US20200171009A1 - Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist - Google Patents

Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist Download PDF

Info

Publication number
US20200171009A1
US20200171009A1 US16/781,587 US202016781587A US2020171009A1 US 20200171009 A1 US20200171009 A1 US 20200171009A1 US 202016781587 A US202016781587 A US 202016781587A US 2020171009 A1 US2020171009 A1 US 2020171009A1
Authority
US
United States
Prior art keywords
compound
product
dose
oxy
dry powder
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
US16/781,587
Inventor
Darrell Baker
Mark Bruce
Glenn Crater
Brian Noga
Marian Thomas
Patrick Wire
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.)
Glaxo Group Ltd
Original Assignee
Glaxo Group Ltd
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=41573013&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20200171009(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Glaxo Group Ltd filed Critical Glaxo Group Ltd
Priority to US16/781,587 priority Critical patent/US20200171009A1/en
Publication of US20200171009A1 publication Critical patent/US20200171009A1/en
Priority to US17/514,685 priority patent/US20220047565A1/en
Priority to US17/714,241 priority patent/US20220235087A1/en
Priority to US18/485,799 priority patent/US20240041846A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • 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
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K9/0075Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a dry powder inhaler [DPI], e.g. comprising micronized drug mixed with lactose carrier particles
    • 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
    • A61P11/02Nasal agents, e.g. decongestants
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B65CONVEYING; PACKING; STORING; HANDLING THIN OR FILAMENTARY MATERIAL
    • B65DCONTAINERS FOR STORAGE OR TRANSPORT OF ARTICLES OR MATERIALS, e.g. BAGS, BARRELS, BOTTLES, BOXES, CANS, CARTONS, CRATES, DRUMS, JARS, TANKS, HOPPERS, FORWARDING CONTAINERS; ACCESSORIES, CLOSURES, OR FITTINGS THEREFOR; PACKAGING ELEMENTS; PACKAGES
    • B65D75/00Packages comprising articles or materials partially or wholly enclosed in strips, sheets, blanks, tubes, or webs of flexible sheet material, e.g. in folded wrappers
    • B65D75/28Articles or materials wholly enclosed in composite wrappers, i.e. wrappers formed by associating or interconnecting two or more sheets or blanks
    • B65D75/30Articles or materials enclosed between two opposed sheets or blanks having their margins united, e.g. by pressure-sensitive adhesive, crimping, heat-sealing, or welding
    • B65D75/32Articles or materials enclosed between two opposed sheets or blanks having their margins united, e.g. by pressure-sensitive adhesive, crimping, heat-sealing, or welding one or both sheets or blanks being recessed to accommodate contents
    • B65D75/36Articles or materials enclosed between two opposed sheets or blanks having their margins united, e.g. by pressure-sensitive adhesive, crimping, heat-sealing, or welding one or both sheets or blanks being recessed to accommodate contents one sheet or blank being recessed and the other formed of relatively stiff flat sheet material, e.g. blister packages, the recess or recesses being preformed
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/20Carbocyclic rings
    • C07H15/203Monocyclic carbocyclic rings other than cyclohexane rings; Bicyclic carbocyclic ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/26Acyclic or carbocyclic radicals, substituted by hetero rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates to pharmaceutical products and compositions for use in the treatment of chronic obstructive pulmonary disease (COPD), asthma and related diseases.
  • COPD chronic obstructive pulmonary disease
  • this invention relates to the combination of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist, and the use of said combination in treating diseases mediated via the M 3 muscarinic acetylcholine receptor and/or the beta-2 adrenoreceptor.
  • this invention is concerned with novel pharmaceutical combination products comprising 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate and 4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide and the use of said combination products in medicine, particularly in treating diseases mediated via the M 3 muscarinic acetylcholine receptor and/or the beta-2 adrenoreceptor, for example in the prophylaxis and treatment of inflammatory or respiratory tract diseases.
  • COPD chronic obstructive pulmonary diseases
  • rhinitis respiratory tract infection and upper respiratory tract disease
  • beta-2 adrenergic receptor agonists beta-2 adrenergic receptor agonists
  • beta-2 agonists beta-2 adrenergic receptor agonists
  • short acting compounds for immediate relief such as salbutamol, biltolterol, pirbuterol and terbutaline.
  • longer acting compounds such as salmeterol and formoterol.
  • Salmeterol is available by prescription for use twice daily in the treatment of asthma.
  • inhaled anticholinergic agents have become well established as well-tolerated and effective bronchodilators for the treatment of COPD.
  • Treatment with anticholinergics significantly improves FEV 1 , (forced expiratory volume in 1 second) resting and dynamic lung hyperinflation, symptoms and exercise capacity, and reduces COPD exacerbations.
  • FEV 1 forced expiratory volume in 1 second
  • ipratroplum bromide ipratropium; dosed four-times-a-day
  • oxitropium bromide oxitropium bromide
  • tiotropium tiotropium; dosed once-daily.
  • WO 03/024439 describes compounds of the general formula:
  • WO2005/104745 specifically describes the compound 4-[hydroxy(diphenyl)methyl]-1 ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide.
  • the present invention provides a novel pharmaceutical combination product comprising the therapeutic agents:
  • Compound (II) may refer to the free base depicted above, and/or one or more salts thereof, as dictated by the context.
  • the pharmaceutical combination product comprises 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate and 4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide.
  • 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate and 4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide are the sole active ingredients in said pharmaceutical combination product.
  • the pharmaceutical combination product of Compound (I) and Compound (II) additionally comprises an inhaled corticosteroid.
  • This invention also provides for use of the pharmaceutical combination product in the manufacture of a medicament for the treatment of conditions for which administration of one or more of the therapeutic compounds is indicated.
  • the use is for the manufacture of a medicament for the treatment of inflammatory or respiratory tract diseases, by simultaneous or sequential administration of Compound (I) and Compound (II).
  • the use is for the manufacture of a medicament for the treatment of chronic obstructive pulmonary disease (COPD) and/or asthma, by simultaneous or sequential administration of Compound (I) and Compound (II).
  • COPD chronic obstructive pulmonary disease
  • the invention also provides said pharmaceutical combination product for use in the treatment of inflammatory or respiratory tract diseases, such as chronic obstructive pulmonary disease (COPD) and/or asthma.
  • COPD chronic obstructive pulmonary disease
  • Another embodiment of the invention is a method for the treatment of inflammatory or respiratory tract diseases, comprising administering either sequentially or simultaneously, to a patient in need thereof, a pharmaceutical combination product comprising Compound (I) and Compound (II).
  • the inflammatory or respiratory tract disease is selected from the group consisting of chronic obstructive pulmonary disease, chronic bronchitis, asthma, chronic respiratory obstruction, pulmonary fibrosis, pulmonary emphysema, allergic rhinitis, small airways disease, bronchiectasis and cystic fibrosis.
  • the pharmaceutical combination product may be used for the treatment of inflammatory or respiratory tract diseases, and more specifically the treatment of chronic obstructive pulmonary disease (COPD) and/or asthma by simultaneous or sequential administration of Compound (I) and Compound (II).
  • COPD chronic obstructive pulmonary disease
  • the present invention is directed to a pharmaceutical combination product comprising
  • the pharmaceutically acceptable anion depicted by X ⁇ may be selected from chloride, bromide, iodide, hydroxide, sulfate, nitrate, phosphate, acetate, trifluoroacetate, fumarate, citrate, tartrate, oxalate, succinate, mandelate, methanesulfonate or p-toluenesulfonate.
  • the pharmaceutically acceptable anion X ⁇ is bromide.
  • the structural formula for the quaternary moiety (cation) of Compound (1) is also referred to as 4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane.
  • Compound (I) is 4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide (also referred to herein as Compound (I) bromide).
  • Pharmaceutically acceptable acid addition salts of Compound (II) include those formed from hydrochloric, hydrobromic, sulphuric, citric, tartaric, phosphoric, lactic, pyruvic, acetic, trifluoroacetic, triphenylacetic, phenylacetic, substituted phenyl acetic eg.
  • the pharmaceutically acceptable salt of Compound (II) is selected from the acetate, 1-naphthoate and (R)-mandelate salts.
  • the pharmaceutically acceptable salt of Compound (II) is the ⁇ -phenylcinnamate salt.
  • the pharmaceutically acceptable salt of Compound (II) is the triphenylacetate salt.
  • Compound (II) is 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate (also referred to as Compound (II) triphenylacetate).
  • the pharmaceutical combination product of the invention comprises 4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide and 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate.
  • the pharmaceutical combination product of Compound (I) and Compound (II) additionally comprises an inhaled corticosteroid, e.g. fluticasone propionate, mometasone furoate, budesonide or 6 ⁇ ,9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester (fluticasone furoate).
  • corticosteroid e.g. fluticasone propionate, mometasone furoate, budesonide or 6 ⁇ ,9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester (fluticasone furoate).
  • said pharmaceutical combination product comprises 4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide, 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate and 6 ⁇ ,9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester (fluticasone furoate).
  • the pharmaceutical combination product of the invention comprises 4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide and 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate as the sole active ingredients.
  • Compound (I) specifically 4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide has been the subject of studies in animal models, and in humans, and has been found to be a long acting high-affinity pan-active muscarinic receptor antagonist which has potential for once-daily administration.
  • Compound (II) specifically 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol and its salts has been extensively tested in animal and human studies and has been found to demonstrate sustained bronchodilation over a 24 hour period in conjunction with a favourable safety profile and thus has the potential for once-daily administration.
  • Compound (I) and Compound (II), and the combination thereof, are considered to have potential in the treatment of inflammatory or respiratory tract diseases such as chronic obstructive pulmonary disease, chronic bronchitis, asthma, chronic respiratory obstruction, pulmonary fibrosis, pulmonary emphysema, allergic rhinitis, small airways disease, bronchiectasis and cystic fibrosis.
  • inflammatory or respiratory tract diseases such as chronic obstructive pulmonary disease, chronic bronchitis, asthma, chronic respiratory obstruction, pulmonary fibrosis, pulmonary emphysema, allergic rhinitis, small airways disease, bronchiectasis and cystic fibrosis.
  • COPD chronic disease characterised by airways obstruction and reduced maximum expiratory flow from the lungs that manifests as persistent daily symptoms, such as shortness of breath (dyspnoea), and limitation of the ability to perform daily activities or exertion. Furthermore, there are periodic exacerbations of the condition that result in worsening of the day-to-day symptoms and activity limitation, and can also lead to hospitalisation of the patient because of the severity of the worsening symptoms/limitation. In addition, there is a progressive decline in lung function (disease progression) over several years.
  • Bronchodilator treatment in COPD includes but is not necessarily limited to reducing symptoms, particularly dyspnoea, to allow a patient to undertake more daily activities and other activities that require exertion, and preventing exacerbations.
  • Asthma is a chronic condition, which is characterised by widespread, variable and reversible airflow obstruction. Symptoms include coughing, wheezing, breathlessness and/or a tight feeling in the chest. Asthma attacks are generally caused by exposure to a trigger, such as pollen, dust or other allergens, which causes constriction of the airways (bronchoconstriction). It will be appreciated that a subject suffering from a condition such as asthma, may variously from time to time display no overt symptoms of the condition, or may suffer from periodic attacks during which symptoms are displayed or may experience exacerbations or worsening of the condition. In this context the term ‘treatment’ is intended to encompass prevention of such periodic attacks or exacerbations of the existing condition. Such treatment may be referred to as ‘maintenance treatment’ or ‘maintenance therapy’.
  • the route of administration is by inhalation via the mouth or nose.
  • the route of administration is by inhalation via the mouth.
  • Compound (I), and specifically (4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide may be administered by inhalation at a dose of from about 1 mcg to about 1000 mcg/daily, e.g. 100, 250 or 500 mcg per day.
  • Compound (I) and specifically (4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide may be administered by inhalation at a dose of 62.5 mcg or 125 mcg per day. In general Compound (I) will be administered as a once-daily dose.
  • Compound (I), and specifically (4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide may be administered by inhalation, once-daily, at a dose of 62.5 mcg per day.
  • Compound (I), and specifically (4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide may be administered by inhalation, once-daily, at a dose of 125 mcg per day.
  • Compound (II) may for example be administered by inhalation at a dose of from about 1 mcg to about 400 mcg/day (calculated as the free base).
  • Compound (II), and specifically 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate may be administered by inhalation at a dose of from about 1 mcg to 100 mcg/day, for example 3, 6.25, 12.5, 25, 50 or 100 mcg/day (calculated as the free base).
  • Compound (II) will be administered as a once-daily dose. In one embodiment Compound (II) may be administered by inhalation at a dose of 12.5 mcg/day. In another embodiment Compound (II) may be administered by inhalation at a dose of 25 mcg/day. In another embodiment Compound (II) may be administered by inhalation at a dose of 50 mcg/day.
  • 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate may be administered by inhalation, once-daily, at a dose of 25 mcg per day.
  • the present invention provides a pharmaceutical combination product for once-daily administration by inhalation, comprising 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate at a dose of 25 mcg per day, and (4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide at a dose of 125 mcg per day.
  • the present invention provides a pharmaceutical combination product for once-daily administration by inhalation, comprising 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate at a dose of 25 mcg per day, and (4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide at a dose of 62.5 mcg per day.
  • the combination additionally includes an inhaled corticosteroid
  • this may be used at doses compatible with those known for monotherapy.
  • the inhaled corticosteroid is fluticasone furoate this may be administered by inhalation at a dose of from about 25 mcg to about 800 mcg daily, and if necessary in divided doses.
  • the daily dose of fluticasone furoate may be for example 25, 50, 100, 200, 300, 400, 600 or 800 mcg, in general as a once-daily dose.
  • the daily dose of fluticasone furoate is 100 mcg.
  • the daily dose of fluticasone furoate is 50 mcg.
  • the individual compounds of the pharmaceutical combination product as described herein may be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations/compositions.
  • Compound (I) and Compound (II) may for example, be formulated separately and presented in separate packs or devices, or said individually formulated components may be presented in a single pack or device.
  • the individual compounds may be admixed within the same formulation, and presented as a fixed pharmaceutical combination.
  • such formulations will include pharmaceutical carriers or excipients as described hereinafter, but combinations of the compounds without any excipients are also within the ambit of this invention.
  • the individual compounds of the pharmaceutical combination product may be administered simultaneously in a combined pharmaceutical formulation or composition.
  • the pharmaceutical combination product additionally includes an inhaled corticosteroid, eg 6 ⁇ ,9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester (fluticasone furoate)
  • an inhaled corticosteroid eg 6 ⁇ ,9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester (fluticasone furoate)
  • this may likewise be formulated separately, either with or without one or more pharmaceutical carriers or excipients, and presented for either sequential or simultaneous administration, or the inhaled corticosteroid may be admixed with either Compound (I) and/or Compound (II).
  • a pharmaceutical combination product comprising Compound (I) and Compound (II) presented separately for sequential or simultaneous administration;
  • each of Compound (I) and/or Compound (II) may be formulated with or without pharmaceutical carriers or excipients.
  • the present invention further provides a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein at least one of Compound (I) and Compound (II) is formulated with a pharmaceutically acceptable carrier or excipient.
  • the present invention further provides a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein each of Compound (I) and Compound (II) is formulated with a pharmaceutically acceptable carrier or excipient.
  • compositions of Compounds (I) and (II) include those suitable for inhalation, including fine particle powders, or mists which may be generated and administered by means of various types of inhalers for example, reservoir dry powder inhalers, unit-dose dry powder inhalers, pre-metered multi-dose dry powder inhalers, nasal inhalers or pressurized metered dose inhalers, nebulisers or insufflators.
  • inhalers for example, reservoir dry powder inhalers, unit-dose dry powder inhalers, pre-metered multi-dose dry powder inhalers, nasal inhalers or pressurized metered dose inhalers, nebulisers or insufflators.
  • compositions may be prepared by any of the methods well known in the art of pharmacy. In general, said methods include the step of bringing the active ingredient(s) into association with the carrier which constitutes one or more accessory ingredients. In general the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired composition.
  • Powder compositions generally contain a powder mix for inhalation of the active ingredient and a suitable powder base (carrier/diluent/excipient substance) such as mono-, di or poly-saccharides (e.g. lactose or starch).
  • a suitable powder base such as mono-, di or poly-saccharides (e.g. lactose or starch).
  • lactose is preferred.
  • the lactose may be for example anhydrous lactose or ⁇ -lactose monohydrate.
  • the carrier is ⁇ -lactose monohydrate.
  • Dry powder compositions may also include, in addition to the active ingredient and carrier, a further excipient (eg a ternary agent) such as a sugar ester, calcium stearate or magnesium stearate.
  • a further excipient eg a ternary agent
  • the active ingredient may be presented without excipients.
  • composition or ‘formulation’ herein refers to the active ingredients either with or without excipients or carriers.
  • the present invention further provides a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein at least one of Compound (I) and Compound (II) is formulated with a pharmaceutically acceptable carrier and a ternary agent.
  • the present invention further provides a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein Compound (II) is formulated with a pharmaceutically acceptable carrier and a ternary agent.
  • the present invention further provides a pharmaceutical formulation comprising a combination of Compound (I) and Compound (II) wherein both Compounds are formulated with a pharmaceutically acceptable carrier and a ternary agent.
  • the present invention further provides a pharmaceutical combination product for inhaled administration comprising 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate and (4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide each formulated separately with a pharmaceutically acceptable carrier and a ternary agent, but held in the same pack or device, for sequential or simultaneous administration.
  • said ternary agent is magnesium stearate.
  • the present invention further provides a pharmaceutical combination product for inhaled administration comprising 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate and (4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide each formulated separately with lactose, as a pharmaceutically acceptable carrier, and magnesium stearate, as a ternary agent, but held in the same pack or device, for sequential or simultaneous administration.
  • a pharmaceutical combination product for inhaled administration comprising 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate
  • compositions may be presented in unit dosage form.
  • Dry powder compositions for topical delivery to the lung by inhalation may, for example, be presented in capsules and cartridges of for example gelatine, or blisters of for example laminated aluminium foil, for use in an inhaler or insufflator.
  • Each capsule, cartridge or blister may generally contain between 1 mcg-1000 mcg, e.g. 100 to 500 mcg of Compound (I) and/or between 1 mcg-400 mcg, e.g 1 to 100 mcg of Compound (II).
  • Packaging of the formulation may be suitable for unit dose or multi-dose delivery.
  • Compound (I) and Compound (II) may be formulated independently or in admixture. Said compounds may thus be incorporated in separate unit doses or may be combined in a single unit dose with or without additional excipients as deemed necessary.
  • each capsule, cartridge or blister may contain 125 mcg or 62.5 mcg of Compound (I) and/or 25 mcg of Compound (II).
  • each capsule, cartridge or blister may contain 125 mcg or 62.5 mcg of (4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide and/or 25 mcg of 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate.
  • a composition suitable for inhaled administration may be incorporated into a plurality of sealed dose containers provided on medicament pack(s) mounted inside a suitable inhalation device.
  • the containers may be rupturable, peelable or otherwise openable one-at-a-time and the doses of the dry powder composition administered by inhalation on a mouthpiece of the inhalation device, as known in the art.
  • the medicament pack may take a number of different forms, for instance a disk-shape or an elongate strip.
  • Representative inhalation devices are the DISKHALERTM and DISKUSTM devices, marketed by GlaxoSmithKline. The DISKUSTM inhalation device is, for example, described in GB 2242134A.
  • a dry powder inhalable composition may also be provided as a bulk reservoir in an inhalation device, the device then being provided with a metering mechanism for metering a dose of the composition from the reservoir to an inhalation channel where the metered dose is able to be inhaled by a patient inhaling at a mouthpiece of the device.
  • exemplary marketed devices of this type are TURBUHALERTM of AstraZeneca, TWISTHALERTM of Schering and CLICKHALERTM of Innovata.
  • a further delivery method for a dry powder inhalable composition is for metered doses of the composition to be provided in capsules (one dose per capsule) which are then loaded into an inhalation device, typically by the patient on demand.
  • the device has means to rupture, pierce or otherwise open the capsule so that the dose is able to be entrained into the patient's lung when they inhale at the device mouthpiece.
  • ROTAHALERTM of GlaxoSmithKline
  • HANDIHALERTM of Boehringer Ingelheim.
  • a dry powder composition may also be presented in a delivery device which permits separate containment of Compound (I) and Compound (II) optionally in admixture with one or more excipients.
  • a delivery device permitting separate containment of actives is an inhaler device having two medicament packs in peelable blister strip form, each pack containing pre-metered doses in blister pockets arranged along its length.
  • Said device has an internal indexing mechanism which, each time the device is actuated, peels opens a pocket of each strip and positions the packs so that each newly exposed dose of each pack is adjacent a manifold which communicates with a mouthpiece of the device. When the patient inhales at the mouthpiece, each dose is simultaneously drawn out of its associated pocket into the manifold and entrained via the mouthpiece into the patient's respiratory tract. Thus, each time the device is used, the patient is administered a combination therapy consisting of a dose from each medicament pack.
  • a further device that permits separate containment of different compounds is DUOHALERTM of Innovata.
  • the present invention provides a dry powder inhaler (Inhaler 1) comprising two compositions presented separately, wherein a first composition comprises
  • the present invention provides Inhaler 1 wherein each composition is in unit dose form.
  • the present invention provides Inhaler 1 wherein the unit dose form is a capsule, cartridge or blister.
  • the present invention provides Inhaler 1 wherein 4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide is present in an amount of about 125 mcg/dose.
  • the present invention provides Inhaler 1 wherein 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate is present in an amount of about 25 mcg/dose.
  • the present invention provides Inhaler 1 wherein the second composition further comprises 6 ⁇ ,9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester (fluticasone furoate).
  • the present invention provides Inhaler 1 wherein 6 ⁇ ,9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester (fluticasone furoate) is present in an amount of about 100 mcg/dose.
  • Spray compositions for inhalation may for example be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurised packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant.
  • Aerosol compositions suitable for inhalation can be either a suspension or a solution and generally contain the pharmaceutical product and a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane or a mixture thereof.
  • the aerosol composition may optionally contain additional formulation excipients well known in the art such as surfactants e.g. oleic acid, lecithin or an oligolactic acid derivative e.g. as described in WO94/21229 and WO98/34596 and/or cosolvents e.g. ethanol.
  • additional formulation excipients well known in the art such as surfactants e.g. oleic acid, lecithin or an oligolactic acid derivative e.g. as described in WO94/21229 and WO98/34596 and/or cosolvents e.g. ethanol.
  • Pressurised formulations will generally be retained in a canister (e.g. an aluminium canister) closed with a valve (e.g. a metering valve) and fitted into an actuator provided with a mouthpiece.
  • a pharmaceutical combination product comprising Compound (I) and Compound (II) formulated individually or in admixture, with a fluorocarbon or hydrogen-containing chlorofluorocarbon as propellant, optionally in combination with a surface-active agent and/or a co-solvent.
  • the propellant is selected from 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane and mixtures thereof.
  • Another aspect of the invention is a pharmaceutical combination product consisting of Compound (I) and Compound (II) formulated individually or in admixture, with a fluorocarbon or hydrogen-containing chlorofluorocarbon as propellant, optionally in combination with a surface-active agent and/or a cosolvent.
  • the propellant is selected from 1,1,1,2-tetrafluoroethane, or 1,1,1,2,3,3,3-heptafluoro-n-propane and mixtures thereof.
  • compositions according to the invention may be buffered by the addition of suitable buffering agents.
  • Active ingredients for administration by inhalation desirably have a controlled particle size.
  • the optimum particle size for inhalation into the bronchial system is usually 1- ⁇ m, preferably 2-5 ⁇ m. Particles having a size above 20 ⁇ m are generally too large when inhaled to reach the small airways.
  • the particles of the active ingredient as produced may be size reduced by conventional means e.g. by micronization.
  • the desired fraction may be separated out by air classification or sieving.
  • the particles will be crystalline.
  • Dry powder compositions according to the invention may comprise a carrier.
  • the carrier when it is lactose e.g. ⁇ -lactose monohydrate, may form from about 91 to about 99%, e.g. 97.7-99.0% or 91.0-99.2% by weight of the formulation.
  • the particle size of the carrier for example lactose, will be much greater than the inhaled medicament within the present invention.
  • the carrier when it is lactose it will typically be present as milled lactose, having a MMD (mass median diameter) of 60-90 ⁇ m.
  • the lactose component may comprise a fine lactose fraction.
  • the ‘fine’ lactose fraction is defined as the fraction of lactose having a particle size of less than 7 ⁇ m, such as less than 6 ⁇ m, for example less than 5 ⁇ m.
  • the particle size of the ‘fine’ lactose fraction may be less than 4.5 ⁇ m.
  • the fine lactose fraction if present, may comprise 2 to 10% by weight of the total lactose component, such as 3 to 6% by weight fine lactose, for example 4.5% by weight fine lactose.
  • Magnesium stearate if present in the composition, is generally used in an amount of about 0.2 to 2%, e.g. 0.6 to 2% or 0.5 to 1.75%, e.g. 0.6%, 0.75%, 1%, 1.25% or 1.5% w/w, based on the total weight of the composition.
  • the magnesium stearate will typically have a particle size in the range 1 to 50 ⁇ m, and more particularly 1-20 ⁇ m, e.g. 1-10 ⁇ m.
  • Commercial sources of magnesium stearate include Peter Greven, Covidien/Mallinckodt and FACI.
  • a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein Compound (I) is (4-[hydroxy(diphenyl)methyl]-1- ⁇ 2-[(phenylmethyl)oxy]ethyl ⁇ -1-azoniabicyclo[2.2.2]octane bromide and is presented as a dry powder composition containing magnesium stearate at an amount of 0.6% w/w based on the total weight of the composition.
  • a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein Compound (II) is 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate and is presented as a dry powder composition containing magnesium stearate at an amount of 1.0% w/w based on the total weight of the composition.
  • Intranasal sprays may be formulated with aqueous or non-aqueous vehicles with the addition of agents such as thickening agents, buffer salts or acid or alkali to adjust the pH, isotonicity adjusting agents or anti-oxidants.
  • agents such as thickening agents, buffer salts or acid or alkali to adjust the pH, isotonicity adjusting agents or anti-oxidants.
  • Solutions for inhalation by nebulization may be formulated with an aqueous vehicle with the addition of agents such as acid or alkali, buffer salts, isotonicity adjusting agents or antimicrobials. They may be sterilized by filtration or heating in an autoclave, or presented as a non-sterile product.
  • agents such as acid or alkali, buffer salts, isotonicity adjusting agents or antimicrobials. They may be sterilized by filtration or heating in an autoclave, or presented as a non-sterile product.
  • the invention also provides a method of preparing a pharmaceutical combination product as defined herein, the method comprising either:
  • 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol, and its salts, including 4- ⁇ (1R)-2-[(6- ⁇ 2-[(2,6-dichlorobenzyl)oxy]ethoxy ⁇ hexyl)amino]-1-hydroxyethyl ⁇ -2-(hydroxymethyl)phenol triphenylacetate may be prepared as described in WO03/024439 (Example 78(i)), which is incorporated by reference herein.
  • Compound (II) as the ⁇ -phenylcinnamate salt and the triphenylacetate salt has been studied in a number of clinical pharmacology studies, including single- and repeat-dose studies. In addition, these studies have evaluated Compound (II) formulated with lactose and either cellobiose octaacetate or magnesium stearate.
  • a combination of Compound (I) bromide and Compound (II) triphenylacetate has been administered to sixteen healthy Japanese volunteers, aged 20 to 65, as part of a clinical trial to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of single inhaled doses of Compound (I) bromide and Compound (II) triphenylacetate as monotherapies and in combination.
  • This study was a randomised, double blind, placebo-controlled, four-way crossover study wherein subjects received a single dose of:
  • ⁇ -lactose monohydrate sourced from DMV Fronterra Excipients, complying with the requirements of Ph.Eur/USNF may be used. Before use, the ⁇ -lactose monohydrate may be sieved through a coarse screen (for example with a mesh size 500 or 800 microns). The level of fines in the ⁇ -lactose monohydrate, which can be measured by Sympatec, may be 4.5% w/w less than 4.5 micron.
  • Compound (I) bromide is micronised before use in an APTM microniser to give a mass median diameter of 1 to 5 microns, such as 2 to 5 microns.
  • Pharmaceutical grade magnesium stearate sourced from Peter Greven, complying with the requirements of Ph.Eur/USNF may be used as supplied with a mass median particle size of 8 to 12 microns.
  • Lactose monohydrate may be passed through a sieve and then combined with magnesium stearate and blended using either a high shear mixer (a QMM, PMA or TRV series mixer, such as TRV25 or TRV65) or a low shear tumbling blender (a Turbula mixer) to provide a magnesium stearate/lactose premix, hereinafter referred to as blend A.
  • a high shear mixer a QMM, PMA or TRV series mixer, such as TRV25 or TRV65
  • a Turbula mixer a low shear tumbling blender
  • Final blend B may be obtained as follows.
  • An quantity of blend A and compound (I) bromide may be screened, for example using a COMILTM, and then blended with the remaining blend A using either a high shear mixer (a QMM, PMA or TRV series mixer, such as TRV25 or TRV65) or a low shear tumbling blender (a Turbula mixer).
  • a high shear mixer a QMM, PMA or TRV series mixer, such as TRV25 or TRV65
  • a Turbula mixer a Turbula mixer
  • Blend Time (mins) Approximate Speed (fpm) A 6 460 B 10 590
  • the blended composition may then be transferred into blister strips (typical nominal mean quantity of blend per blister is 12.5-13.5 mg) of the type generally used for the supply of dry powder for inhalation and the blister strips were sealed in the customary fashion.
  • blister strips typically nominal mean quantity of blend per blister is 12.5-13.5 mg
  • ⁇ -lactose monohydrate which can be sourced from DMV Fronterra Excipients, complying with the requirements of Ph.Eur/USNF may be used. Before use, the ⁇ -lactose monohydrate may be sieved through a coarse screen (typical mesh size 500 microns). The level of fines in the ⁇ -lactose monohydrate, which can be measured by Sympatec, may be 4.5% w/w less than 4.5 micron.
  • Compound (II) triphenylacetate is micronised before use in an APTM microniser to give a MMD (mass median diameter) of from 1 to 5 microns, such as 2 to 5 microns, for example 1.8 microns.
  • MMD mass median diameter
  • Magnesium stearate which can be sourced from Peter Greven, complying with the requirements of Ph.Eur/USNF may be used as supplied with a mass median particle size 8 to 12 microns.
  • Lactose monohydrate may be passed through a sieve and then combined with magnesium stearate (typically 130 g) and blended using either a high shear mixer (a QMM, PMA or TRV series mixer, such as TRV25 or TRV65) or a low shear tumbling blender (a Turbula mixer) to provide a magnesium stearate/lactose premix, hereinafter referred to as blend A.
  • a high shear mixer a QMM, PMA or TRV series mixer, such as TRV25 or TRV65
  • a Turbula mixer a low shear tumbling blender
  • Final blend B may be obtained as follows.
  • An appropriate quantity of blend A and compound (II) triphenylacetate typically 5-165 g may be screened, for example using a COMILTM, and then blended with the remaining blend A using either a high shear mixer (a QMM, PMA or TRV series mixer) or a low shear tumbling blender (a Turbula mixer).
  • the final concentration of compound (II) triphenylacetate in the blends is typically in the range 0.02% w/w-0.8% w/w free base equivalent.
  • the blended composition is transferred into blister strips (typical nominal mean quantity of blend B per blister is 12.5-13.5 mg) or the type generally used for the supply of dry powder for inhalation and the blister strips are then sealed in the customary fashion.
  • Blend Time (mins) Approximate Speed (rpm) A 9 550 B 8.5 550
  • Compound (I) bromide and Compound (II) triphenylacetate as an inhalation powder may be administered in a DPI device containing two blister strips.
  • One strip contains a blend of micronised Compound (I) bromide (approximately 500 micrograms per blister), magnesium stearate and lactose monohydrate.
  • the second strip contains a blend of micronised Compound (II) triphenylacetate (approximately 25 micrograms per blister), magnesium stearate and lactose monohydrate.
  • the DPI device will deliver, when actuated, the contents of a single blister simultaneously from each of the two blister strips.
  • Each blister strip is a double foil laminate containing 30 blisters per strip.
  • Compound (I) bromide and Compound (II) triphenylacetate as an inhalation powder may be administered in a dry powder inhaler device containing two blister strips, wherein one strip contains a blend of micronised Compound (I) bromide (approximately 125 or 62.5 micrograms per blister), magnesium stearate (at an amount of 0.6% w/w of the total powder weight per blister) and lactose monohydrate.
  • the second strip contains a blend of micronised Compound (II) triphenylacetate (approximately 25 micrograms per blister), magnesium stearate and lactose monohydrate.
  • the second strip optionally further comprises 6 ⁇ ,9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester (fluticasone furoate) at an amount of approximately 100 micrograms per blister.
  • the DPI device will deliver, when actuated, the contents of a single blister simultaneously from each of the two blister strips.
  • Each blister strip is a double foil laminate containing 7, 14 or filled blisters per strip.
  • Compound (I) bromide and Compound (II) triphenylacetate as an inhalation powder may be administered in a dry powder inhaler device containing two blister strips, wherein one strip contains a blend of micronised Compound (I) bromide (approximately 125 or 62.5 micrograms per blister), Compound (II) triphenylacetate (approximately 25 micrograms per blister), magnesium stearate and lactose monohydrate.
  • the second strip contains a blend of 6 ⁇ ,9 ⁇ -difluoro-17 ⁇ -[(2-furanylcarbonyl)oxy]-11 ⁇ -hydroxy-16 ⁇ -methyl-3-oxo-androsta-1,4-diene-17 ⁇ -carbothioic acid S-fluoromethyl ester (fluticasone furoate) at an amount of approximately 100 micrograms per blister, and lactose monohydrate.
  • the DPI device will deliver, when actuated, the contents of a single blister simultaneously from each of the two blister strips.
  • Each blister strip is a double foil laminate containing 7, 14 or 30 filled blisters per strip.

Abstract

Combinations of a muscarinic acetylcholine receptor antagonist and a beta 2 agonist for inhaled administration via the nose or mouth, and methods of using them are provided.

Description

    FIELD OF THE INVENTION
  • This invention relates to pharmaceutical products and compositions for use in the treatment of chronic obstructive pulmonary disease (COPD), asthma and related diseases.
  • More particularly this invention relates to the combination of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist, and the use of said combination in treating diseases mediated via the M3 muscarinic acetylcholine receptor and/or the beta-2 adrenoreceptor.
  • More particularly this invention is concerned with novel pharmaceutical combination products comprising 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate and 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide and the use of said combination products in medicine, particularly in treating diseases mediated via the M3 muscarinic acetylcholine receptor and/or the beta-2 adrenoreceptor, for example in the prophylaxis and treatment of inflammatory or respiratory tract diseases.
  • BACKGROUND OF THE INVENTION
  • Selective β2-adrenoreceptor agonists have been used in the prophylaxis and treatment of clinical conditions for which a bronchodilating agent has been indicated. Such conditions include diseases associated with airflow obstruction such as chronic obstructive pulmonary diseases (COPD) (e.g. chronic and wheezy bronchitis, emphysema), asthma, respiratory tract infection and upper respiratory tract disease (e.g. rhinitis, including seasonal and allergic rhinitis).
  • In particular, asthma and other related disorders are typically treated with beta-2 adrenergic receptor agonists (beta-2 agonists) as they provide a bronchodilator effect to the patient, resulting in relief from the symptoms of breathlessness. Within the beta-2 agonist class there are presently available short acting compounds for immediate relief, such as salbutamol, biltolterol, pirbuterol and terbutaline. There are also longer acting compounds commercially available, such as salmeterol and formoterol. Salmeterol is available by prescription for use twice daily in the treatment of asthma.
  • Over the last two decades, inhaled anticholinergic agents have become well established as well-tolerated and effective bronchodilators for the treatment of COPD. Treatment with anticholinergics significantly improves FEV1, (forced expiratory volume in 1 second) resting and dynamic lung hyperinflation, symptoms and exercise capacity, and reduces COPD exacerbations. Currently, only a few inhaled anticholinergic bronchodilators are available: the short-acting ipratroplum bromide (ipratropium; dosed four-times-a-day) and oxitropium bromide, and the long-acting tiotropium bromide (tiotropium; dosed once-daily).
  • WO 03/024439 describes compounds of the general formula:
  • Figure US20200171009A1-20200604-C00001
  • and salts, solvates, and physiologically functional derivatives thereof.
  • The compound 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol is specifically described in WO03/024439, as are pharmaceutically acceptable salts thereof, in particular the acetate, triphenylacetate, α-phenylcinnamate, 1-naphthoate and (R)-mandelate salts. WO2005/104745 describes compounds of the formulae:
  • Figure US20200171009A1-20200604-C00002
  • WO2005/104745 specifically describes the compound 4-[hydroxy(diphenyl)methyl]-1 {2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide.
  • SUMMARY OF THE INVENTION
  • In a first aspect the present invention provides a novel pharmaceutical combination product comprising the therapeutic agents:
  • a) a compound of the formula:
  • Figure US20200171009A1-20200604-C00003
  • wherein
    X is a pharmaceutically acceptable anion;
    and
    b) a compound of the formula:
  • Figure US20200171009A1-20200604-C00004
  • or a pharmaceutically acceptable salt thereof.
  • Hereinafter, Compound (II) may refer to the free base depicted above, and/or one or more salts thereof, as dictated by the context.
  • In one embodiment the pharmaceutical combination product comprises 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate and 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide.
  • In one embodiment 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate and 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide are the sole active ingredients in said pharmaceutical combination product.
  • In another embodiment the pharmaceutical combination product of Compound (I) and Compound (II) additionally comprises an inhaled corticosteroid.
  • This invention also provides for use of the pharmaceutical combination product in the manufacture of a medicament for the treatment of conditions for which administration of one or more of the therapeutic compounds is indicated.
  • In one embodiment the use is for the manufacture of a medicament for the treatment of inflammatory or respiratory tract diseases, by simultaneous or sequential administration of Compound (I) and Compound (II).
  • In another embodiment the use is for the manufacture of a medicament for the treatment of chronic obstructive pulmonary disease (COPD) and/or asthma, by simultaneous or sequential administration of Compound (I) and Compound (II).
  • The invention also provides said pharmaceutical combination product for use in the treatment of inflammatory or respiratory tract diseases, such as chronic obstructive pulmonary disease (COPD) and/or asthma.
  • Another embodiment of the invention is a method for the treatment of inflammatory or respiratory tract diseases, comprising administering either sequentially or simultaneously, to a patient in need thereof, a pharmaceutical combination product comprising Compound (I) and Compound (II).
  • In one embodiment of the invention the inflammatory or respiratory tract disease is selected from the group consisting of chronic obstructive pulmonary disease, chronic bronchitis, asthma, chronic respiratory obstruction, pulmonary fibrosis, pulmonary emphysema, allergic rhinitis, small airways disease, bronchiectasis and cystic fibrosis.
  • In another embodiment of the invention the pharmaceutical combination product may be used for the treatment of inflammatory or respiratory tract diseases, and more specifically the treatment of chronic obstructive pulmonary disease (COPD) and/or asthma by simultaneous or sequential administration of Compound (I) and Compound (II).
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to a pharmaceutical combination product comprising
  • a) a compound of formula:
  • Figure US20200171009A1-20200604-C00005
  • wherein
    X is a pharmaceutically acceptable anion;
    and
    b) a compound of formula:
  • Figure US20200171009A1-20200604-C00006
  • or a pharmaceutically acceptable salt thereof.
  • The pharmaceutically acceptable anion depicted by X may be selected from chloride, bromide, iodide, hydroxide, sulfate, nitrate, phosphate, acetate, trifluoroacetate, fumarate, citrate, tartrate, oxalate, succinate, mandelate, methanesulfonate or p-toluenesulfonate. In one embodiment the pharmaceutically acceptable anion X is bromide.
  • For purposes herein, the structural formula for the quaternary moiety (cation) of Compound (1) is also referred to as 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane.
  • In one embodiment of the invention Compound (I) is 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide (also referred to herein as Compound (I) bromide).
  • Pharmaceutically acceptable acid addition salts of Compound (II) include those formed from hydrochloric, hydrobromic, sulphuric, citric, tartaric, phosphoric, lactic, pyruvic, acetic, trifluoroacetic, triphenylacetic, phenylacetic, substituted phenyl acetic eg. methoxyphenyl acetic, sulphamic, suiphanilic, succinic, oxalic, fumaric, maleic, malic, glutamic, aspartic, oxaloacetic, methanesuiphonic, ethanesulphonic, arylsulponic (for example p-toluenesulphonic, benzenesulphonic, naphthalenesulphonic or naphthalenedisulphonic), salicylic, glutaric, gluconic, tricarballylic, mandelic, cinnamic, substituted cinnamic (for example, methyl, methoxy, halo or phenyl substituted cinnamic, including 4-methyl and 4-methoxycinnamic acid and α-phenyl cinnamic acid), ascorbic, oleic, naphthoic, hydroxynaphthoic (for example 1- or 3-hydroxy-2-naphthoic), naphthaleneacrylic (for example naphthalene-2-acrylic), benzoic, 4-methoxybenzoic, 2- or 4-hydroxybenzoic, 4-chlorobenzoic, 4-phenylbenzoic, bezeneacrylic (for example 1,4-benzenediacrylic) and isethionic acids.
  • In one embodiment the pharmaceutically acceptable salt of Compound (II) is selected from the acetate, 1-naphthoate and (R)-mandelate salts.
  • In another embodiment the pharmaceutically acceptable salt of Compound (II) is the α-phenylcinnamate salt.
  • In another embodiment the pharmaceutically acceptable salt of Compound (II) is the triphenylacetate salt.
  • The structural formula shown above for Compound (II) may be named as 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol.
  • In one embodiment of the invention Compound (II) is 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate (also referred to as Compound (II) triphenylacetate).
  • In one embodiment the pharmaceutical combination product of the invention comprises 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide and 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate.
  • In another embodiment the pharmaceutical combination product of Compound (I) and Compound (II) additionally comprises an inhaled corticosteroid, e.g. fluticasone propionate, mometasone furoate, budesonide or 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate).
  • In one embodiment said pharmaceutical combination product comprises 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide, 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate and 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate).
  • In one embodiment, the pharmaceutical combination product of the invention comprises 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide and 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate as the sole active ingredients.
  • Compound (I), specifically 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide has been the subject of studies in animal models, and in humans, and has been found to be a long acting high-affinity pan-active muscarinic receptor antagonist which has potential for once-daily administration.
  • Compound (II), specifically 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol and its salts has been extensively tested in animal and human studies and has been found to demonstrate sustained bronchodilation over a 24 hour period in conjunction with a favourable safety profile and thus has the potential for once-daily administration.
  • Compound (I) and Compound (II), and the combination thereof, are considered to have potential in the treatment of inflammatory or respiratory tract diseases such as chronic obstructive pulmonary disease, chronic bronchitis, asthma, chronic respiratory obstruction, pulmonary fibrosis, pulmonary emphysema, allergic rhinitis, small airways disease, bronchiectasis and cystic fibrosis.
  • COPD is a chronic disease characterised by airways obstruction and reduced maximum expiratory flow from the lungs that manifests as persistent daily symptoms, such as shortness of breath (dyspnoea), and limitation of the ability to perform daily activities or exertion. Furthermore, there are periodic exacerbations of the condition that result in worsening of the day-to-day symptoms and activity limitation, and can also lead to hospitalisation of the patient because of the severity of the worsening symptoms/limitation. In addition, there is a progressive decline in lung function (disease progression) over several years.
  • Bronchodilator treatment in COPD includes but is not necessarily limited to reducing symptoms, particularly dyspnoea, to allow a patient to undertake more daily activities and other activities that require exertion, and preventing exacerbations.
  • Asthma is a chronic condition, which is characterised by widespread, variable and reversible airflow obstruction. Symptoms include coughing, wheezing, breathlessness and/or a tight feeling in the chest. Asthma attacks are generally caused by exposure to a trigger, such as pollen, dust or other allergens, which causes constriction of the airways (bronchoconstriction). It will be appreciated that a subject suffering from a condition such as asthma, may variously from time to time display no overt symptoms of the condition, or may suffer from periodic attacks during which symptoms are displayed or may experience exacerbations or worsening of the condition. In this context the term ‘treatment’ is intended to encompass prevention of such periodic attacks or exacerbations of the existing condition. Such treatment may be referred to as ‘maintenance treatment’ or ‘maintenance therapy’.
  • The amounts of Compound (I) and Compound (II), and in one embodiment of the invention, 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide and 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate, required to achieve a therapeutic effect will, of course, vary with the route of administration, the subject under treatment, the particular disorder or disease being treated, and the severity of the disease. In one embodiment, the route of administration is by inhalation via the mouth or nose. In a further embodiment, the route of administration is by inhalation via the mouth.
  • In one embodiment Compound (I), and specifically (4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide, may be administered by inhalation at a dose of from about 1 mcg to about 1000 mcg/daily, e.g. 100, 250 or 500 mcg per day. In a further embodiment, Compound (I) and specifically (4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide may be administered by inhalation at a dose of 62.5 mcg or 125 mcg per day. In general Compound (I) will be administered as a once-daily dose.
  • In a further embodiment, Compound (I), and specifically (4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide, may be administered by inhalation, once-daily, at a dose of 62.5 mcg per day.
  • In a further embodiment, Compound (I), and specifically (4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide, may be administered by inhalation, once-daily, at a dose of 125 mcg per day.
  • Compound (II) may for example be administered by inhalation at a dose of from about 1 mcg to about 400 mcg/day (calculated as the free base). In one embodiment Compound (II), and specifically 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate, may be administered by inhalation at a dose of from about 1 mcg to 100 mcg/day, for example 3, 6.25, 12.5, 25, 50 or 100 mcg/day (calculated as the free base). In general Compound (II) will be administered as a once-daily dose. In one embodiment Compound (II) may be administered by inhalation at a dose of 12.5 mcg/day. In another embodiment Compound (II) may be administered by inhalation at a dose of 25 mcg/day. In another embodiment Compound (II) may be administered by inhalation at a dose of 50 mcg/day.
  • In a further embodiment, 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate, may be administered by inhalation, once-daily, at a dose of 25 mcg per day.
  • In a further embodiment, the present invention provides a pharmaceutical combination product for once-daily administration by inhalation, comprising 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate at a dose of 25 mcg per day, and (4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide at a dose of 125 mcg per day.
  • In a further embodiment, the present invention provides a pharmaceutical combination product for once-daily administration by inhalation, comprising 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate at a dose of 25 mcg per day, and (4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide at a dose of 62.5 mcg per day.
  • When the combination additionally includes an inhaled corticosteroid, this may be used at doses compatible with those known for monotherapy. When the inhaled corticosteroid is fluticasone furoate this may be administered by inhalation at a dose of from about 25 mcg to about 800 mcg daily, and if necessary in divided doses. Thus, the daily dose of fluticasone furoate may be for example 25, 50, 100, 200, 300, 400, 600 or 800 mcg, in general as a once-daily dose. In one embodiment, the daily dose of fluticasone furoate is 100 mcg. In a further embodiment, the daily dose of fluticasone furoate is 50 mcg.
  • The individual compounds of the pharmaceutical combination product as described herein may be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations/compositions. Thus Compound (I) and Compound (II) may for example, be formulated separately and presented in separate packs or devices, or said individually formulated components may be presented in a single pack or device. Where appropriate, the individual compounds may be admixed within the same formulation, and presented as a fixed pharmaceutical combination. In general such formulations will include pharmaceutical carriers or excipients as described hereinafter, but combinations of the compounds without any excipients are also within the ambit of this invention. In one embodiment, the individual compounds of the pharmaceutical combination product may be administered simultaneously in a combined pharmaceutical formulation or composition.
  • When the pharmaceutical combination product additionally includes an inhaled corticosteroid, eg 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate) this may likewise be formulated separately, either with or without one or more pharmaceutical carriers or excipients, and presented for either sequential or simultaneous administration, or the inhaled corticosteroid may be admixed with either Compound (I) and/or Compound (II). 6α,9α-Difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester may be formulated for example as described in WO02/12265, or as described hereinafter.
  • In further aspects the invention therefore provides:
  • A pharmaceutical combination product comprising Compound (I) and Compound (II) presented separately for sequential or simultaneous administration;
    A pharmaceutical combination product comprising Compound (I) and Compound (II) presented separately but held in the same pack or device, for sequential or simultaneous administration; and
    A pharmaceutical combination product comprising Compound (I) and Compound (II) in admixture with each other for simultaneous administration.
  • In each case, each of Compound (I) and/or Compound (II) may be formulated with or without pharmaceutical carriers or excipients.
  • The present invention further provides a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein at least one of Compound (I) and Compound (II) is formulated with a pharmaceutically acceptable carrier or excipient.
  • The present invention further provides a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein each of Compound (I) and Compound (II) is formulated with a pharmaceutically acceptable carrier or excipient.
  • In one embodiment of this invention compositions of Compounds (I) and (II) include those suitable for inhalation, including fine particle powders, or mists which may be generated and administered by means of various types of inhalers for example, reservoir dry powder inhalers, unit-dose dry powder inhalers, pre-metered multi-dose dry powder inhalers, nasal inhalers or pressurized metered dose inhalers, nebulisers or insufflators.
  • The compositions may be prepared by any of the methods well known in the art of pharmacy. In general, said methods include the step of bringing the active ingredient(s) into association with the carrier which constitutes one or more accessory ingredients. In general the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired composition.
  • Powder compositions generally contain a powder mix for inhalation of the active ingredient and a suitable powder base (carrier/diluent/excipient substance) such as mono-, di or poly-saccharides (e.g. lactose or starch). Use of lactose is preferred. The lactose may be for example anhydrous lactose or α-lactose monohydrate. In one embodiment, the carrier is α-lactose monohydrate. Dry powder compositions may also include, in addition to the active ingredient and carrier, a further excipient (eg a ternary agent) such as a sugar ester, calcium stearate or magnesium stearate.
  • Alternatively, the active ingredient may be presented without excipients. For the avoidance of doubt use of the term ‘composition’ or ‘formulation’ herein refers to the active ingredients either with or without excipients or carriers.
  • The present invention further provides a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein at least one of Compound (I) and Compound (II) is formulated with a pharmaceutically acceptable carrier and a ternary agent.
  • The present invention further provides a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein Compound (II) is formulated with a pharmaceutically acceptable carrier and a ternary agent.
  • In another embodiment the present invention further provides a pharmaceutical formulation comprising a combination of Compound (I) and Compound (II) wherein both Compounds are formulated with a pharmaceutically acceptable carrier and a ternary agent.
  • The present invention further provides a pharmaceutical combination product for inhaled administration comprising 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate and (4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide each formulated separately with a pharmaceutically acceptable carrier and a ternary agent, but held in the same pack or device, for sequential or simultaneous administration.
  • In one embodiment said ternary agent is magnesium stearate.
  • The present invention further provides a pharmaceutical combination product for inhaled administration comprising 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate and (4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide each formulated separately with lactose, as a pharmaceutically acceptable carrier, and magnesium stearate, as a ternary agent, but held in the same pack or device, for sequential or simultaneous administration.
  • The compositions may be presented in unit dosage form. Dry powder compositions for topical delivery to the lung by inhalation may, for example, be presented in capsules and cartridges of for example gelatine, or blisters of for example laminated aluminium foil, for use in an inhaler or insufflator.
  • Each capsule, cartridge or blister may generally contain between 1 mcg-1000 mcg, e.g. 100 to 500 mcg of Compound (I) and/or between 1 mcg-400 mcg, e.g 1 to 100 mcg of Compound (II). Packaging of the formulation may be suitable for unit dose or multi-dose delivery. As indicated above Compound (I) and Compound (II) may be formulated independently or in admixture. Said compounds may thus be incorporated in separate unit doses or may be combined in a single unit dose with or without additional excipients as deemed necessary.
  • In a further embodiment, each capsule, cartridge or blister may contain 125 mcg or 62.5 mcg of Compound (I) and/or 25 mcg of Compound (II).
  • In yet a further embodiment, each capsule, cartridge or blister may contain 125 mcg or 62.5 mcg of (4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide and/or 25 mcg of 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate.
  • In one embodiment, a composition suitable for inhaled administration may be incorporated into a plurality of sealed dose containers provided on medicament pack(s) mounted inside a suitable inhalation device. The containers may be rupturable, peelable or otherwise openable one-at-a-time and the doses of the dry powder composition administered by inhalation on a mouthpiece of the inhalation device, as known in the art. The medicament pack may take a number of different forms, for instance a disk-shape or an elongate strip. Representative inhalation devices are the DISKHALER™ and DISKUS™ devices, marketed by GlaxoSmithKline. The DISKUS™ inhalation device is, for example, described in GB 2242134A.
  • A dry powder inhalable composition, may also be provided as a bulk reservoir in an inhalation device, the device then being provided with a metering mechanism for metering a dose of the composition from the reservoir to an inhalation channel where the metered dose is able to be inhaled by a patient inhaling at a mouthpiece of the device. Exemplary marketed devices of this type are TURBUHALER™ of AstraZeneca, TWISTHALER™ of Schering and CLICKHALER™ of Innovata.
  • A further delivery method for a dry powder inhalable composition is for metered doses of the composition to be provided in capsules (one dose per capsule) which are then loaded into an inhalation device, typically by the patient on demand. The device has means to rupture, pierce or otherwise open the capsule so that the dose is able to be entrained into the patient's lung when they inhale at the device mouthpiece. As marketed examples of such devices there may be mentioned ROTAHALER™ of GlaxoSmithKline and HANDIHALER™ of Boehringer Ingelheim.
  • A dry powder composition may also be presented in a delivery device which permits separate containment of Compound (I) and Compound (II) optionally in admixture with one or more excipients. Thus, for example, the individual compounds of the combination are administrable simultaneously but are stored separately, e.g. in separate pharmaceutical compositions, for example as described in WO 2003/061743 A1, WO 2007/012871 A1 and/or WO2007/068896. In one embodiment a delivery device permitting separate containment of actives is an inhaler device having two medicament packs in peelable blister strip form, each pack containing pre-metered doses in blister pockets arranged along its length. Said device has an internal indexing mechanism which, each time the device is actuated, peels opens a pocket of each strip and positions the packs so that each newly exposed dose of each pack is adjacent a manifold which communicates with a mouthpiece of the device. When the patient inhales at the mouthpiece, each dose is simultaneously drawn out of its associated pocket into the manifold and entrained via the mouthpiece into the patient's respiratory tract. Thus, each time the device is used, the patient is administered a combination therapy consisting of a dose from each medicament pack. A further device that permits separate containment of different compounds is DUOHALER™ of Innovata.
  • In a further embodiment, the present invention provides a dry powder inhaler (Inhaler 1) comprising two compositions presented separately, wherein a first composition comprises
      • i. 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide, and
      • ii. lactose, and
      • iii. magnesium stearate at an amount of about 0.6% w/w based on the total weight of the first composition;
        and a second composition comprises
      • i. 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate, and
      • ii. lactose, and
      • iii. magnesium stearate at an amount of about 1.0% w/w based on the total weight of the second composition.
  • In a further embodiment, the present invention provides Inhaler 1 wherein each composition is in unit dose form.
  • In a further embodiment, the present invention provides Inhaler 1 wherein the unit dose form is a capsule, cartridge or blister.
  • In a further embodiment, the present invention provides Inhaler 1 wherein 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide is present in an amount of about 125 mcg/dose.
  • In a further embodiment, the present invention provides Inhaler 1 wherein 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate is present in an amount of about 25 mcg/dose.
  • In a further embodiment, the present invention provides Inhaler 1 wherein the second composition further comprises 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate).
  • In a further embodiment, the present invention provides Inhaler 1 wherein 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate) is present in an amount of about 100 mcg/dose.
  • Spray compositions for inhalation may for example be formulated as aqueous solutions or suspensions or as aerosols delivered from pressurised packs, such as a metered dose inhaler, with the use of a suitable liquefied propellant. Aerosol compositions suitable for inhalation can be either a suspension or a solution and generally contain the pharmaceutical product and a suitable propellant such as a fluorocarbon or hydrogen-containing chlorofluorocarbon or mixtures thereof, particularly hydrofluoroalkanes, especially 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane or a mixture thereof. The aerosol composition may optionally contain additional formulation excipients well known in the art such as surfactants e.g. oleic acid, lecithin or an oligolactic acid derivative e.g. as described in WO94/21229 and WO98/34596 and/or cosolvents e.g. ethanol. Pressurised formulations will generally be retained in a canister (e.g. an aluminium canister) closed with a valve (e.g. a metering valve) and fitted into an actuator provided with a mouthpiece.
  • There is thus provided as a further aspect of the invention a pharmaceutical combination product comprising Compound (I) and Compound (II) formulated individually or in admixture, with a fluorocarbon or hydrogen-containing chlorofluorocarbon as propellant, optionally in combination with a surface-active agent and/or a co-solvent. According to another aspect of the invention, the propellant is selected from 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane and mixtures thereof.
  • Another aspect of the invention is a pharmaceutical combination product consisting of Compound (I) and Compound (II) formulated individually or in admixture, with a fluorocarbon or hydrogen-containing chlorofluorocarbon as propellant, optionally in combination with a surface-active agent and/or a cosolvent. In another embodiment of the invention the propellant is selected from 1,1,1,2-tetrafluoroethane, or 1,1,1,2,3,3,3-heptafluoro-n-propane and mixtures thereof.
  • Where appropriate compositions according to the invention may be buffered by the addition of suitable buffering agents.
  • Active ingredients for administration by inhalation desirably have a controlled particle size. The optimum particle size for inhalation into the bronchial system is usually 1-μm, preferably 2-5 μm. Particles having a size above 20 μm are generally too large when inhaled to reach the small airways. To achieve these particle sizes the particles of the active ingredient as produced may be size reduced by conventional means e.g. by micronization. The desired fraction may be separated out by air classification or sieving. Preferably, the particles will be crystalline.
  • Dry powder compositions according to the invention may comprise a carrier. The carrier when it is lactose e.g. α-lactose monohydrate, may form from about 91 to about 99%, e.g. 97.7-99.0% or 91.0-99.2% by weight of the formulation. In general, the particle size of the carrier, for example lactose, will be much greater than the inhaled medicament within the present invention. When the carrier is lactose it will typically be present as milled lactose, having a MMD (mass median diameter) of 60-90 μm.
  • The lactose component may comprise a fine lactose fraction. The ‘fine’ lactose fraction is defined as the fraction of lactose having a particle size of less than 7 μm, such as less than 6 μm, for example less than 5 μm. The particle size of the ‘fine’ lactose fraction may be less than 4.5 μm. The fine lactose fraction, if present, may comprise 2 to 10% by weight of the total lactose component, such as 3 to 6% by weight fine lactose, for example 4.5% by weight fine lactose.
  • Magnesium stearate, if present in the composition, is generally used in an amount of about 0.2 to 2%, e.g. 0.6 to 2% or 0.5 to 1.75%, e.g. 0.6%, 0.75%, 1%, 1.25% or 1.5% w/w, based on the total weight of the composition. The magnesium stearate will typically have a particle size in the range 1 to 50 μm, and more particularly 1-20 μm, e.g. 1-10 μm. Commercial sources of magnesium stearate include Peter Greven, Covidien/Mallinckodt and FACI.
  • In a further embodiment there is provided a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein Compound (I) is (4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide and is presented as a dry powder composition containing magnesium stearate at an amount of 0.6% w/w based on the total weight of the composition.
  • In yet a further embodiment, there is provided a pharmaceutical combination product comprising Compound (I) and Compound (II) wherein Compound (II) is 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate and is presented as a dry powder composition containing magnesium stearate at an amount of 1.0% w/w based on the total weight of the composition.
  • Intranasal sprays may be formulated with aqueous or non-aqueous vehicles with the addition of agents such as thickening agents, buffer salts or acid or alkali to adjust the pH, isotonicity adjusting agents or anti-oxidants.
  • Solutions for inhalation by nebulization may be formulated with an aqueous vehicle with the addition of agents such as acid or alkali, buffer salts, isotonicity adjusting agents or antimicrobials. They may be sterilized by filtration or heating in an autoclave, or presented as a non-sterile product.
  • The invention also provides a method of preparing a pharmaceutical combination product as defined herein, the method comprising either:
  • (a) preparing a separate pharmaceutical composition for administration of the individual compounds of the combination either sequentially or simultaneously, or
    (b) preparing a combined pharmaceutical composition for administration of the individual compounds together in the combination for simultaneous use, wherein the pharmaceutical composition comprises the combination together with one or more pharmaceutically acceptable carriers and/or excipients.
  • 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol, and its salts, including 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate may be prepared as described in WO03/024439 (Example 78(i)), which is incorporated by reference herein.
  • 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide is described as Example 84, in WO2005/104745 which is incorporated by reference herein.
  • Clinical Studies 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl)}-1-azoniabicyclo[2.2.2]octane bromide
  • 4-[hydroxy(diphenyl)methyl]-1-2-[(phenylmethyl)oxy]ethyl)-1-azoniabicyclo[2.2.2]octane bromide has been found to be an effective long-acting potent, pan-active anti-muscarinic bronchodilator which demonstrates slow reversibility at the human M3 receptor in vitro and long duration of action in vivo when administered directly to the lungs in pre-clinical models. The long duration of action of this compound identified using in vitro models, when administered via inhalation in animals, and subsequently in early phase studies in healthy volunteers and COPD subjects supports the potential for use of this compound as a once daily bronchodilator for COPD.
  • Several clinical pharmacology studies have been conducted using 4-[hydroxy(diphenyl)methyl]-1-(2-[(phenylmethyl)oxy]ethyl)-1-azoniabicyclo[2.2.2]octane bromide in both healthy volunteers and COPD patients to investigate the safety, tolerability, pharmacokinetics and pharmacodynamics of this compound. The bronchodilatory effects and duration of action of single inhaled doses of this compound as measured by plethysmography (sGaw, Raw) and spirometry (FEV1) were assessed in some of the above noted studies. These studies showed clinically relevant bronchodilation and 24 h duration of action for the compound.
  • In one such study, designed to evaluate the safety, efficacy and pharmacokinetics of 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide in subjects with COPD, five once-daily doses (62.5 mcg, 125 mcg, 250 mcg, 500 mcg and 1000 mcg), taken over a 14-day treatment period, produced statistically significant improvements in pulmonary function compared to placebo. All once-daily doses showed numerically greater improvement in trough FEV1 than the open label tiotropium active control (18 mcg once-daily). In addition, this study confirmed that 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide has a once-daily profile.
  • A further study evaluated the efficacy and safety of three doses (125 mcg, 250 mcg and 500 mcg) of 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide administered once-daily via a dry powder inhaler over a 28 day period in subjects with COPD. This study confirmed that 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide appears to be safe and efficacious, maintaining significant bronchodilation over twenty four hours.
  • Compound (II) (as the α-phenylcinnamate Salt or the Triphenylacetate Salt)
  • Compound (II) as the α-phenylcinnamate salt and the triphenylacetate salt has been studied in a number of clinical pharmacology studies, including single- and repeat-dose studies. In addition, these studies have evaluated Compound (II) formulated with lactose and either cellobiose octaacetate or magnesium stearate.
  • In asthmatic patients, a statistically and clinically significant improvement in trough (24-hour) FEV1 was observed for all doses of Compound (II) tested, compared to placebo. Single doses of 25 μg to 100 μg of Compound (II) triphenylacetate (containing lactose and magnesium stearate) demonstrated 24 hour duration of action as assessed by a 200 mL or greater increase in mean 23 to 24 hour post-dose FEV1 versus placebo.
  • In COPD patients, treatment with 100 mcg and 400 mcg Compound (II) alpha-phenylcinnamate (with lactose alone) achieved a clinically relevant adjusted mean difference from placebo in weighted mean trough FEV1 (22 to 24 hrs) of >100 mL. Single doses of 25 μg to 100 μg of Compound (II) triphenylacetate (containing lactose and magnesium stearate) demonstrated 24 hour duration of action as assessed by a 190 mL or greater increase in mean 23 to 24 hour post-dose FEV1 versus placebo).
  • Combination Therapy
  • A combination of Compound (I) bromide and Compound (II) triphenylacetate has been administered to sixteen healthy Japanese volunteers, aged 20 to 65, as part of a clinical trial to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of single inhaled doses of Compound (I) bromide and Compound (II) triphenylacetate as monotherapies and in combination. This study was a randomised, double blind, placebo-controlled, four-way crossover study wherein subjects received a single dose of:
      • Compound (I) bromide (500 mcg dose),
      • Compound (II) triphenylacetate (50 mcg dose),
      • Compound (1) bromide (500 mcg dose) and Compound (II) triphenylacetate (50 mcg dose) concurrently, or
      • placebo
        at each of the four treatment periods. On enrolment into the study subjects were assigned to one of four treatment sequences based on a Williams design.
  • This clinical study in healthy Japanese volunteers, evaluated the effect of Compound (I) bromide (500 mcg dose) and Compound (II) triphenylacetate (50 mcg dose) administered as single inhaled doses and concurrently (Compound (I) bromide (500 mcg dose) and Compound (II) triphenylacetate (50 mcg dose)) on lung function parameters. Single inhaled doses and the combination administered using dry powder inhalers were found to be well tolerated. In this study FEV1 values were recorded. FEV1 values were higher for all treatment groups compared with placebo. The group dosed with Compound (I) bromide (500 mcg dose) and Compound (II) triphenylacetate (50 mcg dose) concurrently showing the largest difference relative to placebo.
  • Pharmaceutical Formulations Preparation of Blends Compound (I) Bromide
  • Pharmaceutical grade α-lactose monohydrate, sourced from DMV Fronterra Excipients, complying with the requirements of Ph.Eur/USNF may be used. Before use, the α-lactose monohydrate may be sieved through a coarse screen (for example with a mesh size 500 or 800 microns). The level of fines in the α-lactose monohydrate, which can be measured by Sympatec, may be 4.5% w/w less than 4.5 micron.
  • Compound (I) bromide is micronised before use in an APTM microniser to give a mass median diameter of 1 to 5 microns, such as 2 to 5 microns.
  • Pharmaceutical grade magnesium stearate, sourced from Peter Greven, complying with the requirements of Ph.Eur/USNF may be used as supplied with a mass median particle size of 8 to 12 microns.
  • Blend A
  • Lactose monohydrate may be passed through a sieve and then combined with magnesium stearate and blended using either a high shear mixer (a QMM, PMA or TRV series mixer, such as TRV25 or TRV65) or a low shear tumbling blender (a Turbula mixer) to provide a magnesium stearate/lactose premix, hereinafter referred to as blend A.
  • Blend B
  • Final blend B may be obtained as follows. An quantity of blend A and compound (I) bromide may be screened, for example using a COMIL™, and then blended with the remaining blend A using either a high shear mixer (a QMM, PMA or TRV series mixer, such as TRV25 or TRV65) or a low shear tumbling blender (a Turbula mixer).
  • Representative Batch Formula for Compound (I) Bromide Powder Blend (62.5 Microgram Per Blister)
  • Ingredient Quantity
    Micronised Compound (I) Bromide 74.1 g
    Magnesium Stearate 75 g
    Lactose Monohydrate To 12.5 kg
    Note:
    74.1 g of Compound (I) Bromide is equivalent to 62.5 g of the free cation.
    The quantity of Compound (I) Bromide added may be adjusted to reflect the assigned
    purity of the input drug substance.
  • Representative Batch Formula for Compound (1) Bromide Powder Blend (125 Microgram Per Blister)
  • ingredient Quantity
    Micronised Compound (I) Bromide 148.3 g
    Magnesium Stearate 75 g
    Lactose Monohydrate To 12.5 kg
    Note:
    148.3 g of Compound (I) Bromide is equivalent to 125 g of the free cation.
    The quantity of Compound (I) Bromide added may be adjusted to reflect the assigned
    purity of the input drug substance.
  • Blending Parameters (Using a TRV25, 12.5 kg Scale)
  • Blend Time (mins) Approximate Speed (fpm)
    A 6 460
    B 10 590
  • Blister Strip Preparation
  • The blended composition may then be transferred into blister strips (typical nominal mean quantity of blend per blister is 12.5-13.5 mg) of the type generally used for the supply of dry powder for inhalation and the blister strips were sealed in the customary fashion.
  • Compound (II) Triphenylacetate
  • Pharmaceutical grade α-lactose monohydrate, which can be sourced from DMV Fronterra Excipients, complying with the requirements of Ph.Eur/USNF may be used. Before use, the α-lactose monohydrate may be sieved through a coarse screen (typical mesh size 500 microns). The level of fines in the α-lactose monohydrate, which can be measured by Sympatec, may be 4.5% w/w less than 4.5 micron.
  • Compound (II) triphenylacetate is micronised before use in an APTM microniser to give a MMD (mass median diameter) of from 1 to 5 microns, such as 2 to 5 microns, for example 1.8 microns.
  • Pharmaceutical grade Magnesium stearate, which can be sourced from Peter Greven, complying with the requirements of Ph.Eur/USNF may be used as supplied with a mass median particle size 8 to 12 microns.
  • Blend A
  • Lactose monohydrate may be passed through a sieve and then combined with magnesium stearate (typically 130 g) and blended using either a high shear mixer (a QMM, PMA or TRV series mixer, such as TRV25 or TRV65) or a low shear tumbling blender (a Turbula mixer) to provide a magnesium stearate/lactose premix, hereinafter referred to as blend A.
  • Blend B
  • Final blend B may be obtained as follows. An appropriate quantity of blend A and compound (II) triphenylacetate (typically 5-165 g) may be screened, for example using a COMIL™, and then blended with the remaining blend A using either a high shear mixer (a QMM, PMA or TRV series mixer) or a low shear tumbling blender (a Turbula mixer). The final concentration of compound (II) triphenylacetate in the blends is typically in the range 0.02% w/w-0.8% w/w free base equivalent.
  • Blister Strip Preparation
  • The blended composition is transferred into blister strips (typical nominal mean quantity of blend B per blister is 12.5-13.5 mg) or the type generally used for the supply of dry powder for inhalation and the blister strips are then sealed in the customary fashion.
  • EXAMPLE PREPARATIONS
  • Using the above-described procedure the following exemplary formulations may be prepared:
  • Mass of Mass of compound (II)
    Blend Magnesium triphenylacetate Mass of Quantity per
    No stearate (micronised) | lactose blister
    1 130 g 5.0 g To 13 kg 13 mg
    2 130 g 10.3 g To 13 kg 13 mg
    3 130 g 20.7 g To 13 kg 13 mg
    4 130 g 41.3 g To 13 kg 13 mg
    5 130 g 82.7 g To 13 kg 13 mg
    6 130 g 165.4 g To 13 kg 13 mg
    Note:
    The quantity of compound (II) triphenylacetate used is based on a base to salt
    conversion factor of 1.59. For example, 41 g of Compound (II) triphenylacetate is
    equivalent to 25g of the free base.
  • Example Blending Parameters (Using a TRV25, 13 kg Scale, Compound (II) Triphenylacetate Powder Blend (25 Microgram Blister))
  • Blend Time (mins) Approximate Speed (rpm)
    A 9 550
    B 8.5 550
  • Example Dry Powder Inhaler Devices
  • Compound (I) bromide and Compound (II) triphenylacetate as an inhalation powder may be administered in a DPI device containing two blister strips. One strip contains a blend of micronised Compound (I) bromide (approximately 500 micrograms per blister), magnesium stearate and lactose monohydrate. The second strip contains a blend of micronised Compound (II) triphenylacetate (approximately 25 micrograms per blister), magnesium stearate and lactose monohydrate. The DPI device will deliver, when actuated, the contents of a single blister simultaneously from each of the two blister strips. Each blister strip is a double foil laminate containing 30 blisters per strip.
  • In a further embodiment, Compound (I) bromide and Compound (II) triphenylacetate as an inhalation powder may be administered in a dry powder inhaler device containing two blister strips, wherein one strip contains a blend of micronised Compound (I) bromide (approximately 125 or 62.5 micrograms per blister), magnesium stearate (at an amount of 0.6% w/w of the total powder weight per blister) and lactose monohydrate. The second strip contains a blend of micronised Compound (II) triphenylacetate (approximately 25 micrograms per blister), magnesium stearate and lactose monohydrate. The second strip optionally further comprises 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate) at an amount of approximately 100 micrograms per blister. The DPI device will deliver, when actuated, the contents of a single blister simultaneously from each of the two blister strips. Each blister strip is a double foil laminate containing 7, 14 or filled blisters per strip.
  • In a further embodiment, Compound (I) bromide and Compound (II) triphenylacetate as an inhalation powder may be administered in a dry powder inhaler device containing two blister strips, wherein one strip contains a blend of micronised Compound (I) bromide (approximately 125 or 62.5 micrograms per blister), Compound (II) triphenylacetate (approximately 25 micrograms per blister), magnesium stearate and lactose monohydrate. The second strip contains a blend of 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate) at an amount of approximately 100 micrograms per blister, and lactose monohydrate. The DPI device will deliver, when actuated, the contents of a single blister simultaneously from each of the two blister strips. Each blister strip is a double foil laminate containing 7, 14 or 30 filled blisters per strip.
  • All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth.
  • The above description fully discloses the invention including preferred embodiments thereof. Modifications and improvements of the embodiments specifically disclosed herein are within the scope of the following claims. Without further elaboration, it is believed that one skilled in the art can, using the preceding description, utilize the present invention to its fullest extent. Therefore, the Examples herein are to be construed as merely illustrative and not a limitation of the scope of the present invention in any way. The embodiments of the invention in which an exclusive property or privilege is claimed are defined as follows.

Claims (68)

1. A pharmaceutical combination product comprising
a) a compound of the formula:
Figure US20200171009A1-20200604-C00007
wherein
X is a pharmaceutically acceptable anion; and
b) a compound of the formula:
Figure US20200171009A1-20200604-C00008
or a pharmaceutically acceptable salt thereof (Compound (II)).
2. A product according to claim 1 wherein for Compound (I) the pharmaceutically acceptable anion is selected from the group consisting of chloride, bromide, iodide, hydroxide, sulfate, nitrate, phosphate, acetate, trifluoroacetate, fumarate, citrate, tartrate, oxalate, succinate, mandelate, methanesulfonate or p-toluenesulfonate.
3. A product according to claim 1 or claim 2 wherein Compound (I) is 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide.
4. A product according to any of claims 1 to 3 wherein Compound (II) is 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol α-phenylcinnamate.
5. A product according to any of claims 1 to 3 wherein Compound (II) is 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate.
6. A product according to any of claims 1 to 5 wherein Compound (I) and Compound (II) are presented in a form adapted for separate administration.
7. A product according to any of claims 1 to 5 wherein Compound (I) and Compound (II) are presented in a form adapted for sequential administration.
8. A product according to any of claims 1 to 5 wherein Compound (I) and Compound (II) are presented in a form adapted for simultaneous administration.
9. A product according to claim 8 wherein Compound (I) and Compound (II) are in admixture with each other.
10. A product according to any of claims 1 to 9 wherein at least one of Compound (I) and Compound (II) is formulated with a pharmaceutically acceptable carrier or excipient.
11. A product according to any of claims 1 to 9 in a form suitable for administration by oral or nasal inhalation.
12. A product according to claim 11 wherein the form is suitable for administration by inhalation via a medicament dispenser selected from a reservoir dry powder inhaler, a unit-dose dry powder inhaler, a pre-metered multi-dose dry powder inhaler, a nasal inhaler or a pressurized metered dose inhaler.
13. A product as claimed in claim 12 wherein each of Compound (I) and Compound (II) is presented in the form of a dry powder composition.
14. A product as claimed in claim 13 wherein Compound (I) and Compound (II) are presented as separate compositions.
15. A product as claimed in claim 13 wherein Compound (I) and Compound (II) are presented as admixed compositions.
16. A product as claimed in claim 14 or 15 wherein at least one of said compositions of Compound (I) or Compound (II) contains a carrier.
17. A product as claimed in claim 14 or 15 wherein both compositions of Compound (I) and Compound (II) contain a carrier.
18. A product as claimed in claim 16 or 17 wherein the carrier is lactose.
19. A product as claimed in any of claims 13 to 18 wherein at least one of said compositions contains a ternary agent.
20. A product as claimed in any of claims 13 to 18 wherein both compositions contain a ternary agent.
21. A product as claimed in claim 19 wherein the ternary agent is magnesium stearate.
22. A product as claimed in claim 20, wherein the ternary agent in both compositions is magnesium stearate.
23. A product as claimed in claim 22, wherein magnesium stearate is present in an amount of about 0.6% w/w in a composition of Compound (I), and/or an amount of about 1.0% w/w in a composition of Compound (II).
24. A product as claimed in any one of claims 13 to 23 wherein said separate or admixed compositions are in unit dose form.
25. A product as claimed in claim 24 wherein the unit dose form is in a capsule, cartridge or blister pack.
26. A product as claimed in any of claims 13 to 25 wherein the composition is administered via a dry powder inhaler.
27. A product as claimed in claim 26 wherein said inhaler permits separate containment of the active ingredients.
28. A product as claimed in any one of claims 1 to 27 wherein Compound (I) is present in an amount of about 1 to 1000 mcg/dose.
29. A product as claimed in any one of claims 1 to 27 wherein Compound (I) is present in an amount of 125 mcg/dose.
30. A product as claimed in any one of claims 1 to 27 wherein Compound (I) is present in an amount of 62.5 mcg/dose.
31. A product according to any one of claims 1 to 30 wherein Compound (II) is present in an amount of 1 to 100 mcg/dose.
32. A product according to any one of claims 1 to 30 wherein Compound (II) is present in an amount of 25 mcg/dose.
33. A dry powder inhaler containing a product as defined in any of claims 1 to 32.
34. A product according to claim 11 wherein each of Compound (I) and Compound (II) is presented in the form of a spray composition for inhalation.
35. A product according to claim 34 wherein Compound (I) and Compound (II) are presented as separate or admixed compositions.
36. A product according to claim 34 or claim 35 wherein the spray composition is an aqueous solution or suspension.
37. A product according to claim 34 or 35 wherein the spray composition is an aerosol composition.
38. A product according to claim 37 comprising as propellant a fluorocarbon or hydrogen-containing chlorofluorocarbon.
39. A product according to claim 38 wherein the propellant is a hydrofluoroalkane.
40. A product according to claim 39 wherein the propellant is 1,1,1,2-tetrafluoroethane, 1,1,1,2,3,3,3-heptafluoro-n-propane or a mixture thereof.
41. A product according to any of claims 34 to 40 further comprising a co-solvent.
42. A product according to any of claims 34 to 41 further comprising a surface-active agent.
43. A product according to any of claims 1 to 42 further comprising an inhaled corticosteroid selected from the group consisting of fluticasone propionate, mometasone furoate, budesonide and 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate).
44. A product according to claim 43 wherein the inhaled corticosteroid is 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate).
45. A product according to claim 44, wherein 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate) is present in an amount of 100 mcg/dose.
46. A pressurised metered dose inhaler containing a product as defined in any of claims 1 to 11 and 34 to 45.
47. Use of the product as defined in any of claims 1 to 32 and 34 to 45 in the manufacture of a medicament for the prophylaxis or treatment of conditions for which administration of one or more of Compound (I) and Compound (II) is indicated.
48. The use according to claim 47, for the treatment of inflammatory or respiratory tract diseases, by simultaneous or sequential administration, in any order, of Compound (I) and Compound (II).
49. The use according to claim 47 or 48, for the manufacture of a medicament for the treatment of chronic obstructive pulmonary disease (COPD) and/or asthma by simultaneous or sequential administration of Compound (I) and Compound (II).
50. A method for the prophylaxis or treatment of inflammatory or respiratory tract diseases, comprising administering to a patient in need thereof, a product as defined in to any of claims 1 to 32 and 34 to 45.
51. A method according to claim 50 wherein the active ingredients of said product are administered either sequentially or simultaneously.
52. A method according to claim 50 or 51 wherein the disease is selected from the group consisting of chronic obstructive lung disease, chronic bronchitis, asthma, chronic respiratory obstruction, pulmonary fibrosis, pulmonary emphysema, allergic rhinitis, small airways disease, bronchiectasis and cystic fibrosis.
53. A method according to claim 52 wherein the disease is chronic obstructive lung disease (COPD).
54. A method according to claim 53 for the treatment of chronic obstructive pulmonary disease (COPD), by simultaneous or sequential administration of the active ingredients of a product as defined in any of claims 1 to 32 and 34 to 45.
55. A method according to any of claims 50 to 54 wherein administration is via inhalation by the mouth or nose.
56. A method according to claim 55 wherein administration is via a medicament dispenser selected from a reservoir dry powder inhaler, a pre-metered multi-dose dry powder inhaler, a nasal inhaler or a pressurized metered dose inhaler.
57. The method according to any of claims 50 to 56 wherein the product is administered once per day.
58. A product as defined in any of claims 1 to 32 and 34 to 45 for use in the treatment of inflammatory or respiratory tract diseases, by simultaneous or sequential administration, in any order, of Compound (I) and Compound (II).
59. A product according to claim 58 for use in the treatment of chronic obstructive pulmonary disease (COPD) and/or asthma by simultaneous or successive administration of Compound (I) and Compound (II).
60. A dry powder inhaler comprising two compositions presented separately, wherein the first composition comprises
i. 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide, and
ii. lactose, and
iii. magnesium stearate at an amount of about 0.6% w/w based on the total weight of the first composition;
and the second composition comprises
iv. 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate, and
v. lactose, and
vi. magnesium stearate at an amount of about 1.0% w/w based on the total weight of the second composition.
61. A dry powder inhaler according to claim 60, wherein each composition is in unit dose form.
62. A dry powder inhaler according to claim 61, wherein the unit dose form is a capsule, cartridge or blister.
63. A dry powder inhaler according to claim 61 or 62 wherein 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide is present in an amount of about 125 mcg/dose.
64. A dry powder inhaler according to claim 61 or 62 wherein 4-[hydroxy(diphenyl)methyl]-1-{2-[(phenylmethyl)oxy]ethyl}-1-azoniabicyclo[2.2.2]octane bromide is present in an amount of about 62.5 mcg/dose.
65. A dry powder inhaler according to any of claims 61 to 64 wherein 4-{(1R)-2-[(6-{2-[(2,6-dichlorobenzyl)oxy]ethoxy}hexyl)amino]-1-hydroxyethyl}-2-(hydroxymethyl)phenol triphenylacetate is present in an amount of about 25 mcg/dose.
66. A dry powder inhaler according to claim 60 wherein the second composition further comprises 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate).
67. A dry powder inhaler according to claim 61 to 65 wherein the second composition further comprises 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate).
68. A dry powder inhaler according to claim 67 wherein 6α,9α-difluoro-17α-[(2-furanylcarbonyl)oxy]-11β-hydroxy-16α-methyl-3-oxo-androsta-1,4-diene-17β-carbothioic acid S-fluoromethyl ester (fluticasone furoate) is present in an amount of about 100 mcg/dose.
US16/781,587 2009-12-01 2020-02-04 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist Abandoned US20200171009A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US16/781,587 US20200171009A1 (en) 2009-12-01 2020-02-04 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US17/514,685 US20220047565A1 (en) 2009-12-01 2021-10-29 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US17/714,241 US20220235087A1 (en) 2009-12-01 2022-04-06 Mannose-derived antagonists of fimh useful for treating disease
US18/485,799 US20240041846A1 (en) 2009-12-01 2023-10-12 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GBGB0921075.8A GB0921075D0 (en) 2009-12-01 2009-12-01 Novel combination of the therapeutic agents
GB0921075.8 2009-12-01
PCT/EP2010/068429 WO2011067212A1 (en) 2009-12-01 2010-11-29 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US201213510962A 2012-08-20 2012-08-20
US14/970,945 US9750726B2 (en) 2009-12-01 2015-12-16 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US15/678,246 US11090294B2 (en) 2009-12-01 2017-08-16 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US16/781,587 US20200171009A1 (en) 2009-12-01 2020-02-04 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/678,246 Continuation US11090294B2 (en) 2009-12-01 2017-08-16 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/925,977 Continuation US20200407390A1 (en) 2009-12-01 2020-07-10 Mannose-derived antagonists of fimh useful for treating disease
US17/514,685 Continuation US20220047565A1 (en) 2009-12-01 2021-10-29 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist

Publications (1)

Publication Number Publication Date
US20200171009A1 true US20200171009A1 (en) 2020-06-04

Family

ID=41573013

Family Applications (7)

Application Number Title Priority Date Filing Date
US13/510,962 Abandoned US20120309725A1 (en) 2009-12-01 2010-11-29 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US14/970,945 Active US9750726B2 (en) 2009-12-01 2015-12-16 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US15/678,246 Active US11090294B2 (en) 2009-12-01 2017-08-16 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US16/781,587 Abandoned US20200171009A1 (en) 2009-12-01 2020-02-04 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US17/514,685 Abandoned US20220047565A1 (en) 2009-12-01 2021-10-29 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US17/714,241 Pending US20220235087A1 (en) 2009-12-01 2022-04-06 Mannose-derived antagonists of fimh useful for treating disease
US18/485,799 Pending US20240041846A1 (en) 2009-12-01 2023-10-12 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US13/510,962 Abandoned US20120309725A1 (en) 2009-12-01 2010-11-29 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US14/970,945 Active US9750726B2 (en) 2009-12-01 2015-12-16 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US15/678,246 Active US11090294B2 (en) 2009-12-01 2017-08-16 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist

Family Applications After (3)

Application Number Title Priority Date Filing Date
US17/514,685 Abandoned US20220047565A1 (en) 2009-12-01 2021-10-29 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US17/714,241 Pending US20220235087A1 (en) 2009-12-01 2022-04-06 Mannose-derived antagonists of fimh useful for treating disease
US18/485,799 Pending US20240041846A1 (en) 2009-12-01 2023-10-12 Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist

Country Status (40)

Country Link
US (7) US20120309725A1 (en)
EP (2) EP2506844B1 (en)
JP (1) JP5816631B2 (en)
KR (2) KR101742140B1 (en)
CN (2) CN102724974A (en)
AU (6) AU2010326798B2 (en)
BR (1) BR112012012925A2 (en)
CA (1) CA2781487C (en)
CL (1) CL2012001432A1 (en)
CO (1) CO6541613A2 (en)
CR (1) CR20120265A (en)
CY (2) CY1120058T1 (en)
DK (1) DK2506844T3 (en)
DO (1) DOP2012000148A (en)
EA (1) EA023839B1 (en)
ES (1) ES2659330T3 (en)
FR (1) FR18C1022I2 (en)
GB (1) GB0921075D0 (en)
HK (1) HK1249407A1 (en)
HR (1) HRP20180312T1 (en)
HU (2) HUE036216T2 (en)
IL (1) IL219893A (en)
LT (2) LT2506844T (en)
LU (1) LUC00077I2 (en)
MA (1) MA33853B1 (en)
ME (1) ME02965B (en)
MX (1) MX351290B (en)
MY (1) MY184864A (en)
NO (1) NO2506844T3 (en)
NZ (1) NZ600026A (en)
PE (2) PE20170915A1 (en)
PL (1) PL2506844T3 (en)
PT (1) PT2506844T (en)
RS (1) RS56848B1 (en)
SG (2) SG10201407864UA (en)
SI (1) SI2506844T1 (en)
TR (1) TR201802921T4 (en)
UA (1) UA106775C2 (en)
WO (1) WO2011067212A1 (en)
ZA (1) ZA201203890B (en)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MY144753A (en) * 2004-04-27 2011-10-31 Glaxo Group Ltd Muscarinic acetylcholine receptor antagonists
ES2739352T3 (en) 2009-02-26 2020-01-30 Glaxo Group Ltd Pharmaceutical formulations comprising 4 - {(1R) -2 - [(6- {2 - [(2,6-dichlorobenzyl) oxy] ethoxy} hexyl) amino] -1-hydroxyethyl} -2- (hydroxymethyl) phenol
GB0921075D0 (en) 2009-12-01 2010-01-13 Glaxo Group Ltd Novel combination of the therapeutic agents
ES2546422T5 (en) * 2010-08-31 2023-01-20 Glaxosmithkline Ip Dev Ltd Dry powder inhalation medicinal products having moisture control properties and methods of administration thereof
KR20140041700A (en) * 2011-06-08 2014-04-04 글락소 그룹 리미티드 Dry powder inhaler compositions comprising umeclidinium
JP2014516062A (en) * 2011-06-08 2014-07-07 グラクソ グループ リミテッド Combinations containing umecridinium and corticosteroids
EP2836204B1 (en) * 2012-04-13 2020-07-08 GlaxoSmithKline Intellectual Property Development Limited Aggregate particles
GB201222679D0 (en) * 2012-12-17 2013-01-30 Glaxo Group Ltd Pharmaceutical combination products
GB201305825D0 (en) 2013-03-28 2013-05-15 Vectura Ltd New use
EP4252742A3 (en) * 2014-05-28 2023-12-27 GlaxoSmithKline Intellectual Property Development Ltd Fluticasone furoate in the treatment of copd
TR201407010A2 (en) 2014-06-16 2015-12-21 Arven Ilac Sanayi Ve Ticaret Anonim Sirketi Vilanterol formulations.
EP2957552B1 (en) 2014-06-16 2020-01-22 Arven Ilac Sanayi Ve Ticaret A.S. Vilanterol formulations
EP3157567A1 (en) 2014-06-18 2017-04-26 Cipla Limited Pharmaceutical composition comprising a beta-2-agonist and anticholinergic agent
EP3880169A1 (en) * 2018-11-12 2021-09-22 Kindeva Drug Delivery L.P. Umeclidinium and vilanterol formulation and inhaler
CN111840256A (en) * 2019-04-29 2020-10-30 上海谷森医药有限公司 Atomized inhalant and preparation method thereof
US11642333B2 (en) 2019-05-23 2023-05-09 Anovent Pharmaceutical (U.S.), Llc Inhalable formulation of a solution containing vilanterol trifenatate and umeclidinium bromide
US11304897B2 (en) * 2020-06-09 2022-04-19 Anovent Pharmaceutical (U.S.), Llc Pharmaceutical formulation containing umeclidinium bromide and vilanterol trifenatate

Family Cites Families (181)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB124009A (en) 1918-01-12 1919-03-12 Walter Edward Kimber Improvements in and connected with the Manufacture of Tools for Turning, Planing, Boring and similar purposes.
GB124010A (en) 1919-01-10 1919-03-20 Rene Emile Dior Improved Process for the Manufacture of Synthetic Resins.
GB1242211A (en) 1967-08-08 1971-08-11 Fisons Pharmaceuticals Ltd Pharmaceutical composition
GB1381872A (en) 1971-06-22 1975-01-29 Fisons Ltd Pharmaceutical compositions for inhalation
FR2182892B1 (en) 1972-03-25 1976-11-05 Degussa
GB2064336B (en) 1979-12-06 1984-03-14 Glaxo Group Ltd Device for dispensing medicaments
CY1492A (en) 1981-07-08 1990-02-16 Draco Ab Powder inhalator
CA1224992A (en) 1982-10-08 1987-08-04 Robert E. Newell Device for administering medicament to patients
US4778054A (en) 1982-10-08 1988-10-18 Glaxo Group Limited Pack for administering medicaments to patients
GB2169265B (en) 1982-10-08 1987-08-12 Glaxo Group Ltd Pack for medicament
DE3682457D1 (en) 1985-07-30 1991-12-19 Glaxo Group Ltd DEVICE FOR THE ADMINISTRATION OF MEDICINES TO PATIENTS.
GB8530365D0 (en) 1985-12-10 1986-01-22 Univ Bath Manufacture of moulded products
IT1204826B (en) 1986-03-04 1989-03-10 Chiesi Farma Spa INHALATION PHARMACEUTICAL COMPOSITIONS
CA1329526C (en) 1987-08-20 1994-05-17 Hiroshi Ikura Powdery medicine applicator device
US4994276A (en) 1988-09-19 1991-02-19 Edward Mendell Co., Inc. Directly compressible sustained release excipient
GB8820353D0 (en) 1988-08-26 1988-09-28 Staniforth J N Controlled release tablet
US5135757A (en) 1988-09-19 1992-08-04 Edward Mendell Co., Inc. Compressible sustained release solid dosage forms
US5376386A (en) 1990-01-24 1994-12-27 British Technology Group Limited Aerosol carriers
SK280967B6 (en) 1990-03-02 2000-10-09 Glaxo Group Limited Inhalation device
US6536427B2 (en) 1990-03-02 2003-03-25 Glaxo Group Limited Inhalation device
UA26230A (en) 1990-03-02 1999-07-19 Глексо Груп Лімітед SHARED USER INHALATOR WITH TREATMENT UNIT AND TREATMENT UNIT
GB9004781D0 (en) 1990-03-02 1990-04-25 Glaxo Group Ltd Device
FR2660634B1 (en) 1990-04-06 1992-10-16 Airsec Ind Sa PACKAGING FOR PRODUCTS REQUIRING THE USE OF A DEHYDRANT.
JPH0471560A (en) 1990-07-10 1992-03-06 Mect Corp Packing body
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
DE4140689B4 (en) 1991-12-10 2007-11-22 Boehringer Ingelheim Kg Inhalable powders and process for their preparation
DE69330672T2 (en) 1992-06-12 2002-06-27 Teijin Ltd PHARMACEUTICAL PREPARATION FOR APPLICATION IN THE AIRWAY
AU659328B2 (en) 1992-06-12 1995-05-11 Teijin Limited Ultrafine powder for inhalation and production thereof
GB2269992A (en) 1992-08-14 1994-03-02 Rh Ne Poulenc Rorer Limited Powder inhalation formulations
US5560490A (en) 1992-09-09 1996-10-01 Fisons Plc Pharmaceutical packaging with capsule sealing means
SE9203743D0 (en) 1992-12-11 1992-12-11 Astra Ab EFFICIENT USE
JP2924924B2 (en) 1992-12-18 1999-07-26 シェリング・コーポレーション Inhaler for powdered drugs
US5441060A (en) 1993-02-08 1995-08-15 Duke University Dry powder delivery system
JP3987963B2 (en) 1993-03-17 2007-10-10 スリーエム カンパニー Aerosol formulations containing ester-, amide- or mercaptoester-derived dispersion promoters
US5730785A (en) 1993-04-01 1998-03-24 Multisorb Technologies, Inc. Desiccant canister for desiccants and other particulate material
IS1796B (en) 1993-06-24 2001-12-31 Ab Astra Inhaled polypeptide formulation composition which also contains an enhancer compound
US5506203C1 (en) 1993-06-24 2001-02-06 Astra Ab Systemic administration of a therapeutic preparation
DE4323636A1 (en) 1993-07-15 1995-01-19 Hoechst Ag Pharmaceutical preparations from coated, poorly water-soluble pharmaceutical substances for inhalation pharmaceutical forms and processes for their preparation
GB9322014D0 (en) 1993-10-26 1993-12-15 Co Ordinated Drug Dev Improvements in and relating to carrier particles for use in dry powder inhalers
US5503662A (en) 1994-03-29 1996-04-02 Multiform Desiccants, Inc. Canister with porous plastic ends
FI95441C (en) 1994-05-31 1996-02-12 Leiras Oy Inhaler drug chamber
US6156231A (en) 1994-09-08 2000-12-05 Multisorb Technologies, Inc. Oxygen absorbing composition with cover layer
US5641425A (en) 1994-09-08 1997-06-24 Multiform Desiccants, Inc. Oxygen absorbing composition
SA95160463B1 (en) 1994-12-22 2005-10-04 استرا أكتيبولاج powders for inhalation
GB9501841D0 (en) 1995-01-31 1995-03-22 Co Ordinated Drug Dev Improvements in and relating to carrier particles for use in dry powder inhalers
DE69611608T2 (en) 1995-04-19 2001-05-03 Capitol Vial Inc CLOSED CONTAINER WITH DRYER
US6279736B1 (en) 1995-04-19 2001-08-28 Capitol Specialty Plastics, Inc. Barrier pack having an absorbing agent applied to the interior of the pack
GB9515182D0 (en) 1995-07-24 1995-09-20 Co Ordinated Drug Dev Improvements in and relating to powders for use in dry powder inhalers
GB9700226D0 (en) 1997-01-08 1997-02-26 Glaxo Group Ltd Inhalation device
US6103141A (en) 1997-01-23 2000-08-15 Multisorb Technologies, Inc. Desiccant deposit
US6126919A (en) 1997-02-07 2000-10-03 3M Innovative Properties Company Biocompatible compounds for pharmaceutical drug delivery systems
WO1998040169A1 (en) 1997-03-14 1998-09-17 The University Of Houston Methods for making fluorinated surface modifying agents, methods of using same and products made using same
US5985415A (en) 1997-11-17 1999-11-16 Reconnx, Inc. Finger joint architecture for wood products, and method and apparatus for formation thereof
US6086376A (en) 1998-01-30 2000-07-11 Rtp Pharma Inc. Dry aerosol suspension of phospholipid-stabilized drug microparticles in a hydrofluoroalkane propellant
EP1614974A1 (en) 1998-02-05 2006-01-11 E.I.Du pont de nemours and company Humidity augmenting apparatus
PL343491A1 (en) 1998-04-18 2001-08-27 Glaxo Group Ltd Pharmaceutical aerosol formulation
US7521068B2 (en) 1998-11-12 2009-04-21 Elan Pharma International Ltd. Dry powder aerosols of nanoparticulate drugs
RO122660B1 (en) 1998-11-13 2009-11-30 Skypepharma Ag Dry powder for inhalation
US20070212422A1 (en) 1999-11-10 2007-09-13 Manfred Keller Dry powder for inhalation
GB9827145D0 (en) 1998-12-09 1999-02-03 Co Ordinated Drug Dev Improvements in or relating to powders
US6315112B1 (en) 1998-12-18 2001-11-13 Smithkline Beecham Corporation Method and package for storing a pressurized container containing a drug
US6352152B1 (en) 1998-12-18 2002-03-05 Smithkline Beecham Corporation Method and package for storing a pressurized container containing a drug
US6119853A (en) 1998-12-18 2000-09-19 Glaxo Wellcome Inc. Method and package for storing a pressurized container containing a drug
US6390291B1 (en) 1998-12-18 2002-05-21 Smithkline Beecham Corporation Method and package for storing a pressurized container containing a drug
CZ298489B6 (en) 1999-03-05 2007-10-17 Chiesi Farmaceutici S. P. A. Powder for use in a dry powder inhaler
IT1309592B1 (en) 1999-03-05 2002-01-24 Chiesi Farma Spa VEHICLE PARTICLES MODIFIED FOR USE IN THE PREPARATION OF PHARMACEUTICAL FORMULATIONS IN THE FORM OF POLYMERS FOR INHALATION AND
USD440874S1 (en) 1999-07-29 2001-04-24 The Gillette Company Container
WO2001027210A1 (en) 1999-10-11 2001-04-19 Ml Laboratories Plc Medicament delivery device with moisture resistant coating
DE19957802A1 (en) 1999-12-01 2001-06-07 Trw Repa Gmbh Belt retractor system
GB0008660D0 (en) 2000-04-07 2000-05-31 Arakis Ltd The treatment of respiratory diseases
WO2001078735A1 (en) 2000-04-13 2001-10-25 Innovata Biomed Limited Medicaments for treating respiratory disorders comprising formoterol and fluticasone
PE20011227A1 (en) 2000-04-17 2002-01-07 Chiesi Farma Spa PHARMACEUTICAL FORMULATIONS FOR DRY POWDER INHALERS IN THE FORM OF HARD AGGLOMERATES
EP1353854A2 (en) 2000-05-15 2003-10-22 Glaxo Group Limited Aerosol mdi overcap containing desiccant
GB0015043D0 (en) 2000-06-21 2000-08-09 Glaxo Group Ltd Medicament dispenser
AU2001270074A1 (en) 2000-06-22 2002-01-02 Glaxo Group Limited Method and package for storing a pressurized container containing a drug
GB0019172D0 (en) 2000-08-05 2000-09-27 Glaxo Group Ltd Novel compounds
US6759398B2 (en) 2000-08-05 2004-07-06 Smithkline Beecham Corporation Anti-inflammatory androstane derivative
CA2634715A1 (en) 2000-08-05 2002-02-14 Glaxo Group Limited 6.alpha.,9.alpha.-difluoro-17.alpha.-[(2-furanylcarboxyl)oxy}-11.beta.-hydroxy-16.alpha-methyl-3-oxo-androsta-1,4-diene-17.beta.carbothioic acids-fluoromethyl ester as an anti-inflammatory agent
DE10056855A1 (en) 2000-11-16 2002-05-29 Lohmann Therapie Syst Lts Product, e.g. pharmaceutical, consisting of a laminate comprising a moisture- and/or oxygen-sensitive layer and a moisture- and/or oxygen-absorbing layer in air- and moisture-impermeable packaging
AU2002220857A1 (en) 2000-11-30 2002-06-11 Vectura Limited Particles for use in a pharmaceutical composition
ATE382335T1 (en) 2000-11-30 2008-01-15 Vectura Ltd METHOD FOR PREPARING MICROPARTICLES FOR USE IN PHARMACEUTICAL COMPOSITIONS FOR INHALATION
EP1241110A1 (en) 2001-03-16 2002-09-18 Pfizer Products Inc. Dispensing unit for oxygen-sensitive drugs
EP1243524A3 (en) 2001-03-16 2004-04-07 Pfizer Products Inc. Pharmaceutical kit for oxygen-sensitive drugs
UA77656C2 (en) 2001-04-07 2007-01-15 Glaxo Group Ltd S-fluoromethyl ester of 6-alpha, 9-alpha-difluoro-17-alpha-[(2-furanylcarbonyl)oxy]-11-beta-hydroxy-16- alpha-methyl-3-oxoandrosta-1,4-dien-17-beta-carbothioacid as anti-inflammatory agent
DE10126924A1 (en) 2001-06-01 2002-12-05 Boehringer Ingelheim Pharma Inhalation capsule contains powdered mixture of tiotropium and auxiliary, for treating asthma or chronic obstructive pulmonary disease, having capsule material of low moisture content to improve stability
JP4191034B2 (en) 2001-09-14 2008-12-03 グラクソ グループ リミテッド Phenetanolamine derivatives for the treatment of respiratory diseases
AU2002351271A1 (en) 2001-12-21 2003-07-24 Nektar Therapeutics Capsule package with moisture barrier
GB0201677D0 (en) 2002-01-25 2002-03-13 Glaxo Group Ltd Medicament dispenser
GB0208608D0 (en) * 2002-04-13 2002-05-22 Glaxo Group Ltd Composition
US20030203141A1 (en) 2002-04-25 2003-10-30 Blum John B. Blister package
US8003179B2 (en) 2002-06-20 2011-08-23 Alcan Packaging Flexible France Films having a desiccant material incorporated therein and methods of use and manufacture
US20040131805A1 (en) 2002-06-20 2004-07-08 Merical Rick L. Films having a desiccant material incorporated therein and methods of use and manufacture
US20060269708A1 (en) 2002-06-20 2006-11-30 Rick Merical Films having a desiccant material incorporated therein and methods of use and manufacture
US20030235664A1 (en) 2002-06-20 2003-12-25 Rick Merical Films having a desiccant material incorporated therein and methods of use and manufacture
US20060236868A1 (en) 2003-05-19 2006-10-26 Bester Pansegrouw Dessicant container
GB0312148D0 (en) 2003-05-28 2003-07-02 Aventis Pharma Ltd Stabilized pharmaceutical products
SE527191C2 (en) 2003-06-19 2006-01-17 Microdrug Ag Inhaler device and combined doses of tiotropium and fluticasone
GB0315889D0 (en) 2003-07-08 2003-08-13 Aventis Pharma Ltd Stable pharmaceutical products
MXPA06000370A (en) 2003-07-09 2006-03-28 Chong Kun Dang Pharm Corp The solid dispersion of tacrolimus.
GB0316341D0 (en) 2003-07-11 2003-08-13 Glaxo Group Ltd Pharmaceutical formulations
EP1643973A1 (en) * 2003-07-11 2006-04-12 Glaxo Group Limited Pharmaceutical formulations comprising magnesium stearate
US20050072958A1 (en) 2003-10-02 2005-04-07 Thomas Powers Oxygen scavenger for low moisture environment and methods of using the same
AR046103A1 (en) 2003-10-14 2005-11-23 Glaxo Group Ltd COMPOSITE OF 8-AZONIABICICLO [3.2.1] OCTANO, PHARMACEUTICAL COMPOSITION FOR THE TREATMENT OF DISEASES MEDIATED BY MUSCARINIC ACETILCOLINE RECEPTORS AND USE OF THE COMPOUND TO PREPARE SUCH COMPOSITION
WO2005044186A2 (en) 2003-10-28 2005-05-19 Glaxo Group Limited Inhalable pharmaceutical formulations employing desiccating agents and methods of administering the same
GB0326632D0 (en) * 2003-11-14 2003-12-17 Jagotec Ag Dry powder formulations
KR101315690B1 (en) 2003-12-03 2013-10-22 베링거 인겔하임 인터내셔날 게엠베하 Pre-metered dry powder inhaler for moisture-sensitive medicaments
SE0303269L (en) 2003-12-03 2005-06-04 Microdrug Ag Medical product
SE0303569L (en) 2003-12-03 2005-06-04 Microdrug Ag DPI for delivery of moisture-sensitive drugs
SE0303570L (en) 2003-12-03 2005-06-04 Microdrug Ag Moisture-sensitive medical product
SE0303270L (en) 2003-12-03 2005-06-04 Microdrug Ag Method of administration of tiotropium
AU2004294889B2 (en) 2003-12-03 2010-08-26 Boehringer Ingelheim International Gmbh Medical product containing tiotropium
GB2410192B (en) 2004-01-23 2005-12-07 Eg Technology Ltd An inhaler
MY144753A (en) 2004-04-27 2011-10-31 Glaxo Group Ltd Muscarinic acetylcholine receptor antagonists
GB0409703D0 (en) 2004-04-30 2004-06-02 Vectura Ltd Pharmaceutical compositions
SI1891974T1 (en) 2004-05-31 2010-11-30 Almirall Sa Combinations comprising antimuscarinic agents and PDE4 inhibitors
ES2257152B1 (en) 2004-05-31 2007-07-01 Laboratorios Almirall S.A. COMBINATIONS THAT INCLUDE ANTIMUSCARINIC AGENTS AND BETA-ADRENERGIC AGONISTS.
SE530006C2 (en) 2004-06-18 2008-02-05 Mederio Ag Inhaler using tub
SE0401654D0 (en) 2004-06-24 2004-06-24 Astrazeneca Ab A support structure for a drug
GB0416397D0 (en) 2004-07-22 2004-08-25 Glaxo Group Ltd Pharmaceutical formulations
US7528253B2 (en) 2004-08-16 2009-05-05 Theravance, Inc. Compounds having β2 adrenergic receptor agonist and muscarinic receptor antagonist activity
CA2583995A1 (en) 2004-10-21 2006-05-04 Boehringer Ingelheim International Gmbh Blister for inhalers
US7501011B2 (en) 2004-11-09 2009-03-10 Multisorb Technologies, Inc. Humidity control device
SE0402976L (en) 2004-12-03 2006-06-04 Mederio Ag Medical product
PE20060826A1 (en) 2004-12-06 2006-10-08 Smithkline Beecham Corp OLEOPHINIC DERIVATIVE OF 8-AZONIABICICLO [3.2.1] OCTANE AND THE PHARMACEUTICAL COMBINATION THAT INCLUDES IT
PE20061162A1 (en) 2004-12-06 2006-10-14 Smithkline Beecham Corp COMPOUNDS OLEFINIC DERIVATIVES OF 8-AZONIABICICLO [3.2.1] OCTANES
WO2006063050A1 (en) 2004-12-08 2006-06-15 Perfecseal, Inc. A sterilizable package with a high barrier to oxygen
KR20070100720A (en) 2004-12-27 2007-10-11 킹 파머슈티칼스 리서치 앤드 디벨로프먼트 아이엔씨 Oxygen-impervious packaging with optional oxygen scavenger, stabilized thyroid hormone compositions and methods for storing thyroid hormone pharmaceutical compositions
US20060144733A1 (en) 2004-12-30 2006-07-06 3M Innovative Properties Company Container assembly and method for humidity control
WO2006086130A2 (en) 2005-02-10 2006-08-17 Glaxo Group Limited Process for crystallizing lactose particles for use in pharmaceutical formulations
CN101155590A (en) 2005-02-10 2008-04-02 葛兰素集团有限公司 Processes for making lactose utilizing pre-classification techniques and pharmaceutical formulations formed therefrom
DE202005002409U1 (en) 2005-02-15 2005-07-07 Microdrug Ag Medicinal product containing tiotropium, for treatment of asthma or chronic obstructive pulmonary disease, comprising powdered drug and carrier in container with high protection dry seal, for use in powder inhaler
DE202005004659U1 (en) 2005-03-22 2005-07-07 Microdrug Ag Predosed dry powder inhaler containing tiotropium, for treatment of respiratory tract diseases such as asthma, with powdered drug and carrier enclosed in container with high protection dry seal
GB0507165D0 (en) 2005-04-08 2005-05-18 Glaxo Group Ltd Novel crystalline pharmaceutical product
JP4659523B2 (en) 2005-04-26 2011-03-30 共同印刷株式会社 Blister film and blister packaging container
WO2006124556A2 (en) 2005-05-13 2006-11-23 Glaxo Group Limited Inhalable pharmaceutical formulations employing lactose anhydrate and methods of administering the same
DE102005022862A1 (en) 2005-05-18 2006-12-14 Airsec S.A.S Capsules for inhalers
GB0523653D0 (en) * 2005-11-21 2005-12-28 Novartis Ag Organic compounds
US20060278965A1 (en) 2005-06-10 2006-12-14 Foust Donald F Hermetically sealed package and methods of making the same
GB0515584D0 (en) 2005-07-28 2005-09-07 Glaxo Group Ltd Medicament dispenser
EP1937341A1 (en) 2005-09-28 2008-07-02 Mederio AG Inhaler device for moisture sensitive drugs and method of operating an inhaler device.
GB0520794D0 (en) 2005-10-12 2005-11-23 Innovata Biomed Ltd Inhaler
EP1780005B1 (en) 2005-10-18 2009-03-11 Alcan Technology & Management Ltd. Coldformable laminate for blister bottom sheet
EP1787800A1 (en) 2005-11-17 2007-05-23 Alcan Technology & Management Ltd. Cold-workable laminate used for the base of a blister pack
AR058289A1 (en) 2005-12-12 2008-01-30 Glaxo Group Ltd COLLECTOR TO BE USED IN MEDICINAL DISPENSER
CA2642988C (en) 2006-02-27 2015-10-27 Takeda Pharmaceutical Company Limited Pharmaceutical product comprising a benzimidazole in combination with a desiccant
KR100764640B1 (en) 2006-03-08 2007-10-10 세원셀론텍(주) Adipose tissue regeneration kit
WO2007109606A2 (en) 2006-03-17 2007-09-27 Csp Technologies, Inc. Moisture-tight primary packaging for a dry powder inhaler
US20090123391A1 (en) 2006-03-22 2009-05-14 3M Innovative Properties Company Novel Formulations
TW200803870A (en) 2006-03-22 2008-01-16 Glaxo Group Ltd Novel formulations
AU2006341188A1 (en) 2006-03-24 2007-10-04 Ian Simon Tracton Stable packaged dosage form and process therefor
CA2650813A1 (en) 2006-05-24 2007-11-29 Boehringer Ingelheim International Gmbh New long-acting drug combinations for the treatment of respiratory diseases
GB0615108D0 (en) 2006-07-28 2006-09-06 Glaxo Group Ltd Novel formulations
RU2448026C2 (en) 2006-08-03 2012-04-20 Мерк Патент Гмбх Package with pharmaceutical preparations
US20090291146A1 (en) 2006-08-09 2009-11-26 Glaxo Group Limited Process for manufacturing lactose
US20090298742A1 (en) 2006-08-09 2009-12-03 Glaxo Group Limited Process for manufacturing lactose
FI20065636A0 (en) 2006-10-04 2006-10-04 Lab Pharma Ltd Desiccant system for controlling the moisture of inhalable powders
WO2008049842A2 (en) 2006-10-26 2008-05-02 Boehringer Ingelheim International Gmbh Egfr kinase inhibitor combinations for the treatment of respiratory and gastrointestinal disorders
US20080178559A1 (en) 2007-01-26 2008-07-31 Multisorb Technologies, Inc. Sorbent Packaging
MY158271A (en) 2007-03-29 2016-09-30 Multisorb Tech Inc Method for selecting adsorptive composite barriers for packaging applications
US7549272B2 (en) 2007-04-16 2009-06-23 Multisorb Technologies, Inc. Sorbent canister with beveled edges
WO2008135570A1 (en) 2007-05-07 2008-11-13 Airsec Container with moisture control capacity
UY31233A1 (en) 2007-07-21 2009-03-02 PACKING UNITS WITH A DESIRING AGENT
UY31235A1 (en) 2007-07-21 2009-03-02 NEW PULVERULENT DRUGS CONTAINING TIOTROPY AND SALMETEROL, AS WELL AS LACTOSE AS EXCIPIENT
WO2009029029A1 (en) 2007-08-24 2009-03-05 Astrazeneca Ab Inhaler for powdered substances with desiccant compartment
KR20100063116A (en) 2007-09-12 2010-06-10 글락소 그룹 리미티드 Novel combination of therapeutic agents
EP2080523A1 (en) 2008-01-15 2009-07-22 CHIESI FARMACEUTICI S.p.A. Compositions comprising an antimuscarinic and a long-acting beta-agonist
CA2716124A1 (en) 2008-02-20 2009-08-27 Boehringer Ingelheim International Gmbh Powder inhalers
JP4983657B2 (en) 2008-03-11 2012-07-25 富士通株式会社 E-mail management program, recording medium recording the program, communication terminal, and e-mail management method
EP2127628A1 (en) 2008-05-27 2009-12-02 Ranbaxy Laboratories Limited Unit dose pack
US9095324B2 (en) 2008-06-20 2015-08-04 Boston Scientific Scimed, Inc. Package assembly
ES2461496T3 (en) 2008-10-02 2014-05-20 Respivert Limited MAP p38 kinase inhibitors
UY32297A (en) 2008-12-22 2010-05-31 Almirall Sa MESILATE SALT OF 5- (2 - {[6- (2,2-DIFLUORO-2-PHENYLITOXI) HEXIL] AMINO} -1-HYDROXYETHYL) -8-HYDROXYCHINOLIN-2 (1H) -ONA AS A RECEIVER AGONIST B (BETA ) 2 ACRENERGIC
WO2010097114A1 (en) * 2009-02-26 2010-09-02 Glaxo Group Limited Novel combination of therapeutic agents
ES2739352T3 (en) * 2009-02-26 2020-01-30 Glaxo Group Ltd Pharmaceutical formulations comprising 4 - {(1R) -2 - [(6- {2 - [(2,6-dichlorobenzyl) oxy] ethoxy} hexyl) amino] -1-hydroxyethyl} -2- (hydroxymethyl) phenol
CN102458543B (en) 2009-05-18 2015-02-04 3M创新有限公司 Dry powder inhalers
EP2277799B8 (en) 2009-07-23 2012-04-11 Airsec S.A.S. Hydrated humidity control substance and process for its preparation
GB0921075D0 (en) 2009-12-01 2010-01-13 Glaxo Group Ltd Novel combination of the therapeutic agents
ES2546422T5 (en) 2010-08-31 2023-01-20 Glaxosmithkline Ip Dev Ltd Dry powder inhalation medicinal products having moisture control properties and methods of administration thereof
JP2014516062A (en) 2011-06-08 2014-07-07 グラクソ グループ リミテッド Combinations containing umecridinium and corticosteroids
KR20140041700A (en) 2011-06-08 2014-04-04 글락소 그룹 리미티드 Dry powder inhaler compositions comprising umeclidinium
GB201222679D0 (en) 2012-12-17 2013-01-30 Glaxo Group Ltd Pharmaceutical combination products

Also Published As

Publication number Publication date
IL219893A (en) 2017-01-31
US9750726B2 (en) 2017-09-05
EP3335707A1 (en) 2018-06-20
RS56848B1 (en) 2018-04-30
UA106775C2 (en) 2014-10-10
PT2506844T (en) 2018-02-28
NZ600026A (en) 2014-06-27
ZA201203890B (en) 2013-11-27
PE20170915A1 (en) 2017-07-12
AU2016262698A1 (en) 2016-12-15
PE20130042A1 (en) 2013-02-09
KR20170061719A (en) 2017-06-05
AU2018282427A1 (en) 2019-01-17
AU2016262698B2 (en) 2018-10-25
US20160095840A1 (en) 2016-04-07
SG181087A1 (en) 2012-07-30
US20220047565A1 (en) 2022-02-17
EP3335707B1 (en) 2024-04-17
KR101830728B1 (en) 2018-04-04
LTC2506844I2 (en) 2022-10-10
AU2018282427B2 (en) 2021-04-01
CY2018017I2 (en) 2018-09-05
MX2012006310A (en) 2012-06-19
HUS1800027I1 (en) 2018-08-28
BR112012012925A2 (en) 2016-08-16
EP2506844B1 (en) 2017-12-20
AU2018282427C1 (en) 2022-06-30
KR101742140B1 (en) 2017-05-31
CA2781487A1 (en) 2011-06-09
AU2014204459A1 (en) 2014-07-31
WO2011067212A1 (en) 2011-06-09
GB0921075D0 (en) 2010-01-13
FR18C1022I2 (en) 2020-03-20
IL219893A0 (en) 2012-07-31
CY1120058T1 (en) 2018-09-05
AU2021204302A1 (en) 2021-07-22
CR20120265A (en) 2012-08-14
EA201290266A1 (en) 2013-02-28
MX351290B (en) 2017-10-09
FR18C1022I1 (en) 2018-07-13
AU2023219901A1 (en) 2023-09-14
EP2506844A1 (en) 2012-10-10
US20240041846A1 (en) 2024-02-08
CN102724974A (en) 2012-10-10
KR20120092163A (en) 2012-08-20
US20220235087A1 (en) 2022-07-28
CN107412229A (en) 2017-12-01
SI2506844T1 (en) 2018-04-30
AU2014204459B2 (en) 2016-08-25
MY184864A (en) 2021-04-28
AU2010326798A1 (en) 2012-06-21
EA023839B1 (en) 2016-07-29
JP5816631B2 (en) 2015-11-18
MA33853B1 (en) 2012-12-03
DOP2012000148A (en) 2012-07-31
HUE036216T2 (en) 2018-06-28
ES2659330T3 (en) 2018-03-14
US11090294B2 (en) 2021-08-17
CO6541613A2 (en) 2012-10-16
TR201802921T4 (en) 2018-03-21
LUC00077I2 (en) 2018-08-14
CY2018017I1 (en) 2018-09-05
SG10201407864UA (en) 2014-12-30
LT2506844T (en) 2018-03-12
PL2506844T3 (en) 2018-06-29
NO2506844T3 (en) 2018-05-19
LTPA2018011I1 (en) 2018-06-25
DK2506844T3 (en) 2018-02-05
JP2013512270A (en) 2013-04-11
ME02965B (en) 2018-07-20
US20180008582A1 (en) 2018-01-11
HRP20180312T1 (en) 2018-03-23
CL2012001432A1 (en) 2012-10-05
CA2781487C (en) 2018-10-16
US20120309725A1 (en) 2012-12-06
HK1249407A1 (en) 2018-11-02
AU2010326798B2 (en) 2014-08-07

Similar Documents

Publication Publication Date Title
US11090294B2 (en) Combinations of a muscarinic receptor antagonist and a beta-2 adrenoreceptor agonist
US20140113888A1 (en) Novel Combination of Therapeutic Agents
US20140116434A1 (en) Dry Powder Inhaler Compositions
US9795561B2 (en) Combination of umeclidinium, fluticasone propionate and salmeterol xinafoate for use in the treatment of inflammatory or respiratory tract diseases
WO2010097114A1 (en) Novel combination of therapeutic agents

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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