitization as assessed by the similar levels of OVA specific IgE measured by ELISA in serum (FIG. 6).
  • Example 2 Use of Compositions Comprising 2-O-Methyl-Cyclodextrin for Therapy of Allergen-Induced Airway Inflammation and Bronchial Hyperresponsiveness in a Mouse Model of Asthma Materials
  • Materials are identical to example 1 with the exception of the cyclodextrin compound which is here 2-O-methyl-cyclodextrin, KLEPTOSE CRYSMEB®, a product commercialised by Roquette. It has, on average, 4 methyl groups per native cyclodextrin molecule and is characterized by an average molecular weight of 1135 and a average molar degree of substitution of 0.57.
  • Sterile, apyrogenic and isotonic CD solutions were prepared with 10, 20, 50 and 75 mM for 2-O-methyl-cyclodextrin. Cyclodextrins were tested following the Bacterial Endotoxin Test described in USP XXVI using Limulus Amebocyte Lysate (LAL). Osmolalities of all the solutions were measured by a Knauer Automatic semi-micro Osmometer and adjusted to the value of 280-300 mOsm/kg by the addition of an adequate amount of NaCl. A terminal sterilization of the solutions was performed by steam sterilization process.
  • Methods
  • Same methods are used as in example 1 but in the present example we did expose mice to aerosolized CRYSMEB (10, 20, 50, 100 or 200 mM) in a standard exposure box (20×30×15 cm) for 30 min/day during 7 days.
  • Pharmacological Results: Inflammatory Cells in the BAL.
  • The cellular composition of the bronchoalveolar lavage was not significantly altered by the exposure to CRYSMEB. In particular, there were no differences regarding eosinophil and neutrophil counts (see table 1).
  • Bronchoalveolar lavage eosinophilia was significantly decreased in the groups treated by CRYSMEB. The decrease in lavage eosinophilia was comparable with that obtained with different concentrations of HP-beta-cyclodextrins or fluticasone, a commonly used inhalation steroid used as a reference therapy (table 2)
  • Peribronchial Inflammation
  • After allergen exposure, mice treated with placebo displayed a significant increase in peribronchial inflammation as quantified by the peribronchial inflammation score. Mice treated with CRYSMEB 20 mM were shown to have decreased inflammation score when compared to placebo treated mice (FIG. 9A). Peribronchial inflammation score was measured and was significantly decreased in every treatment group as compared to placebo (FIG. 9B)
  • Bronchial Responsiveness
  • The inhalation of CRYSMEB 10 mM reduced the methacholine-induced Penh increase (FIG. 10).
  • The responsiveness to methacholine was increased after allergen exposure and placebo and was significantly reduced by the treatment with CRYSMEB in an extent comparable to that obtained with fluticasone therapy (FIG. 10)
  • Cytokine Measurements in BAL and Lung Protein Extracts
  • In order to unveil mechanisms implicated in the pharmacological effect of CRYSMEB, we measured IL-13, a major Th2 cytokine implicated in the airway hyperresponsiveness and inflammation. We found that levels of IL-13 measured by ELISA in whole lung protein extracts were significantly decreased by the exposure to CRYSMEB as well as fluticasone and HP-beta-CD 50 mM. (see FIG. 11)
  • Example 3 Pharmaceutical Composition to be Administered in an Aerosol to a Patient in Need of Treatment for Bronchial Inflammatory Disease
  • HP betaCD 75 mM
    Solution osmolality is 308 mOs/kg. pH is 7.2.
  • The concentration of CD compound can be modified in function of the requirements. It is preferred to adjust the tonicity by addition of NaCl.
  • A preferred composition for nebulization is:
  • For 200 ml of solution:
      • HPβCD exempt from pyrogenic 20.15 g (75 mM)
      • Sodium chloride 1.42 g (isotonicity)
      • Pyrogen-free, sterile, purified water, q.s. ad 200 ml
      • a) Weigh 20.15 g of HPβCD exempt from pyrogenic (3.2% H2O, from ROQUETTE) and dissolve them in 100 ml of purified water.
      • b) Weigh 1.42 g of sodium chloride and add them to solution (a) by energetically stirring so as to dissolve them.
      • c) Complete with water so as to obtain 200 ml of solution.
      • Sterilize by filtration through a 0.22 μm polypropylene membrane.
  • TABLE 1
    differential cell counts in the bronchoalveolar lavage measured after the exposure to
    different concentrations of inhaled CRYSMEB.
    PLACEBO Crysmeb 20 mM Crysmeb 50 mM Crysmeb 75 mM
    Epithelial cells (%) 15.9714 ± 5.154   29.9 ± 5.909 36.1375 ± 4.52   30.8875 ± 1.349 
    Eosinophils (%) 0.0428 ± 0.0428 0.0375 ± 0.0375 0.1125 ± 0.0789 0.0375 ± 0.0375
    Neutrophils (%) 0.1285 ± 0.236  0.0375 ± 0.1061   0.2 ± 0.3505 0.0375 ± 0.1061
    Lymphocytes (%) 0.1857 ± 0.1421  0.425 ± 0.1485  0.275 ± 0.1161  0.075 ± 0.0491
    Macrophages (%) 83.5857 ± 1.179°    69.5 ± 5.956 63.1625 ± 4.695   68.9 ± 1.334
  • TABLE 2
    differential cell counts in the bronchoalveolar lavage measured after the exposure to
    different concentrations of inhaled CRYSMEB.
    HPBeta-CD HPBeta-CD HPBeta-CD CRYSMEB
    PLACEBO Fluticasone 1 mM 10 mM 50 mM 10 mM
    Epithelial 3.86 ± 2.551 22.85 ± 5.343  25.08 ± 3.413  34.44 ± 3.723 45.04 ± 5.534  36.83 ± 5.644 
    cells (%)
    Eosinophils (%) 53.08 ± 4.683  32.92 ± 7.306* 34.2 ± 7.705* 27.24 ± 4.98* 12.18 ± 4.366* 8.84 ± 2.946*
    Neutrophils (%) 3.03 ± 1.333 1.85 ± 1.093 0.36 ± 0.1563  0.83 ± 0.5838  0.72 ± 0.3992 1.26 ± 0.4587
    Lymphocytes (%) 3.62 ± 1.576  1.68 ± 0.7115 0.48 ± 0.1869   0.21 ± 0.08571 0.12 ± 0.12  0.214 ± 0.1079 
    Macrophages (%) 36.25 ± 5.016  40.52 ± 3.122  39.75 ± 5.427  37.114 ± 3.878  41.86 ± 9.043  52.714 ± 6.49  
    Total cells 220.42 ± 81.709  75.92 ± 11.922 74.92 ± 14.396  114.43 ± 33.245 37.33 ± 10.683 131.93 ± 33.637 
    (104/ml)
  • Example 4 Used Inhaled Hydroxypropyl-β-Cyclodextrin to Address Allergen- or LPS-Induced Inflammation in Rodents Experimental Protocol:
  • In ovalbumin (OVA)-induced inflammation model, mice were immunized by intraperitoneal injection of OVA (10 μg) (Sigma Aldrich, Schnelldorf, Germany) and aluminium hydroxyde on days 0 and 7. From days 21 to 25, mice were exposed to inhalation of 1% OVA or PBS (phosphate buffer saline) for 30 minutes. Airway hyperresponsiveness was measured on day 26 before sacrifice. Mice were either exposed to aerosolized hydroxypropyl-beta-cyclodextrin (15 mM) or PBS for 30 min 6 hours before allergen exposure.
  • After sacrifice, thorax was opened and the right lungs were excised and snap frozen in liquid nitrogen for protein extraction. The left lung was insufflated at constant pressure with 4% paraformaldehyde and embedded in paraffin for further histological analysis. A peribronchial inflammation score was applied on each hematoxylin-eosin stained slide as previously reported (Cataldo et al Am J Pathol 2002). A value from 0 to 2 was adjudged to each bronchus. Score of 0 corresponded to bronchi without inflammation; score 1 to occasional mononuclear cells observed around bronchi, and score 2 to 1 to 5 layer(s) of inflammatory cells around bronchi. Six bronchi per mice were counted and statistical analysis was performed by using GraphPad Program. Lung tissues were crushed and total protein extracts were prepared by incubating crushed lung tissues in a 2M urea solution. Tissue lysates were centrifuged for 15 min at 16100×g. ELISA for CXCL-1 on lung protein extracts were assessed using antibodies from R&D Systems and the R&D Duoset® Elisa Development kit (R&D Systems, Minneapolis, Minn., USA).
  • Results:
  • We already reported that overall inflammation was lower as well as bronchial responsiveness to methacholine. As allergens are able to generate neutrophilic inflammation by activation of chemotactic factors, we addressed the potential efficacy of these compounds to decrease levels of neutrophil chemoattractants in mouse lungs after allergen exposure. Levels of CXCL-1, a major neutrophil chemoattractant were significantly decreased in lung protein extracts of cyclodextrins-treated mice (HPb in FIG. 12) as compared to placebo-exposed mice (OVA in FIG. 12). In summary, there is a decreased production of chemotactic agents for neutrophils after exposure to cyclodextrins in an allergen-induced inflammation model in mice.
  • Example 5 Used Inhaled Hydroxypropyl-β-Cyclodextrin to Address Allergen- or LPS-Induced Inflammation in Rodents
  • Experimental Protocol:
  • In lipopolysaccharide (LPS)-induced inflammation model, mice were exposed to 3 μg LPS diluted in 100 μl of PBS or PBS (phosphate buffer saline) only administered by direct tracheal instillations at day 1 and 5 of the protocol. Mice were daily exposed either to aerosolized hydroxypropyl-beta-cyclodextrin (15 mM) or PBS for 30 min from day 0 to 5 (6 hours after LPS or sham exposure at days 1 and 5). Mice were sacrificed at day 6.
  • After sacrifice, thorax was opened and the right lungs were excised and snap frozen in liquid nitrogen for protein extraction. The left lung was insufflated at constant pressure with 4% paraformaldehyde and embedded in paraffin for further histological analysis. A peribronchial inflammation score was applied on each hematoxylin-eosin stained slide as previously reported (Cataldo et al Am J Pathol 2002). A value from 0 to 2 was adjudged to each bronchus. Score of 0 corresponded to bronchi without inflammation; score 1 to occasional mononuclear cells observed around bronchi, and score 2 to 1 to 5 layer(s) of inflammatory cells around bronchi. Six bronchi per mice were counted and statistical analysis was performed by using GraphPad Program.
  • Lung tissues were crushed and total protein extracts were prepared by incubating crushed lung tissues in a 2M urea solution. Tissue lysates were centrifuged for 15 min at 16100×g. ELISA for CXCL-1 on lung protein extracts were assessed using antibodies from R&D Systems and the R&D Duoset® Elisa Development kit (R&D Systems, Minneapolis, Minn., USA).
  • Results:
  • Levels of CXCL-1 (a chemotactic agent for neutrophils) were significantly decreased after treatment of animals inhaled with hydroxypropyl-β-cyclodextrin as compared to placebo-treated mice (FIG. 13). Peribronchial inflammation was significantly lower in the hydroxypropyl-β-cyclodextrin-treated group as compared to placebo-exposed animals (FIG. 14).
  • CONCLUSION
  • Overall, these data demonstrate that hydroxypropyl-β-cyclodextrin is able to decrease the production of CXCL-1, a potent chemotactic agent for neutrophils in various inflammatory conditions and LPS-induced peribronchial inflammation suggesting a usefulness of this compound for treatment of COPD.

Claims (2)

1-3. (canceled)
4. A method for the treatment of COPD (chronic obstruction pulmonary disease) in a host mammal in need of such treatment, comprising the step of administering in an aerosol, a pharmaceutical composition consisting essentially of hydroxypropyl beta-cyclodextrin in a concentration of from 10-75 mM.
US14/699,563 2004-10-10 2015-04-29 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases Abandoned US20150258133A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/699,563 US20150258133A1 (en) 2004-10-10 2015-04-29 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
EP04104957.8 2004-10-10
EP04104957A EP1655034A1 (en) 2004-10-10 2004-10-10 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases.
PCT/EP2005/054966 WO2006037769A1 (en) 2004-10-10 2005-09-30 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases.
US66499907A 2007-04-10 2007-04-10
US12/846,241 US20110028432A1 (en) 2004-10-10 2010-07-29 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases
US13/478,743 US20120295872A1 (en) 2004-10-10 2012-05-23 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases
US13/804,626 US9034846B2 (en) 2004-10-10 2013-03-14 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases
US14/699,563 US20150258133A1 (en) 2004-10-10 2015-04-29 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/804,626 Continuation US9034846B2 (en) 2004-10-10 2013-03-14 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases

Publications (1)

Publication Number Publication Date
US20150258133A1 true US20150258133A1 (en) 2015-09-17

Family

ID=48870741

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/804,626 Expired - Fee Related US9034846B2 (en) 2004-10-10 2013-03-14 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases
US14/699,563 Abandoned US20150258133A1 (en) 2004-10-10 2015-04-29 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/804,626 Expired - Fee Related US9034846B2 (en) 2004-10-10 2013-03-14 Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases

Country Status (1)

Country Link
US (2) US9034846B2 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR3014694B1 (en) 2013-12-13 2016-11-11 Roquette Freres METHYL-CYCLODEXTRIN-BASED COMPOSITIONS FOR THE TREATMENT AND / OR PREVENTION OF DISEASES BY INCREASING THE CHOLESTEROL-HDL RATE
EP2923702A1 (en) 2014-03-28 2015-09-30 Universite De Liege Composition of cyclodextrin with budesonide derivatives for treatment and prevention of pulmonary inflammatory disease
CN106139161A (en) * 2016-08-12 2016-11-23 合肥久诺医药科技有限公司 A kind of roflumilast clathrate and solid preparation thereof
AU2018341235A1 (en) 2017-09-28 2020-05-07 Asdera Llc Use of cyclodextrins in diseases and disorders involving phospholipid dysregulation

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4054736A (en) 1970-06-10 1977-10-18 Ono Pharmaceutical Co., Ltd. Clathrate compounds of prostaglandins or their analogues with cyclodextrin
US4020160A (en) * 1975-08-15 1977-04-26 American Cyanamid Company Cyclodextrin sulfate salts as complement inhibitors
US5840713A (en) 1995-04-03 1998-11-24 Weisz; Paul B. Therapy for tissue membrane insufficiency
ATE499109T1 (en) * 2004-12-07 2011-03-15 Proteolix Inc PROTEAsome INHIBITION COMPOSITION

Also Published As

Publication number Publication date
US20130196947A1 (en) 2013-08-01
US9034846B2 (en) 2015-05-19

Similar Documents

Publication Publication Date Title
US7829550B2 (en) Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases
JP3480736B2 (en) Use of mometasone furoate for the treatment of airway channels and lung disease
US6677323B2 (en) Use of mometasone furoate for treating airway passage and lung diseases
US20150258133A1 (en) Use of cyclodextrin for treatment and prevention of bronchial inflammatory diseases
AU2004311478A1 (en) Inhalant formulation containing sulfoalkyl ether cyclodextrin and corticosteroid
US9023815B2 (en) Method for treating rhinitis and sinusitis by rhamnolipids
JP2022520990A (en) How to treat respiratory illness
Doggrell Triamcinolone: new and old indications
US20070225253A1 (en) Use of a trioxopyrimidine for the treatment and prevention of bronchial inflammatory diseases

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITE DE LIEGE, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CATALDO, DIDIER;EVRARD, BRIGITTE;NOEL, AGNES;AND OTHERS;SIGNING DATES FROM 20150608 TO 20150819;REEL/FRAME:036426/0833

STCB Information on status: application discontinuation

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