US20180153918A1 - Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof - Google Patents

Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof Download PDF

Info

Publication number
US20180153918A1
US20180153918A1 US15/875,308 US201815875308A US2018153918A1 US 20180153918 A1 US20180153918 A1 US 20180153918A1 US 201815875308 A US201815875308 A US 201815875308A US 2018153918 A1 US2018153918 A1 US 2018153918A1
Authority
US
United States
Prior art keywords
mycobacterium
encapsulated
aminoglycoside
free
amikacin
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
US15/875,308
Inventor
Jeff Weers
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.)
Insmed Inc
Original Assignee
Insmed Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=38123420&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20180153918(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Insmed Inc filed Critical Insmed Inc
Priority to US15/875,308 priority Critical patent/US20180153918A1/en
Assigned to TRANSAVE, INC. reassignment TRANSAVE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WEERS, JEFF
Assigned to TRANSAVE, LLC reassignment TRANSAVE, LLC MERGER (SEE DOCUMENT FOR DETAILS). Assignors: TRANSAVE, INC.
Assigned to TRANSAVE, INC. reassignment TRANSAVE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WEERS, JEFF
Assigned to INSMED INCORPORATED reassignment INSMED INCORPORATED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TRANSAVE, LLC
Publication of US20180153918A1 publication Critical patent/US20180153918A1/en
Priority to US16/746,205 priority patent/US20200390758A1/en
Priority to US18/087,506 priority patent/US20230133762A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • 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
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • 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
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/7036Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin having at least one amino group directly attached to the carbocyclic ring, e.g. streptomycin, gentamycin, amikacin, validamycin, fortimicins
    • 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
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0078Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a nebulizer such as a jet nebulizer, ultrasonic nebulizer, e.g. in the form of aqueous drug solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K9/008Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy comprising drug dissolved or suspended in liquid propellant for inhalation via a pressurized metered dose inhaler [MDI]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M11/00Sprayers or atomisers specially adapted for therapeutic purposes
    • A61M11/001Particle size control
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M11/00Sprayers or atomisers specially adapted for therapeutic purposes
    • A61M11/005Sprayers or atomisers specially adapted for therapeutic purposes using ultrasonics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M11/00Sprayers or atomisers specially adapted for therapeutic purposes
    • A61M11/006Sprayers or atomisers specially adapted for therapeutic purposes operated by applying mechanical pressure to the liquid to be sprayed or atomised
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M11/00Sprayers or atomisers specially adapted for therapeutic purposes
    • A61M11/02Sprayers or atomisers specially adapted for therapeutic purposes operated by air or other gas pressure applied to the liquid or other product to be sprayed or atomised
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M15/00Inhalators
    • A61M15/009Inhalators using medicine packages with incorporated spraying means, e.g. aerosol cans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M15/00Inhalators
    • A61M15/0091Inhalators mechanically breath-triggered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M15/00Inhalators
    • A61M15/08Inhaling devices inserted into the nose
    • 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
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • 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
    • A61P31/08Antibacterial agents for leprosy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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

  • respiratory diseases are the number one cause of world-wide mortality, with at least 20% of the world's population afflicted. Over 25 million Americans have chronic lung disease, making it the number one disabler of American workers ( ⁇ $50B/yr), and the number three cause of mortality.
  • inhaled tobramycin (TOBI®, Chiron Corp, Emeryville, Calif.), is a nebulized form of tobramycin, that has been shown to have improved efficacy and reduced nephro- and oto-toxicity relative to injectable aminoglycosides.
  • Sustained release formulations of antiinfectives are achieved by encapsulating the antiinfective in a liposome. Improving pulmonary targeting with sustained release formulations would further improve the therapeutic index by increasing local concentrations of drug and reducing systemic exposure. Improvements in targeting are also expected to reduce dose requirements.
  • Achieving sustained release of drugs in the lung is a difficult task, given the multiple clearance mechanisms that act in concert to rapidly remove inhaled drugs from the lung.
  • These clearance methods include: (a) rapid clearance from the conducing airways over a period of hours by the mucociliary escalator, (b) clearance of particulates from the deep lung by alveolar macrophages; (c) degradation of the therapeutic by pulmonary enzymes and; (d) rapid absorption of small molecule drugs into the systemic circulation.
  • Absorption of small molecule drugs has been shown to be nearly quantitative, with an absorption time for hydrophilic small molecules of about 1 hr, and an absorption time for lipophilic drugs of about 1 min.
  • the absorption half-life from the lung is on the order of 1.5 hr.
  • High initial peak concentrations of drug can lead to adaptive resistance, while a substantial time with levels below or near the effective minimum inhibitory concentration (MIC), may lead to selection of resistant phenotypes.
  • MIC effective minimum inhibitory concentration
  • liposomal sustained release formulations It is assumed that only the “free” (un-encapsulated) drug has bactericidal activity.
  • One potential disadvantage of liposomal sustained release formulations is that the encapsulation of drug in the liposomal formulation decreases the concentration of free drug reaching the lung pathogens, drug which is needed to achieve efficient killing of bacteria immediately following administration.
  • the subject invention results from the realization that administering a pharmaceutical composition comprising both free and liposome encapsulated antiinfective results in improved treatment of pulmonary infections.
  • the present invention relates to a system for treating or providing prophylaxis against a pulmonary infection, wherein the system comprises a pharmaceutical formulation comprising mixtures of free and lipid-based composition encapsulated antiinfective, wherein the amount of free antiinfective is sufficient to provide for immediate bactericidal activity, and the amount of encapsulated antiinfective is sufficient to provide sustained bactericidal activity, and reduce the development of resistant strains of the infectious agent, and an inhalation delivery device.
  • the free form of the antiinfective is available to provide a bolus of immediate antimicrobial activity.
  • the slow release of antiinfective from the lipid-based composition following pulmonary administration is analogous to continuous administration of the antiinfective, thereby providing for sustained levels of antiinfective in the lungs.
  • the sustained AUC levels provides prolonged bactericidal activity between administration. Further, the sustained levels provided by the release of antiinfective from the lipid-based composition is expected to provide improved protection against the development of resistant microbial strains.
  • Combinations of free and encapsulated drug can be achieved by: (a) formulation of mixtures of free and encapsulated drug that are stable to the nebulization; (b) formulation of encapsulated drug which leads to burst on nebulization.
  • the ratio of free to encapsulated drug is contemplated to be between about 1:100 w:w and about 100:1 w:w, and may be determined by the minimum inhibitory concentration of the infectious agent and the sustained release properties of the formulation.
  • the ratio of free to encapsulated drug can be optimized for a given infectious agent and drug formulation based on known pharmacodynamic targets for bacterial killing and prevention of resistance. Schentag, J. J. J Chemother. 1999, 11, 426-439.
  • the present invention relates to the aforementioned system wherein the antiinfective is selected from the group consisting of antibiotic agents, antiviral agents, and antifungal agents.
  • the antiinfective is an antibiotic selected from the group consisting of cephalosporins, quinolones, fluoroquinolones, penicillins, beta lactamase inhibitors, carbepenems, monobactams, macrolides, lincosamines, glycopeptides, rifampin, oxazolidonones, tetracyclines, aminoglycosides, streptogramins, and sulfonamides.
  • the antiinfective is an aminoglycoside.
  • the antiinfective is amikacin, gentamicin, or tobramycin.
  • the lipid-based composition is a liposome.
  • the liposome comprises a mixture of unilamellar vesicles and multilamellar vesicles.
  • the liposome comprises a phospholipid and a sterol.
  • the liposome comprises a phosphatidylcholine and a sterol.
  • the liposome comprises dipalmitoylphosphatidylcholine (DPPC) and a sterol.
  • the liposome comprises dipalmitoylphosphatidylcholine (DPPC) and cholesterol.
  • the present relates to the aforementioned system wherein the antiinfective is an aminogylcoside and the liposome comprises DPPC and cholesterol.
  • the antiinfective is amikacin
  • the liposome comprises DPPC and cholesterol
  • the liposome comprises a mixture of unilamellar vesicles and multilamellar vesicles.
  • the present invention relates to the aforementioned system, wherein the ratio by weight of free antiinfective to antiinfective encapsulated in a lipid-based composition is between about 1:100 an about 100:1. In a further embodiment, the ratio by weight is between about 1:10 and about 10:1. In a further embodiment, the ratio by weight is between about 1:2 and about 2:1.
  • the present invention relates to a method for treating or providing prophylaxis against a pulmonary infection in a patient, the method comprising: administering an aerosolized pharmaceutical formulation comprising the antiinfective to the lungs of the patient, wherein the pharmaceutical formulation comprises mixtures of free and lipid-based composition encapsulated antiinfectives, and the amount of free antiinfective is sufficient to provide for bactericidal activity, and the amount of encapsulated antiinfective is sufficient to reduce the development of resistant strains of the infectious agent.
  • the aforementioned method comprises first determining the minimum inhibitory concentration (MIC) of an antiinfective for inhibiting pulmonary infections, and wherein the amount of free antiinfective is at least 2 times the MIC, preferably greater than 4 times the MIC, and preferably greater than 10 times the MIC of the antiinfective agent, where the MIC is defined as either the minimum inhibitory concentration in the epithelial lining of the lung, or alternatively the minimum inhibitory concentration in the solid tissue of the lung (depending on the nature of the infection).
  • MIC minimum inhibitory concentration
  • the present invention relates to the aforementioned method, wherein the aerosolized pharmaceutical formulation is administered at least once per week.
  • the present invention relates to the aforementioned method, wherein the antiinfective is selected from the group consisting of antibiotic agents, antiviral agents, and antifungal agents.
  • the antiinfective is an antibiotic selected from the group consisting of cephalosporins quinolones, fluoroquinolones, penicillins, beta lactamase inhibitors, carbepenems, monobactams, macrolides, lincosamines, glycopeptides, rifampin, oxazolidonones, tetracyclines, aminoglycosides, streptogramins, and sulfonamides.
  • the antiinfective is an aminoglycoside.
  • the antiinfective is amikacin, gentamicin, or tobramycin.
  • the lipid-based composition is a liposome.
  • the liposome encapsulated antiinfective comprises a phosphatidylcholine in admixture with a sterol.
  • the sterol is cholesterol.
  • the liposome encapsulated antiinfective comprises a mixture of unilamellar vesicles and multilamellar vesicles.
  • the liposome encapsulated antiinfective comprises a phosphatidytcholine in admixture with cholesterol, and wherein the liposome encapsulated antiinfective comprises a mixture of unilamellar vesicles and multilamellar vesicles.
  • the ratio of the area under the lung concentration/time curve to the MIC at 24 hr is greater than 25, preferably greater than 100, and preferably greater than 250.
  • the therapeutic ratio of free/encapsulated drug and the required nominal dose can be determined with standard pharmacokinetic models, once the efficiency of pulmonary delivery and clearance of the drug product are established with the aerosol delivery device of choice.
  • the present invention relates to a method of treating a patient for a pulmonary infection comprising a cycle of treatment with lipid-based composition encapsulated antiinfective to enhance bacterial killing and reduce development of phenotypic resistance, following by a cycle of no treatment to reduce the development of adaptive resistance.
  • the treatment regimen may be determined by clinical research.
  • the treatment regime may be an on-cycle treatment for about 7, 14, 21, or 30 days, followed by an off-cycle absence of treatment for about 7, 14, 21, or 30 days.
  • the present invention relates to a method for reducing the loss of antiinfective encapsulated in lipid-based compositions upon nebulization comprising administering the antiinfective encapsulated in lipid-based compositions with free antiinfective.
  • the systems and methods of the present invention are useful for treating, for example, lung infections in cystic fibrosis patients, chronic obstructive pulmonary disease (COPD), bronchiectasis, acterial pneumonia, and in acute bronchial exacerbations of chronic bronchitis (ABECB).
  • COPD chronic obstructive pulmonary disease
  • bronchiectasis acterial pneumonia
  • ABSECB chronic bronchial exacerbations of chronic bronchitis
  • the technology is useful in the treatment of intracellular infections including Mycobacterium tuberculosis, and inhaled agents of bioterror (e.g., anthrax and tularemia).
  • bioterror e.g., anthrax and tularemia
  • the technology may also be used as a phophylactic agent to treat opportunistic fungal infections (e.g., aspergillosis) in immunocompromised patients (e.g., organ transplant or AIDS patients).
  • a system comprising a pharmaceutical composition comprising both free and lipid-based composition encapsulated antiinfective and an inhalation device.
  • Formulating the antiinfective as a mixture of free and lipid-based composition encapsulated antiinfective provides several advantages, some of which include: (a) provides for a bolus of free antiinfective for immediate bactericidal activity and a sustained level of antiinfective for prevention of resistance; (b) simplifies the manufacturing process, as less free antiinfective need be removed via diafiltration; and (c) allows for greater antiinfective contents to be achieved in the drug product.
  • FIG. 1 depicts the plot of lung concentration ( ⁇ g/ml) as a function of time following nebulization of unencapsulated tobramycin at a nominal dose of 300 mg (TOBI®, Chiron Corp., Emeryville, Calif.), and liposomal amikacin at a nominal dose of 100 mg.
  • Lung concentrations for both drug products are calculated assuming a volume of distribution for aminoglycosides in the lung of 200 ml.
  • the tobramycin curve was determined by pharmacokinetic modeling of the temporal tobramycin plasma concentration curve (Le Brun thesis, 2001).
  • an element means one element or more than one element.
  • antibacterial is are-recognized and refers to the ability of the compounds of the present invention to prevent, inhibit or destroy the growth of microbes of bacteria.
  • antiinfective and “antiinfective agent” are used interchangeably throughout the specification to describe a biologically active agent which can kill or inhibit the growth of certain other harmful pathogenic organisms, including but not limited to bacteria, yeasts and fungi, viruses, protozoa or parasites, and which can be administered to living organisms, especially animals such as mammals, particularly humans.
  • antimicrobial is are-recognized and refers to the ability of the compounds of the present invention to prevent, inhibit or destroy the growth of microbes such as bacteria, fungi, protozoa and viruses.
  • bioavailable is art-recognized and refers to a form of the subject invention that allows for it, or a portion of the amount administered, to be absorbed by, incorporated to, or otherwise physiologically available to a subject or patient to whom it is administered.
  • illness refers to any illness caused by or related to infection by an organism.
  • lipid-based composition refers to compositions that primarily comprise lipids.
  • Non-limiting examples of lipid-based compositions may take the form of coated lipid particles, liposomes, emulsions, micelles, and the like.
  • mammals include humans, primates, bovines, porcines, canines, felines, and rodents (e.g., mice and rats).
  • microbe is art-recognized and refers to a microscopic organism. In certain embodiments the term microbe is applied to bacteria. In other embodiments the term refers to pathogenic forms of a microscopic organism.
  • a “patient,” “subject” or “host” to be treated by the subject method may mean either a human or non-human animal.
  • pharmaceutically-acceptable salts refers to the relatively non-toxic, inorganic and organic acid addition salts of compounds, including, for example, those contained in compositions of the present invention.
  • prodrug is art-recognized and is intended to encompass compounds which, under physiological conditions, are converted into the antibacterial agents of the present invention.
  • a common method for making a prodrug is to select moieties which are hydrolyzed under physiological conditions to provide the desired compound.
  • the prodrug is converted by an enzymatic activity of the host animal or the target bacteria.
  • treating is are-recognized and refers to curing as well as anneliorating at least one symptom of any condition or disease.
  • the lipids used in the pharmaceutical formulations of the present invention can be synthetic, semi-synthetic or naturally-occurring lipids, including phospholipids, tocopherols, sterols, fatty acids, glycoproteins such as albumin, negatively-charged lipids and cationic lipids.
  • phosholipids they could include such lipids as egg phosphatidylcholine (EPC), egg phosphatidylglycerol (EPG), egg phosphatidylinositol (EPI), egg phosphatidylserine (EPS), phosphatidylethanolamine (EPE), and phosphatidic acid (EPA); the soya counterparts, soy phosphatidylcholine (SPC); SPG, SPS, SPI, SPE, and SPA; the hydrogenated egg and soya counterparts (e.g., HEPC, HSPC), other phospholipids made up of ester linkages of fatty acids in the 2 and 3 of glycerol positions containing chains of 12 to 26 carbon atoms and different head groups in the I position of glycerol that include choline, glycerol, inositol, serine, ethanolamine, as well as the corresponding phosphatidic acids.
  • EPC egg phosphatidyl
  • the chains on these fatty acids can be saturated or unsaturated, and the phospholipid may be made up of fatty acids of different chain lengths and different degrees of unsaturation.
  • the compositions of the formulations can include dipalmitoylphosphatidylcholine (DPPC), a major constituent of naturally-occurring lung surfactant.
  • DPPC dipalmitoylphosphatidylcholine
  • DMPC dimyristoylphosphatidycholine
  • DMPG dimyristoylphosphatidylglycerol
  • DPPQ dipalmitoylphosphatideholine
  • DPPG dipalmitoylphosphatidylglycerol
  • DSPQ dipalmitoylphosphatidylglycerol
  • DSPQ dipalmitoylphosphatidylglycerol
  • DSPQ dipalmitoylphosphatidylglycerol
  • DSPQ dipalmitoylphosphatidylglycerol
  • DOPE dioleylphosphatidyl-ethanolarnine
  • PSPC palmitoylstearoylphosphatidyl-choline
  • PSPG palmitoylstearolphosphatidylglycerol
  • MOPE mono-oleoyl-phosphatidylethanolarnine
  • the sterols can include, cholesterol, esters of cholesterol including cholesterol hemi-succinate, salts of cholesterol including cholesterol hydrogen sulfate and cholesterol sulfate, ergosterol, esters of ergosterol including ergosterol hemi-succinate, salts of ergosterol including ergosterol hydrogen sulfate and ergosterol sulfate, lanosterol, esters of lanosterol including lanosterol hemi-succinate, salts of lanosterol including lanosterol hydrogen sulfate and lanosterol sulfate.
  • the tocopherols can include tocopherols, esters of tocopherols including tocopherol hemi-succinates, salts of tocopherols including tocopherol hydrogen sulfates and tocopherol sulfates.
  • the term “sterol compound” includes sterols, tocopherols and the like.
  • the cationic lipids used can include ammonium salts of fatty acids, phospholids and glycerides.
  • the fatty acids include fatty acids of carbon chain lengths of 12 to 26 carbon atoms that are either saturated or unsaturated. Some specific examples include: myristylamine, palmitylamine, laurylamine and stearylamine, dilauroyl ethylphosphocholine (DLEP), dimyristoyl ethylphosphocholine (DMEP), dipalmitoyl ethylphosphocholine (DPEP) and distearoyl ethylphosphocholine (DSEP), N-(2,3-di-(9-(Z)-octadecenyloxy)-prop-1-yl-N,N,N-trimethylammonium chloride (DOTMA) and 1,2-bis(oleoyloxy)-3-(trimethylammonio)propane (DOTAP).
  • DLEP dilauroyl e
  • the negatively-charged lipids which can be used include phosphatidyl-glycerols (PGs), phosphatidic acids (PAs), phosphatidylinositols (Pls) and the phosphatidyl serines (PSs).
  • PGs phosphatidyl-glycerols
  • PAs phosphatidic acids
  • Pls phosphatidylinositols
  • PSs phosphatidyl serines
  • Examples include DMPG, DPPG, DSPG, DMPA, DPPA, DSPA, DMPI, DPPI, DSPI, DMPS, DPPS and DSPS.
  • Phosphatidylcholines such as DPPC aid in the uptake by the cells in the lung (e.g., the alveolar macrophages) and helps to sustain release of the bioactive agent in the lung.
  • the negatively charged lipids such as the PGs, PAs, PSs and PIs, in addition to reducing particle aggregation, are believed to play a role in the sustained release characteristics of the inhalation formulation as well as in the transport of the formulation across the lung (transcytosis) for systemic uptake.
  • the sterol compounds are believed to affect the release characteristics of the formulation.
  • Liposomes are completely closed lipid bilayer membranes containing an entrapped aqueous volume. Liposomes may be unilamellar vesicles (possessing a single membrane bilayer) or multilamellar vesicle (onion-like structures characterized by multiple membrane bilayers, each separated from the next by an aqueous layer).
  • the bilayer is composed of two lipid monolayers having a hydrophobic “tail” region and a hydrophilic “head” region.
  • the structure of the membrane bilayer is such that the hydrophobic (nonpolar) “tails” of the lipid monolayers orient toward the center of the bilayer while the hydrophilic “heads” orient towards the aqueous phase.
  • Liposomes can be produced by a variety of methods (for a review, see, e.g., Cullis et al. (1987)). Bangham's procedure (j. Mol. Biol. (1965)) produces ordinary multilamellar vesicles (MLVs).
  • MUVs multilamellar vesicles
  • Lenk et al. U.S. Pat. Nos. 4,522,803, 5,030,453 and 5,169,637)
  • Fountain et al. U.S. Pat. No. 4,588,578)
  • Cullis et al. U.S. Pat. No. 4,975,282
  • Paphadjopoulos et al., U.S. Pat. No. 4,235,871 discloses preparation of oligolamellar liposomes by reverse phase evaporation.
  • Unilamellar vesicles can be produced from MLVs by a number of techniques, for example, the extrusion of Cullis et al. (U.S. Pat. No. 5,008,050) and Loughrey et al. (U.S. Pat. No. 5,059,421)). Sonication and homogenization an be so used to produce smaller unilamellar liposomes from larger liposomes (see, for example, Paphajopoulos et at. (1968); Deamer and Uster (1983); and Chapman et al. (1968)).
  • the original liposome preparation of Bangham et al. involves suspending phospholipids in an organic solvent which is then evaporated to dryness leaving a phosphoipid film on the reaction vessel. Next, an appropriate amount of aqueous phase is added, the 60 mixture is allowed to “swell”, and the resulting liposomes which consist of multilamellar vesicles (MLVs) are dispersed by mechanical means.
  • MLVs multilamellar vesicles
  • LUVs large unilamellar vesicles
  • vesicles include those that form reverse-phase evaporation vesicles (REV), Papahadjopoulos et al., U.S. Pat. No. 4,235,871.
  • REV reverse-phase evaporation vesicles
  • Another class of liposomes that may be used are those characterized as having substantially equal lamellar solute distribution. This class of liposomes is denominated as stable plurilamellar vesicles (SPLV) as defined in U.S. Pat. No. 4,522,803 to Lenk, et al. and includes monophasic vesicles as described in U.S. Pat. No. 4,588,578 to Fountain, et al. and frozen and thawed multilamellar vesicles (FATMLV) as described above.
  • SPLV stable plurilamellar vesicles
  • FATMLV frozen and thawed multilamellar vesicles
  • sterols and their water soluble derivatives such as cholesterol hemisuccinate have been used to form liposomes; see specifically Janoff et al., U.S. Pat. No. 4,721,612, issued Jan. 26, 1988, entitles “Steroidal Liposomes,” Mayhew et al., PCT Publication No. WO 85/00968, published Mar. 14, 1985, described a method for reducing the toxicity of drugs by encapsulating them in liposomes comprising alpha-tocopherol and certain derivatives thereof. Also, a variety of tocopherols and their water soluble derivatives have been used to form liposomes, see Janoff et al., PCT Publication No. 87/02219, published Apr. 23, 1987, entitled “Alpha Tocopherol-Based Vesicles”.
  • the liposomes are comprised of particles with a mean diameter of approximately 0.01 microns to approximately 3.0 microns, preferably in the range about 0.2 to 1.0 microns.
  • the sustained release property of the liposomal product can be regulated by the nature of the lipid membrane and by inclusion of other excipients (e.g., sterols) in the composition.
  • the infective agent included in the scope of the present invention may be a bacteria.
  • the bacteria can be selected from: Pseudomonas aeruginosia, bacillus anthracis, Listeria monocytogenes, Staphylococcus aureus, Salmenellosis, Yersina pestis, Mycobacterium leprae, M. africanum, M. asiaticum, M. avium - intracellulaire, M. chelonei abscessus, M. fallax, M. fortuitum, M. kansasii, M. leprae, M. malmonense, M. shimoidei, M simiae, M. szulgai, M.
  • the infective agent included in the scope of the present invention can be a virus.
  • the virus can be selected from: hantavirus, respiratory syncytial virus, influenza, and viral pneumonia.
  • the infective agent included in the scope of the present invention can be a fungus.
  • Fungal diseases of note include: aspergillosis, disseminated candidiasis, blastomycosis, coccidioidomycosis, cryptococcosis, histoplasmosis, mucormycosis, and sporotrichosis.
  • antiinfective agent is used throughout the specification to describe a biologically active agent which can kill or inhibit the growth of certain other harmful pathogenic organisms, including but not limited to bacteria, yeasts and fungi, viruses, protozoa or parasities, and which can be administered to living organisms, especially animals such as mammals, particularly humans.
  • antibiotic agents that may be used in the antiinfective compositions of the present invention include cephalosporins, quinolones and fluoroquinolones, penicillins, and beta lactamas inhibitors, carbepenems, monobactams, macrolides and lincosamines, glycopeptides, rifampin, oxazolidonones, tetracyclines, aminoglycosides, streptogramins, sulfonamides, and others. Each family comprises many members.
  • Cephalosporins are further categorized by generation.
  • cephalosporins by generation include the following.
  • cephalosporins I generation include Cefadroxil, Cefazolin, Cephalexin, Cephalothin, Cephaprin, and Cephradine.
  • cephalosporins II generation include Cefaclor, Cefamandol, Cefonicid, Cefotetan, Cefoxitin, Cefproil, Ceftmetazole, Cefuroxime, Cefuroxime axetil, and Loracarbef.
  • cephalosporins III generation examples include Cefdinir, Ceftibuten, Cefditoren, Cefetamet, Cefpodoxime, Cefprozil, Cefuroxime (axetil), Cefuroximer (sodium), Cefoperazone, Cefixime, Cefotaxime, Cefpodoxime proxetil, Ceftazidime, Ceftizoxime, and Ceftriaxone.
  • cephalosporins IV generation examples include Cefepime.
  • Non-limiting examples of quinolones and fluoroquinolones include Cinoxacin, Ciprofloxacin, Enoxacin, Gatifloxacin, Grepafloxacin, Levofloxacin, Lomefloxacin, Moxifloxacin, Nalidixic acid, Norfloxacin, Ofloxacin, Sparfloxacin, Trovafloxacin, Oxolinic acid, Gemifloxacin, and Perfloxacin.
  • Non-limiting examples of penicillins include Amoxicillin, Ampicillin, Bacampicillin, Carbenicillin Indanyl, Mezlocillin, Piperacillin, and Ticarcillin.
  • Non-limiting examples of penicillins and beta lactamase inhibitors include Amoxicillin-Clavulanic Acid, Ampicillin-Sulbactam, Sulfactam, Tazobactam, Benzylpenicillin, Cloxacillin, Dicloxacillin, Methicillin, Oxacillin, Penicillin G (Benzathine, Potassium, Procaine), Penicillin V, Penicillinase-resistant penicillins, Isoxazoylpenicillins, Aminopenicillins, Ureidopenicillins, Piperacillin+Tazobactam, Ticarcillin+Clavulanic Acid, and Nafcillin.
  • Non-limiting examples of carbepenems include Imipenem-Cilastatin and Meropenem.
  • a non-limiting example of a monobactam includes Aztreonam.
  • Non-limiting examples of macrolides and lincosamines include Azithromycin, Clarithromycin, Clindamycin, Dirithromycin, Erythromycin, Lincomycin, and Troleandomycin.
  • glycopeptides include Teicoplanin and Vancomycia.
  • Non-limiting examples of rifampins include Rifabutin, Rifampin, and Rifapentine.
  • Non-limiting example of oxazolidonones includes Linezolid.
  • Non-limiting examples of tetracyclines include Demeclocycline, Doxycycline, Methacycline, Minocycline, Oxytetracycline, Tetracycline, and Chlortetracycline.
  • Non-limiting examples of aminoglycosides include Amikacin, Gentamicin, Kanamycin, Neomycin, Netilmicin, Streptomycin, Tobramycin, and Paromomycin.
  • Non-limiting example of streptogramins includes Quinopristin+Dalfopristin.
  • Non-limiting examples of sulfonamides include Mafenide, Silver Sulfadiazine, Sulfacetamide, Sulfadiazine, Sulfamethoxazole, Sulfasalazine, Sulfisoxazole, Trimethoprim-Sulfamethoxazole, and Sulfamethizole.
  • Non-limiting examples of other antibiotic agents include Bacitracin, Chloramphenicol, Colistemetate, Fosfomycin, Isoniazid, Methenamine, Metronidazol, Mupirocin, Nitroflurantoin, Nitrofurazone, Novobiocin, Polymyxin B, Spectinomycin, Trimethoprine, Trimethoprine/Sulfamethoxazole, Cationic peptides, Colistin, Iseganan, Cycloserine, Capreomycin,Pyrazinamide, Para-aminosalicyclic acid, and Erythromycin ethylsuccinate+sulfisoxazole.
  • Antiviral agents include, but are not limited to: zidovudine, acyclovir, ganciclovir, vidarabine, idoxuridine, trifluridine, ribavirin, interferon alpha-2a, interferon alpha-2b, interferon beta, interferon gamma).
  • Antifungal agents include, but are not limited to: amphotericin B, nystatin, hamycin, natamycin, pimaricin, ambruticin, itraconazole, terconazole, ketoconazole, voriconazole, miconazole, nikkomycin Z, griseofulvin, candicidin, cilofungin, chlotrimazole, clioquinol, caspufungin, tolnaflate.
  • any compositions of the present invention will vary depending on the symptoms, age and body weight of the patient, the nature and severity of the disorder to be treated or prevented, the route of administration, and the form of the subject composition. Any of the subject formulations may be administered in a single dose or in divided doses. Dosages for the compositions of the present invention may be readily determined by techniques known to those of skill in the art or as taught herein.
  • the dosage of the subject compounds will generally be in the range of about 0.01 ng to about 10 g per kg body weight, specifically in the range of about 1 ng to about 0.1 g per kg, and more specifically in the range of about 100 ng to about 10 mg per kg.
  • An effective dose or amount, and any possible affects on the timing of administration of the formulation may need to be identified for any particular composition of the present invention. This may be accomplished by routine experiment as described herein, using one or more groups of animals (preferably at least 5 animals per group), or in human trials if appropriate.
  • the effectiveness of any subject composition and method of treatment or prevention may be assessed by administering the composition and assessing the effect of the administration by measuring one or more applicable indices, and comparing the post-treatment values of these indices to the values of the same indices prior to treatment.
  • the precise time of administration and amount of any particular subject composition that will yield the most effective treatment in a given patient will depend upon the activity, pharmacokinetics, and bioavailability of a subject composition, physiological condition of the patient (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage and type of medication), route of administration, and the like.
  • the guidelines presented herein may be used to optimize the treatment, e.g., determining the optimum time and/or amount of administration, which will require no more than routine experimentation consisting of monitoring the subject and adjusting the dosage and/or timing.
  • the health of the patient may be monitored by measuring one or more of the relevant indices at predetermined times during the treatment period.
  • Treatment including composition, amounts, times of administration and formulation, may be optimized according to the results of such monitoring.
  • the patient may be periodically reevaluated to determine the extent of improvement by measuring the same parameters. Adjustments to the amount(s) of subject composition administered and possibly to the time of administration may be made based on these reevaluations.
  • Treatment may be initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage may be increased by small increments until the optimum therapeutic effect is attained.
  • compositions may reduce the required dosage for any individual agent contained in the compositions (e.g., the Fabl inhibitor) because the onset and duration of effect of the different agents may be complimentary.
  • Toxicity and therapeutic efficacy of subject compositions may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 and the ED 50 .
  • the data obtained from the cell culture assays and animal studies may be used in formulating a range of dosage for use in humans.
  • the dosage of any subject composition lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose may be estimated initially from cell culture assays.
  • the pharmaceutical formulation of the antiinfective may be comprised of either an aqueous dispersion of liposomes and free antiinfective, or a dehydrated powder containing liposomes and free antiinfective.
  • the formulation may contain lipid excipients to form the liposomes, and salts/buffers to provide the appropriate osmolarity and pH.
  • the dry powder formulations may contain additional excipients to prevent the leakage of encapsulated antiinfective during the drying and potential milling steps needed to create a suitable particle size for inhalation (i.e., 1-5 ⁇ m). Such excipients are designed to increase the glass transition temperature of the antiinfective formulation.
  • the pharmaceutical excipient may be a liquid or solid filler, diluent, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof from one organ or portion of the body, to another organ, or portion of the body.
  • Each excipient must be “acceptable” in the sense of being compatible with the subject composition and its components and not injurious to the patient.
  • Suitable excipients include trehalose, raffinose, mannitol, sucrose, leucine, trileucine, and calcium chloride.
  • excipients examples include (1) sugars, such as lactose, and glucose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulos and cellulos acetate; 4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (10)
  • the pharmaceutical formulations of the present invention may be used in any dosage dispensing device adapted for intranasal administration.
  • the device should be constructed with a view to ascertaining optimum metering accuracy and compatibility of its constructive elements, such as container, valve and actuator with the nasal formulation and could be based on a mechanical pump system, e.g., that of a metered-dose nebulizer, dry powder inhaler, soft mist inhaler, or a nebulizer.
  • Suitable devices include jet nebulizers (e.g., PARI LC Star, AKITA), soft mist inhalers (e.g., PARI e-Flow), and capsule-based dry powder inhalers (e.g., PH&T Turbospin).
  • Suitable propellants may be selected among such gases as fluorocarbons, hydrocarbons, nitrogen and dinitrogen oxide or mixtures thereof.
  • the inhalation delivery device can be a nebulizer or a metered dose inhaler (MDI), or any other suitable inhalation delivery device known to one or ordinary skill in the art.
  • the device can contain and be used to deliver a single dose of the antiinfective compositions or the device can contain and be used to deliver multi-doses of the compositions of the present invention.
  • a nebulizer type inhalation delivery device can contain the compositions of the present invention as a solution, usually aqueous, or a suspension.
  • the nebulizer type delivery device may be driven ultrasonically, by compressed air, by other gases, electronically or mechanically.
  • the ultrasonic nebulizer device usually works by imposing a rapidly oscillating waveform onto the liquid film of the formulation via an electrochemical vibrating surface. At a given amplitude the waveform becomes unstable, whereby it disintegrates the liquids film, and it produces small droplets of the formulation.
  • the nebulizer device driven by air or other gases operates on the basis that a high pressure gas stream produces a local pressure drop that draws the liquid formulation into the stream of gases via capillary action. This fine liquid stream is then disintegrated by shear forces.
  • the nebulizer may be portable and hand held in design, and may be equipped with a self contained electrical unit.
  • the nebulizer device may comprise a nozzle that has two coincident outlet channels of defined aperture size through which the liquid formulation can be accelerated. This results in impaction of the two streams and atomization of the formulation.
  • the nebulizer may use a mechanical actuator to force the liquid formulation through a multiorifice nozzle of defined aperture size(s) to produce an aerosol of the formulation for inhalation.
  • blister packs containing single doses of the formulations may be employed.
  • the nebulizer may be employed to ensure the sizing of particles is optimal for positioning of the particle within, for example, the pulmonary membrane.
  • a metered dose inhalator may be employed as the inhalation delivery device for the compositions of the present invention.
  • This device is pressurized (pMDI) and its basic structure comprises a metering valve, an actuator and a container.
  • a propellant is used to discharge the formulation from the device.
  • the composition may consist of particles of a defined size suspended in the pressurized propellant(s) liquid, or the composition can be in a solution or suspension of pressurized liquid propellant(s).
  • the propellants used are primarily atmospheric friendly hydroflourocarbons (HFCs) such as 134a and 227. Traditional chloroflourocarbons like CFC-11, 12 and 114 are used only when essential.
  • the device of the inhalation system may deliver a single dose via, e.g., a blister pack, or it may be multi dose in design.
  • the pressurized metered dose inhalator of the inhalation system can be breath actuated to deliver an accurate dose of the lipid-containing formulation.
  • the delivery of the formulation may be programmed via a microprocessor to occur at a certain point in the inhalation cycle.
  • the MDI may be portable and hand held.
  • FIG. 1 depicts the lung concentration of amikacin and TOBI® (administered 100% free), based on pharmacokinetic modeling of serum concentrations over time. Both curves assume a volume of distribution for aminoglycosides in the lung of 200 ml.
  • the peak levels of antiinfective in the lung are approximately equivalent for the 100 mg dose of liposomal amikacin, and the 300 mg dose of TOBI®. This is a consequence of the rapid clearance of the free tobramycin from the lung by absorption into the systemic circulation with a half-life of about 1.5 hr.
  • These results serve as a demonstration of the improved lung targeting afforded by liposomal encapsulation
  • the presence of free and encapsulated antiinfective in the amikacin formulation is demonstrated by the two component pharmacokinetic profile observed. Free amikacin is rapidly absorbed into the systemic circulation (with a half-life similar to TOBI), while the encapsulated drug has a lung half-life of approximately 45 hr.
  • the free amikacin is available to provide bactericidal activity, while the encapsulated drug provides sustained levels of drug in the lung, enabling improved killing of resistant bacterial strains.
  • the measured lung concentrations for the liposomal compartment are significantly above the MIC 50 of 1240 clinical isolates of Pseudomonas aeruginosa, potentially reducing the development of resistance.
  • Liposomal preparations of amikacin may exhibit significant leakage of encapsulated drug during nebulization.
  • Table 1 the presence of free amikacin in solution was shown to surprisingly decrease the leakage of antiinfective by about four-fold from the liposome. While not wishing to be limited to any particular theory, it is hypothesized that liposomes break-up and re-form during nebulization, losing encapsulated antiinfective in the process. Alternatively, encapsulated antiinfective is lost during nebulization because the liposome membrane becomes leaky. When an excess of free antiinfective is present in solution, the free antiinfective is readily available in close proximity to the liposome, and is available to be taken back up into the liposome on re-formation.

Abstract

A system for treating or providing prophylaxus against a pulmonary infection is disclosed comprising: a) a pharmaceutical formulation comprising a mixture of free antiinfective and antiinfective encapsulated in a lipid-based composition, and b) an inhalation delivery device. A method for providing prophylaxis against a pulmonary infection in a patient and a method of reducing the loss of antiinfective encapsulated in a lipid-based composition upon nebulization comprising administering an aerosolized pharmaceutical formulation comprising a mixture of free antiinfective and antiinfective encapsulated in a lipid-based composition is also disclosed.

Description

    RELATED APPLICATIONS
  • This application claims the benefit of priority to U.S. Provisional Patent Application Ser. No. 60/748,468, filed Dec. 8, 2005, which is hereby incorporated by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • According to the World Health Organization, respiratory diseases are the number one cause of world-wide mortality, with at least 20% of the world's population afflicted. Over 25 million Americans have chronic lung disease, making it the number one disabler of American workers (<$50B/yr), and the number three cause of mortality.
  • Currently, most infections are treated with oral or injectable antiinfectives, even when the pathogen enters through the respiratory tract. Often the antiinfective has poor penetration into the lung, and may be dose-limited due to systemic side-effects. Many of these issues can be overcome by local delivery of the antiinfective to the lungs of patients via inhalation. For example, inhaled tobramycin (TOBI®, Chiron Corp, Emeryville, Calif.), is a nebulized form of tobramycin, that has been shown to have improved efficacy and reduced nephro- and oto-toxicity relative to injectable aminoglycosides. Unfortunately, rapid absorption of the drug necessitates that the drug product be administered twice daily over a period of ca., 20 min per administration. For pediatrics and young adults with cystic fibrosis this treatment regimen can be taxing, especially when one takes into account the fact that these patients are on multiple time-consuming therapies. Any savings in terms of treatment times would be welcomed, and would likely lead to improvements in patient compliance. Achieving improved compliance with other patient populations (e.g., chronic obstructive pulmonary disease (COPD), acute bronchial exacerbations of chronic bronchitis) will be critically dependent on the convenience and efficacy of the treatment. Hence, it is an object of the present invention to improve patient compliance by providing formulations with sustained activity in the lungs. Sustained release formulations of antiinfectives are achieved by encapsulating the antiinfective in a liposome. Improving pulmonary targeting with sustained release formulations would further improve the therapeutic index by increasing local concentrations of drug and reducing systemic exposure. Improvements in targeting are also expected to reduce dose requirements.
  • Achieving sustained release of drugs in the lung is a difficult task, given the multiple clearance mechanisms that act in concert to rapidly remove inhaled drugs from the lung. These clearance methods include: (a) rapid clearance from the conducing airways over a period of hours by the mucociliary escalator, (b) clearance of particulates from the deep lung by alveolar macrophages; (c) degradation of the therapeutic by pulmonary enzymes and; (d) rapid absorption of small molecule drugs into the systemic circulation. Absorption of small molecule drugs has been shown to be nearly quantitative, with an absorption time for hydrophilic small molecules of about 1 hr, and an absorption time for lipophilic drugs of about 1 min.
  • For TOBI® the absorption half-life from the lung is on the order of 1.5 hr. High initial peak concentrations of drug can lead to adaptive resistance, while a substantial time with levels below or near the effective minimum inhibitory concentration (MIC), may lead to selection of resistant phenotypes. It is hypothesized that keeping the level of antiinfective above the MIC for an extended period of time (i.e., eliminating sub-therapeutic trough levels) with a pulmonary sustained release formulation may reduce the potential for development of resistant phenotypes. Hence, it is a further object of the present invention to maintain the ratio of the area under the lung concentration/time curve to the MIC at 24 hr (i.e., the AUIC), not only at an adequate sustained therapeutic level, but above a critical level, so as to reduce the potential for selection of resistant strains.
  • It is assumed that only the “free” (un-encapsulated) drug has bactericidal activity. One potential disadvantage of liposomal sustained release formulations is that the encapsulation of drug in the liposomal formulation decreases the concentration of free drug reaching the lung pathogens, drug which is needed to achieve efficient killing of bacteria immediately following administration. Hence, it is further an object of the present invention to provide a formulation that contains sufficient free drug, to be bactericidal immediately following administration.
  • The disclosures of the foregoing are incorporated herein by reference in their entirety.
  • SUMMARY OF THE INVENTION
  • It is an object of the present invention to use lipid-based composition encapsulation to improve the therapeutic effects of antiinfectives administered to an individual via the pulmonary route.
  • The subject invention results from the realization that administering a pharmaceutical composition comprising both free and liposome encapsulated antiinfective results in improved treatment of pulmonary infections.
  • In one aspect, the present invention relates to a system for treating or providing prophylaxis against a pulmonary infection, wherein the system comprises a pharmaceutical formulation comprising mixtures of free and lipid-based composition encapsulated antiinfective, wherein the amount of free antiinfective is sufficient to provide for immediate bactericidal activity, and the amount of encapsulated antiinfective is sufficient to provide sustained bactericidal activity, and reduce the development of resistant strains of the infectious agent, and an inhalation delivery device.
  • The free form of the antiinfective is available to provide a bolus of immediate antimicrobial activity. The slow release of antiinfective from the lipid-based composition following pulmonary administration is analogous to continuous administration of the antiinfective, thereby providing for sustained levels of antiinfective in the lungs. The sustained AUC levels provides prolonged bactericidal activity between administration. Further, the sustained levels provided by the release of antiinfective from the lipid-based composition is expected to provide improved protection against the development of resistant microbial strains.
  • Combinations of free and encapsulated drug can be achieved by: (a) formulation of mixtures of free and encapsulated drug that are stable to the nebulization; (b) formulation of encapsulated drug which leads to burst on nebulization.
  • The ratio of free to encapsulated drug is contemplated to be between about 1:100 w:w and about 100:1 w:w, and may be determined by the minimum inhibitory concentration of the infectious agent and the sustained release properties of the formulation. The ratio of free to encapsulated drug can be optimized for a given infectious agent and drug formulation based on known pharmacodynamic targets for bacterial killing and prevention of resistance. Schentag, J. J. J Chemother. 1999, 11, 426-439.
  • In a further embodiment, the present invention relates to the aforementioned system wherein the antiinfective is selected from the group consisting of antibiotic agents, antiviral agents, and antifungal agents. In a further embodiment, the antiinfective is an antibiotic selected from the group consisting of cephalosporins, quinolones, fluoroquinolones, penicillins, beta lactamase inhibitors, carbepenems, monobactams, macrolides, lincosamines, glycopeptides, rifampin, oxazolidonones, tetracyclines, aminoglycosides, streptogramins, and sulfonamides. In a further embodiment, the antiinfective is an aminoglycoside. In a further embodiment the antiinfective is amikacin, gentamicin, or tobramycin.
  • In a further embodiment, the lipid-based composition is a liposome. In a further embodiment, the liposome comprises a mixture of unilamellar vesicles and multilamellar vesicles. In a further embodiment, the liposome comprises a phospholipid and a sterol. In a further embodiment, the liposome comprises a phosphatidylcholine and a sterol. In a further embodiment, the liposome comprises dipalmitoylphosphatidylcholine (DPPC) and a sterol. In a further embodiment, the liposome comprises dipalmitoylphosphatidylcholine (DPPC) and cholesterol.
  • In a further embodiment, the present relates to the aforementioned system wherein the antiinfective is an aminogylcoside and the liposome comprises DPPC and cholesterol. In a further embodiment, the antiinfective is amikacin, the liposome comprises DPPC and cholesterol, and the liposome comprises a mixture of unilamellar vesicles and multilamellar vesicles.
  • In a further embodiment, the present invention relates to the aforementioned system, wherein the ratio by weight of free antiinfective to antiinfective encapsulated in a lipid-based composition is between about 1:100 an about 100:1. In a further embodiment, the ratio by weight is between about 1:10 and about 10:1. In a further embodiment, the ratio by weight is between about 1:2 and about 2:1.
  • In another embodiment, the present invention relates to a method for treating or providing prophylaxis against a pulmonary infection in a patient, the method comprising: administering an aerosolized pharmaceutical formulation comprising the antiinfective to the lungs of the patient, wherein the pharmaceutical formulation comprises mixtures of free and lipid-based composition encapsulated antiinfectives, and the amount of free antiinfective is sufficient to provide for bactericidal activity, and the amount of encapsulated antiinfective is sufficient to reduce the development of resistant strains of the infectious agent.
  • In a further embodiment, the aforementioned method comprises first determining the minimum inhibitory concentration (MIC) of an antiinfective for inhibiting pulmonary infections, and wherein the amount of free antiinfective is at least 2 times the MIC, preferably greater than 4 times the MIC, and preferably greater than 10 times the MIC of the antiinfective agent, where the MIC is defined as either the minimum inhibitory concentration in the epithelial lining of the lung, or alternatively the minimum inhibitory concentration in the solid tissue of the lung (depending on the nature of the infection).
  • In a further embodiment, the present invention relates to the aforementioned method, wherein the aerosolized pharmaceutical formulation is administered at least once per week. In a further embodiment, the present invention relates to the aforementioned method, wherein the antiinfective is selected from the group consisting of antibiotic agents, antiviral agents, and antifungal agents. In a further embodiment, the antiinfective is an antibiotic selected from the group consisting of cephalosporins quinolones, fluoroquinolones, penicillins, beta lactamase inhibitors, carbepenems, monobactams, macrolides, lincosamines, glycopeptides, rifampin, oxazolidonones, tetracyclines, aminoglycosides, streptogramins, and sulfonamides. In a further embodiment, the antiinfective is an aminoglycoside. In a further embodiment, the antiinfective is amikacin, gentamicin, or tobramycin.
  • In a further embodiment, the lipid-based composition is a liposome. In a further embodiment, the liposome encapsulated antiinfective comprises a phosphatidylcholine in admixture with a sterol. In a further aspect, the sterol is cholesterol. In a further aspect, the liposome encapsulated antiinfective comprises a mixture of unilamellar vesicles and multilamellar vesicles. In a further aspect, the liposome encapsulated antiinfective comprises a phosphatidytcholine in admixture with cholesterol, and wherein the liposome encapsulated antiinfective comprises a mixture of unilamellar vesicles and multilamellar vesicles.
  • The ratio of the area under the lung concentration/time curve to the MIC at 24 hr (i.e., the AUIC) is greater than 25, preferably greater than 100, and preferably greater than 250.
  • The therapeutic ratio of free/encapsulated drug and the required nominal dose can be determined with standard pharmacokinetic models, once the efficiency of pulmonary delivery and clearance of the drug product are established with the aerosol delivery device of choice.
  • In one aspect, the present invention relates to a method of treating a patient for a pulmonary infection comprising a cycle of treatment with lipid-based composition encapsulated antiinfective to enhance bacterial killing and reduce development of phenotypic resistance, following by a cycle of no treatment to reduce the development of adaptive resistance. The treatment regimen may be determined by clinical research. In one embodiment, the treatment regime may be an on-cycle treatment for about 7, 14, 21, or 30 days, followed by an off-cycle absence of treatment for about 7, 14, 21, or 30 days.
  • In another aspect, the present invention relates to a method for reducing the loss of antiinfective encapsulated in lipid-based compositions upon nebulization comprising administering the antiinfective encapsulated in lipid-based compositions with free antiinfective.
  • The systems and methods of the present invention are useful for treating, for example, lung infections in cystic fibrosis patients, chronic obstructive pulmonary disease (COPD), bronchiectasis, acterial pneumonia, and in acute bronchial exacerbations of chronic bronchitis (ABECB). In addition, the technology is useful in the treatment of intracellular infections including Mycobacterium tuberculosis, and inhaled agents of bioterror (e.g., anthrax and tularemia). The technology may also be used as a phophylactic agent to treat opportunistic fungal infections (e.g., aspergillosis) in immunocompromised patients (e.g., organ transplant or AIDS patients).
  • With bacteria and other infective agents becoming increasingly resistant to traditional treatments, new and more effective treatments for infective agent related illnesses are needed. The present invention addresses these issues by providing a system comprising a pharmaceutical composition comprising both free and lipid-based composition encapsulated antiinfective and an inhalation device. Formulating the antiinfective as a mixture of free and lipid-based composition encapsulated antiinfective provides several advantages, some of which include: (a) provides for a bolus of free antiinfective for immediate bactericidal activity and a sustained level of antiinfective for prevention of resistance; (b) simplifies the manufacturing process, as less free antiinfective need be removed via diafiltration; and (c) allows for greater antiinfective contents to be achieved in the drug product.
  • These embodiments of the present invention, other embodiments, and their features and characteristics, will be apparent from the description, drawings and claims that follow.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts the plot of lung concentration (μg/ml) as a function of time following nebulization of unencapsulated tobramycin at a nominal dose of 300 mg (TOBI®, Chiron Corp., Emeryville, Calif.), and liposomal amikacin at a nominal dose of 100 mg. Lung concentrations for both drug products are calculated assuming a volume of distribution for aminoglycosides in the lung of 200 ml. The tobramycin curve was determined by pharmacokinetic modeling of the temporal tobramycin plasma concentration curve (Le Brun thesis, 2001).
  • DETAILED DESCRIPTION OF THE INVENTION
  • Definitions
  • For convenience, before further description of the present invention, certain terms employed in the specification, examples and appended claims are collected here. These definitions should be read in light of the remainder of the disclosure and understood as by a person of skill in the art. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art.
  • The articles “a” and “an” are used herein to refer to one or to more than one (i.e. to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.
  • The term “antibacterial” is are-recognized and refers to the ability of the compounds of the present invention to prevent, inhibit or destroy the growth of microbes of bacteria.
  • The terms “antiinfective” and “antiinfective agent” are used interchangeably throughout the specification to describe a biologically active agent which can kill or inhibit the growth of certain other harmful pathogenic organisms, including but not limited to bacteria, yeasts and fungi, viruses, protozoa or parasites, and which can be administered to living organisms, especially animals such as mammals, particularly humans.
  • The term “antimicrobial” is are-recognized and refers to the ability of the compounds of the present invention to prevent, inhibit or destroy the growth of microbes such as bacteria, fungi, protozoa and viruses.
  • The term “bioavailable” is art-recognized and refers to a form of the subject invention that allows for it, or a portion of the amount administered, to be absorbed by, incorporated to, or otherwise physiologically available to a subject or patient to whom it is administered.
  • The terms “comprise” and “comprising” are used in the inclusive, open sense, meaning that additional elements may be included.
  • The term “illness” as used herein refers to any illness caused by or related to infection by an organism.
  • The term “including” is used herein to mean “including but not limited to”. “Including” and “including but not limited to” are used interchangeably.
  • The term “lipid-based composition” as used herein refers to compositions that primarily comprise lipids. Non-limiting examples of lipid-based compositions may take the form of coated lipid particles, liposomes, emulsions, micelles, and the like.
  • The term “mammal” is known in the art, and exemplary mammals include humans, primates, bovines, porcines, canines, felines, and rodents (e.g., mice and rats).
  • The term “microbe” is art-recognized and refers to a microscopic organism. In certain embodiments the term microbe is applied to bacteria. In other embodiments the term refers to pathogenic forms of a microscopic organism.
  • A “patient,” “subject” or “host” to be treated by the subject method may mean either a human or non-human animal.
  • The term “pharmaceutically-acceptable salts” is art-recognized and refers to the relatively non-toxic, inorganic and organic acid addition salts of compounds, including, for example, those contained in compositions of the present invention.
  • The term “prodrug” is art-recognized and is intended to encompass compounds which, under physiological conditions, are converted into the antibacterial agents of the present invention. A common method for making a prodrug is to select moieties which are hydrolyzed under physiological conditions to provide the desired compound. In other embodiments, the prodrug is converted by an enzymatic activity of the host animal or the target bacteria.
  • The term “treating” is are-recognized and refers to curing as well as anneliorating at least one symptom of any condition or disease.
  • Lipids
  • The lipids used in the pharmaceutical formulations of the present invention can be synthetic, semi-synthetic or naturally-occurring lipids, including phospholipids, tocopherols, sterols, fatty acids, glycoproteins such as albumin, negatively-charged lipids and cationic lipids. In terms of phosholipids, they could include such lipids as egg phosphatidylcholine (EPC), egg phosphatidylglycerol (EPG), egg phosphatidylinositol (EPI), egg phosphatidylserine (EPS), phosphatidylethanolamine (EPE), and phosphatidic acid (EPA); the soya counterparts, soy phosphatidylcholine (SPC); SPG, SPS, SPI, SPE, and SPA; the hydrogenated egg and soya counterparts (e.g., HEPC, HSPC), other phospholipids made up of ester linkages of fatty acids in the 2 and 3 of glycerol positions containing chains of 12 to 26 carbon atoms and different head groups in the I position of glycerol that include choline, glycerol, inositol, serine, ethanolamine, as well as the corresponding phosphatidic acids. The chains on these fatty acids can be saturated or unsaturated, and the phospholipid may be made up of fatty acids of different chain lengths and different degrees of unsaturation. In particular, the compositions of the formulations can include dipalmitoylphosphatidylcholine (DPPC), a major constituent of naturally-occurring lung surfactant. Other examples include dimyristoylphosphatidycholine (DMPC) and dimyristoylphosphatidylglycerol (DMPG) dipalmitoylphosphatideholine (DPPQ) and dipalmitoylphosphatidylglycerol (DPPG) distearoylphosphatidylcholine (DSPQ) and distearoylphosphatidylglycerol (DSPG), dioleylphosphatidyl-ethanolarnine (DOPE) and mixed phospholipids like palmitoylstearoylphosphatidyl-choline (PSPC) and palmitoylstearolphosphatidylglycerol (PSPG), and single acylated phospholipids like mono-oleoyl-phosphatidylethanolarnine (MOPE).
  • The sterols can include, cholesterol, esters of cholesterol including cholesterol hemi-succinate, salts of cholesterol including cholesterol hydrogen sulfate and cholesterol sulfate, ergosterol, esters of ergosterol including ergosterol hemi-succinate, salts of ergosterol including ergosterol hydrogen sulfate and ergosterol sulfate, lanosterol, esters of lanosterol including lanosterol hemi-succinate, salts of lanosterol including lanosterol hydrogen sulfate and lanosterol sulfate. The tocopherols can include tocopherols, esters of tocopherols including tocopherol hemi-succinates, salts of tocopherols including tocopherol hydrogen sulfates and tocopherol sulfates. The term “sterol compound” includes sterols, tocopherols and the like.
  • The cationic lipids used can include ammonium salts of fatty acids, phospholids and glycerides. The fatty acids include fatty acids of carbon chain lengths of 12 to 26 carbon atoms that are either saturated or unsaturated. Some specific examples include: myristylamine, palmitylamine, laurylamine and stearylamine, dilauroyl ethylphosphocholine (DLEP), dimyristoyl ethylphosphocholine (DMEP), dipalmitoyl ethylphosphocholine (DPEP) and distearoyl ethylphosphocholine (DSEP), N-(2,3-di-(9-(Z)-octadecenyloxy)-prop-1-yl-N,N,N-trimethylammonium chloride (DOTMA) and 1,2-bis(oleoyloxy)-3-(trimethylammonio)propane (DOTAP).
  • The negatively-charged lipids which can be used include phosphatidyl-glycerols (PGs), phosphatidic acids (PAs), phosphatidylinositols (Pls) and the phosphatidyl serines (PSs). Examples include DMPG, DPPG, DSPG, DMPA, DPPA, DSPA, DMPI, DPPI, DSPI, DMPS, DPPS and DSPS.
  • Phosphatidylcholines, such as DPPC, aid in the uptake by the cells in the lung (e.g., the alveolar macrophages) and helps to sustain release of the bioactive agent in the lung. The negatively charged lipids such as the PGs, PAs, PSs and PIs, in addition to reducing particle aggregation, are believed to play a role in the sustained release characteristics of the inhalation formulation as well as in the transport of the formulation across the lung (transcytosis) for systemic uptake. The sterol compounds are believed to affect the release characteristics of the formulation.
  • Liposomes
  • Liposomes are completely closed lipid bilayer membranes containing an entrapped aqueous volume. Liposomes may be unilamellar vesicles (possessing a single membrane bilayer) or multilamellar vesicle (onion-like structures characterized by multiple membrane bilayers, each separated from the next by an aqueous layer). The bilayer is composed of two lipid monolayers having a hydrophobic “tail” region and a hydrophilic “head” region. The structure of the membrane bilayer is such that the hydrophobic (nonpolar) “tails” of the lipid monolayers orient toward the center of the bilayer while the hydrophilic “heads” orient towards the aqueous phase.
  • Liposomes can be produced by a variety of methods (for a review, see, e.g., Cullis et al. (1987)). Bangham's procedure (j. Mol. Biol. (1965)) produces ordinary multilamellar vesicles (MLVs). Lenk et al. (U.S. Pat. Nos. 4,522,803, 5,030,453 and 5,169,637), Fountain et al. (U.S. Pat. No. 4,588,578) and Cullis et al. (U.S. Pat. No. 4,975,282) disclose methods for producing multilamellar liposomes having substantially equal interlamellar solute distribution in each of their aqueous compartments. Paphadjopoulos et al., U.S. Pat. No. 4,235,871, discloses preparation of oligolamellar liposomes by reverse phase evaporation.
  • Unilamellar vesicles can be produced from MLVs by a number of techniques, for example, the extrusion of Cullis et al. (U.S. Pat. No. 5,008,050) and Loughrey et al. (U.S. Pat. No. 5,059,421)). Sonication and homogenization an be so used to produce smaller unilamellar liposomes from larger liposomes (see, for example, Paphajopoulos et at. (1968); Deamer and Uster (1983); and Chapman et al. (1968)).
  • The original liposome preparation of Bangham et al. (J. Mol. Biol, 1965, 13:238-252) involves suspending phospholipids in an organic solvent which is then evaporated to dryness leaving a phosphoipid film on the reaction vessel. Next, an appropriate amount of aqueous phase is added, the 60 mixture is allowed to “swell”, and the resulting liposomes which consist of multilamellar vesicles (MLVs) are dispersed by mechanical means. This preparation provides the basis for the development of the small sonicated unilamellar vesicles described by Papahadjopoulos et al. (Biochim. Biophys, Acta., 1967, 135:624-638), and large unilamellar vesicles.
  • Techniques for producing large unilamellar vesicles (LUVs), such as, reverse phase evaporation, infusion procedures, and detergent dilution, can be used to produce liposomes. A review of these and other methods for producing liposomes may be found in the text Liposomes. Marc Ostro, ed., Marcel Dekker, Inc., New York, 1983, Chapter 1, the pertinent portions of which are incorporated herein by reference. See also Szoka, Jr. et al., (1980, ann. Rev. Biophys. Bioeng., 9:467), the pertinent portions of which are also incorporated herein by reference.
  • Other techniques that are used to prepare vesicles include those that form reverse-phase evaporation vesicles (REV), Papahadjopoulos et al., U.S. Pat. No. 4,235,871. Another class of liposomes that may be used are those characterized as having substantially equal lamellar solute distribution. This class of liposomes is denominated as stable plurilamellar vesicles (SPLV) as defined in U.S. Pat. No. 4,522,803 to Lenk, et al. and includes monophasic vesicles as described in U.S. Pat. No. 4,588,578 to Fountain, et al. and frozen and thawed multilamellar vesicles (FATMLV) as described above.
  • A variety of sterols and their water soluble derivatives such as cholesterol hemisuccinate have been used to form liposomes; see specifically Janoff et al., U.S. Pat. No. 4,721,612, issued Jan. 26, 1988, entitles “Steroidal Liposomes,” Mayhew et al., PCT Publication No. WO 85/00968, published Mar. 14, 1985, described a method for reducing the toxicity of drugs by encapsulating them in liposomes comprising alpha-tocopherol and certain derivatives thereof. Also, a variety of tocopherols and their water soluble derivatives have been used to form liposomes, see Janoff et al., PCT Publication No. 87/02219, published Apr. 23, 1987, entitled “Alpha Tocopherol-Based Vesicles”.
  • The liposomes are comprised of particles with a mean diameter of approximately 0.01 microns to approximately 3.0 microns, preferably in the range about 0.2 to 1.0 microns. The sustained release property of the liposomal product can be regulated by the nature of the lipid membrane and by inclusion of other excipients (e.g., sterols) in the composition.
  • Infective Agent
  • The infective agent included in the scope of the present invention may be a bacteria. The bacteria can be selected from: Pseudomonas aeruginosia, bacillus anthracis, Listeria monocytogenes, Staphylococcus aureus, Salmenellosis, Yersina pestis, Mycobacterium leprae, M. africanum, M. asiaticum, M. avium-intracellulaire, M. chelonei abscessus, M. fallax, M. fortuitum, M. kansasii, M. leprae, M. malmonense, M. shimoidei, M simiae, M. szulgai, M. xenopi, M. tuberculosis, Brucella melitensis, Brucella suis, Brucella abortus, Brucella canis, Legionella pneumonophilia, Francisella tularensis, Pneumocystis carinii, mycoplasma, and Burkholderia cepacia.
  • The infective agent included in the scope of the present invention can be a virus. The virus can be selected from: hantavirus, respiratory syncytial virus, influenza, and viral pneumonia.
  • The infective agent included in the scope of the present invention can be a fungus. Fungal diseases of note include: aspergillosis, disseminated candidiasis, blastomycosis, coccidioidomycosis, cryptococcosis, histoplasmosis, mucormycosis, and sporotrichosis.
  • Antiinfectives
  • The term antiinfective agent is used throughout the specification to describe a biologically active agent which can kill or inhibit the growth of certain other harmful pathogenic organisms, including but not limited to bacteria, yeasts and fungi, viruses, protozoa or parasities, and which can be administered to living organisms, especially animals such as mammals, particularly humans.
  • Non-limiting examples of antibiotic agents that may be used in the antiinfective compositions of the present invention include cephalosporins, quinolones and fluoroquinolones, penicillins, and beta lactamas inhibitors, carbepenems, monobactams, macrolides and lincosamines, glycopeptides, rifampin, oxazolidonones, tetracyclines, aminoglycosides, streptogramins, sulfonamides, and others. Each family comprises many members.
  • Cephalosporins
  • Cephalosporins are further categorized by generation. Non-limiting examples of cephalosporins by generation include the following. Examples of cephalosporins I generation include Cefadroxil, Cefazolin, Cephalexin, Cephalothin, Cephaprin, and Cephradine. Examples of cephalosporins II generation include Cefaclor, Cefamandol, Cefonicid, Cefotetan, Cefoxitin, Cefproil, Ceftmetazole, Cefuroxime, Cefuroxime axetil, and Loracarbef. Examples of cephalosporins III generation include Cefdinir, Ceftibuten, Cefditoren, Cefetamet, Cefpodoxime, Cefprozil, Cefuroxime (axetil), Cefuroximer (sodium), Cefoperazone, Cefixime, Cefotaxime, Cefpodoxime proxetil, Ceftazidime, Ceftizoxime, and Ceftriaxone. Examples of cephalosporins IV generation include Cefepime.
  • Quinolones and Fluoroquinolones
  • Non-limiting examples of quinolones and fluoroquinolones include Cinoxacin, Ciprofloxacin, Enoxacin, Gatifloxacin, Grepafloxacin, Levofloxacin, Lomefloxacin, Moxifloxacin, Nalidixic acid, Norfloxacin, Ofloxacin, Sparfloxacin, Trovafloxacin, Oxolinic acid, Gemifloxacin, and Perfloxacin.
  • Penicillins
  • Non-limiting examples of penicillins include Amoxicillin, Ampicillin, Bacampicillin, Carbenicillin Indanyl, Mezlocillin, Piperacillin, and Ticarcillin.
  • Pinicillins and Beta Lactamase Inhibitors
  • Non-limiting examples of penicillins and beta lactamase inhibitors include Amoxicillin-Clavulanic Acid, Ampicillin-Sulbactam, Sulfactam, Tazobactam, Benzylpenicillin, Cloxacillin, Dicloxacillin, Methicillin, Oxacillin, Penicillin G (Benzathine, Potassium, Procaine), Penicillin V, Penicillinase-resistant penicillins, Isoxazoylpenicillins, Aminopenicillins, Ureidopenicillins, Piperacillin+Tazobactam, Ticarcillin+Clavulanic Acid, and Nafcillin.
  • Carbepenems
  • Non-limiting examples of carbepenems include Imipenem-Cilastatin and Meropenem.
  • Monobactams
  • A non-limiting example of a monobactam includes Aztreonam.
  • Macrolides and Lincosamines
  • Non-limiting examples of macrolides and lincosamines include Azithromycin, Clarithromycin, Clindamycin, Dirithromycin, Erythromycin, Lincomycin, and Troleandomycin.
  • Glycopeptides
  • Non-limiting examples of glycopeptides include Teicoplanin and Vancomycia.
  • Rifampin
  • Non-limiting examples of rifampins include Rifabutin, Rifampin, and Rifapentine.
  • Oxazolidonones
  • Non-limiting example of oxazolidonones includes Linezolid.
  • Tetracyclines
  • Non-limiting examples of tetracyclines include Demeclocycline, Doxycycline, Methacycline, Minocycline, Oxytetracycline, Tetracycline, and Chlortetracycline.
  • Aminoglycosides
  • Non-limiting examples of aminoglycosides include Amikacin, Gentamicin, Kanamycin, Neomycin, Netilmicin, Streptomycin, Tobramycin, and Paromomycin.
  • Streptogramins
  • Non-limiting example of streptogramins includes Quinopristin+Dalfopristin.
  • Sulfonamides
  • Non-limiting examples of sulfonamides include Mafenide, Silver Sulfadiazine, Sulfacetamide, Sulfadiazine, Sulfamethoxazole, Sulfasalazine, Sulfisoxazole, Trimethoprim-Sulfamethoxazole, and Sulfamethizole.
  • Others
  • Non-limiting examples of other antibiotic agents include Bacitracin, Chloramphenicol, Colistemetate, Fosfomycin, Isoniazid, Methenamine, Metronidazol, Mupirocin, Nitroflurantoin, Nitrofurazone, Novobiocin, Polymyxin B, Spectinomycin, Trimethoprine, Trimethoprine/Sulfamethoxazole, Cationic peptides, Colistin, Iseganan, Cycloserine, Capreomycin,Pyrazinamide, Para-aminosalicyclic acid, and Erythromycin ethylsuccinate+sulfisoxazole.
  • Antiviral agents include, but are not limited to: zidovudine, acyclovir, ganciclovir, vidarabine, idoxuridine, trifluridine, ribavirin, interferon alpha-2a, interferon alpha-2b, interferon beta, interferon gamma).
  • Antifungal agents include, but are not limited to: amphotericin B, nystatin, hamycin, natamycin, pimaricin, ambruticin, itraconazole, terconazole, ketoconazole, voriconazole, miconazole, nikkomycin Z, griseofulvin, candicidin, cilofungin, chlotrimazole, clioquinol, caspufungin, tolnaflate.
  • Dosages
  • The dosage of any compositions of the present invention will vary depending on the symptoms, age and body weight of the patient, the nature and severity of the disorder to be treated or prevented, the route of administration, and the form of the subject composition. Any of the subject formulations may be administered in a single dose or in divided doses. Dosages for the compositions of the present invention may be readily determined by techniques known to those of skill in the art or as taught herein.
  • In certain embodiments, the dosage of the subject compounds will generally be in the range of about 0.01 ng to about 10 g per kg body weight, specifically in the range of about 1 ng to about 0.1 g per kg, and more specifically in the range of about 100 ng to about 10 mg per kg.
  • An effective dose or amount, and any possible affects on the timing of administration of the formulation, may need to be identified for any particular composition of the present invention. This may be accomplished by routine experiment as described herein, using one or more groups of animals (preferably at least 5 animals per group), or in human trials if appropriate. The effectiveness of any subject composition and method of treatment or prevention may be assessed by administering the composition and assessing the effect of the administration by measuring one or more applicable indices, and comparing the post-treatment values of these indices to the values of the same indices prior to treatment.
  • The precise time of administration and amount of any particular subject composition that will yield the most effective treatment in a given patient will depend upon the activity, pharmacokinetics, and bioavailability of a subject composition, physiological condition of the patient (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage and type of medication), route of administration, and the like. The guidelines presented herein may be used to optimize the treatment, e.g., determining the optimum time and/or amount of administration, which will require no more than routine experimentation consisting of monitoring the subject and adjusting the dosage and/or timing.
  • While the subject is being treated, the health of the patient may be monitored by measuring one or more of the relevant indices at predetermined times during the treatment period. Treatment, including composition, amounts, times of administration and formulation, may be optimized according to the results of such monitoring. The patient may be periodically reevaluated to determine the extent of improvement by measuring the same parameters. Adjustments to the amount(s) of subject composition administered and possibly to the time of administration may be made based on these reevaluations.
  • Treatment may be initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage may be increased by small increments until the optimum therapeutic effect is attained.
  • The use of the subject compositions may reduce the required dosage for any individual agent contained in the compositions (e.g., the Fabl inhibitor) because the onset and duration of effect of the different agents may be complimentary.
  • Toxicity and therapeutic efficacy of subject compositions may be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 and the ED50.
  • The data obtained from the cell culture assays and animal studies may be used in formulating a range of dosage for use in humans. The dosage of any subject composition lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For compositions of the present invention, the therapeutically effective dose may be estimated initially from cell culture assays.
  • Pharmaceutical Formulation
  • The pharmaceutical formulation of the antiinfective may be comprised of either an aqueous dispersion of liposomes and free antiinfective, or a dehydrated powder containing liposomes and free antiinfective. The formulation may contain lipid excipients to form the liposomes, and salts/buffers to provide the appropriate osmolarity and pH. The dry powder formulations may contain additional excipients to prevent the leakage of encapsulated antiinfective during the drying and potential milling steps needed to create a suitable particle size for inhalation (i.e., 1-5 μm). Such excipients are designed to increase the glass transition temperature of the antiinfective formulation. The pharmaceutical excipient may be a liquid or solid filler, diluent, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof from one organ or portion of the body, to another organ, or portion of the body. Each excipient must be “acceptable” in the sense of being compatible with the subject composition and its components and not injurious to the patient. Suitable excipients include trehalose, raffinose, mannitol, sucrose, leucine, trileucine, and calcium chloride. Examples of other suitable excipients include (1) sugars, such as lactose, and glucose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulos and cellulos acetate; 4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations.
  • Inhalation Device
  • The pharmaceutical formulations of the present invention may be used in any dosage dispensing device adapted for intranasal administration. The device should be constructed with a view to ascertaining optimum metering accuracy and compatibility of its constructive elements, such as container, valve and actuator with the nasal formulation and could be based on a mechanical pump system, e.g., that of a metered-dose nebulizer, dry powder inhaler, soft mist inhaler, or a nebulizer. Due to the large administered dose, preferred devices include jet nebulizers (e.g., PARI LC Star, AKITA), soft mist inhalers (e.g., PARI e-Flow), and capsule-based dry powder inhalers (e.g., PH&T Turbospin). Suitable propellants may be selected among such gases as fluorocarbons, hydrocarbons, nitrogen and dinitrogen oxide or mixtures thereof.
  • The inhalation delivery device can be a nebulizer or a metered dose inhaler (MDI), or any other suitable inhalation delivery device known to one or ordinary skill in the art. The device can contain and be used to deliver a single dose of the antiinfective compositions or the device can contain and be used to deliver multi-doses of the compositions of the present invention.
  • A nebulizer type inhalation delivery device can contain the compositions of the present invention as a solution, usually aqueous, or a suspension. In generating the nebulized spray of the compositions for inhalation, the nebulizer type delivery device may be driven ultrasonically, by compressed air, by other gases, electronically or mechanically. The ultrasonic nebulizer device usually works by imposing a rapidly oscillating waveform onto the liquid film of the formulation via an electrochemical vibrating surface. At a given amplitude the waveform becomes unstable, whereby it disintegrates the liquids film, and it produces small droplets of the formulation. The nebulizer device driven by air or other gases operates on the basis that a high pressure gas stream produces a local pressure drop that draws the liquid formulation into the stream of gases via capillary action. This fine liquid stream is then disintegrated by shear forces. The nebulizer may be portable and hand held in design, and may be equipped with a self contained electrical unit. The nebulizer device may comprise a nozzle that has two coincident outlet channels of defined aperture size through which the liquid formulation can be accelerated. This results in impaction of the two streams and atomization of the formulation. The nebulizer may use a mechanical actuator to force the liquid formulation through a multiorifice nozzle of defined aperture size(s) to produce an aerosol of the formulation for inhalation. In the design of single dose nebulizers, blister packs containing single doses of the formulations may be employed.
  • In the present invention the nebulizer may be employed to ensure the sizing of particles is optimal for positioning of the particle within, for example, the pulmonary membrane.
  • A metered dose inhalator (MDI) may be employed as the inhalation delivery device for the compositions of the present invention. This device is pressurized (pMDI) and its basic structure comprises a metering valve, an actuator and a container. A propellant is used to discharge the formulation from the device. The composition may consist of particles of a defined size suspended in the pressurized propellant(s) liquid, or the composition can be in a solution or suspension of pressurized liquid propellant(s). The propellants used are primarily atmospheric friendly hydroflourocarbons (HFCs) such as 134a and 227. Traditional chloroflourocarbons like CFC-11, 12 and 114 are used only when essential. The device of the inhalation system may deliver a single dose via, e.g., a blister pack, or it may be multi dose in design. The pressurized metered dose inhalator of the inhalation system can be breath actuated to deliver an accurate dose of the lipid-containing formulation. To insure accuracy of dosing, the delivery of the formulation may be programmed via a microprocessor to occur at a certain point in the inhalation cycle. The MDI may be portable and hand held.
  • Exemplification Example 1
  • Pharmacokinetics of amikacin delivered as both free and encapsulated amikacin in healthy volunteers. The nebulized liposomal amikacin contains a mixture of encapsulated (ca., 60%) and free amikacin (ca., 40%). Following inhalation in healthy volunteers the corrected nominal dose was 100 mg as determined by gamma scintigraphy. FIG. 1 depicts the lung concentration of amikacin and TOBI® (administered 100% free), based on pharmacokinetic modeling of serum concentrations over time. Both curves assume a volume of distribution for aminoglycosides in the lung of 200 ml. Interestingly, the peak levels of antiinfective in the lung are approximately equivalent for the 100 mg dose of liposomal amikacin, and the 300 mg dose of TOBI®. This is a consequence of the rapid clearance of the free tobramycin from the lung by absorption into the systemic circulation with a half-life of about 1.5 hr. These results serve as a demonstration of the improved lung targeting afforded by liposomal encapsulation The presence of free and encapsulated antiinfective in the amikacin formulation is demonstrated by the two component pharmacokinetic profile observed. Free amikacin is rapidly absorbed into the systemic circulation (with a half-life similar to TOBI), while the encapsulated drug has a lung half-life of approximately 45 hr. The free amikacin is available to provide bactericidal activity, while the encapsulated drug provides sustained levels of drug in the lung, enabling improved killing of resistant bacterial strains. The measured lung concentrations for the liposomal compartment are significantly above the MIC50 of 1240 clinical isolates of Pseudomonas aeruginosa, potentially reducing the development of resistance.
  • Example 2
  • Impact of free amikacin on the percentage of amikacin encapsulated in liposomes following nebulization. Liposomal preparations of amikacin may exhibit significant leakage of encapsulated drug during nebulization. As detailed in Table 1 below, the presence of free amikacin in solution was shown to surprisingly decrease the leakage of antiinfective by about four-fold from the liposome. While not wishing to be limited to any particular theory, it is hypothesized that liposomes break-up and re-form during nebulization, losing encapsulated antiinfective in the process. Alternatively, encapsulated antiinfective is lost during nebulization because the liposome membrane becomes leaky. When an excess of free antiinfective is present in solution, the free antiinfective is readily available in close proximity to the liposome, and is available to be taken back up into the liposome on re-formation.
  • TABLE 1
    Effect of free amikacin on the leakage of amikacin from liposome-
    encapsulated amikacin.
    Formu- % Free Amikacin % Free Amikacin % Free Amikacin
    lation (Pre-nebulization) (Post-nebulization) (Due to nebulization)
    A 3.3 (n = 1) 42.4 ± 3.2 (n = 3) 39.1 ± 3.2 (n = 3)
    B 53.6 ± 5.4 (n = 9) 63.3 ± 4.7 (n = 9)  9.8 ± 5.8 (n = 9)

    Wherein n is the number of measurements.
  • Incorporation by Reference
  • All of the patents and publications cited herein are hereby incorporated by reference.
  • Equivalents
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims (23)

1.-45. (canceled)
46. A method for treating or providing prophylaxis against a pulmonary infection in a patient in need thereof comprising, administering to the lungs of the patient an aerosolized pharmaceutical composition comprising free aminoglycoside in an amount effective to provide immediate bactericidal activity against the pulmonary infection and liposomal encapsulated aminoglycoside in an amount effective to provide sustained bactericidal activity against the pulmonary infection, wherein the lipid component of the liposome consists of electrically neutral lipids, wherein the electrically neutral lipids comprise a phosphatidylcholine and a sterol, and the aerosolized pharmaceutical composition comprises droplets of an aqueous solution or suspension.
47. the method of claim 46, wherein the aminoglycoside is amikacin.
48. The method of claim 46, wherein the aminoglycoside is gentamicin, kanamycin, neomycin, netilmicin, streptomycin, tobramycin or paromomycin.
49. The method of claim 46, wherein the sterol is cholesterol.
50. The method of claim 46, wherein the pulmonary infection is Pseudomonas aeruginosa.
51. The method of claim 46 wherein the pulmonary infection is a mycobacterial infection.
52. The method of claim 46, wherein the pulmonary infection is a Burkholderia infection.
53. The method of claim 51, wherein the mycobacterial infection is Mycobacterium leprae, Mycobacterium africamum, Mycobacterium asiaticum Mycobacterium avium-intracellulare, Mycobacterium chelonae, Mycobacterium abscessus, Mycobacterium chelonae-abscessus, Mycobacterium fallax, Mycobacterium fortuitum, Mycobacterium kansasil, Mycobacterium leprae, Mycobacterium malmoense, Mycobacterium shimoidei, Mycobacterium simiae, Mycobacterium szulgai, Mycobacterium xenopi or Mycobacterium tuberculosis.
54. The method of claim 53, wherein the mycobacterial infection is Mycobacterium abscessus or Mycobacterium avium intracellulare.
55. The method of claim 46, wherein the liposomal encapsulated aminoglycoside comprises a mixture of unilamellar vesicles and multilamellar vesicles.
56. The method of claim 46, wherein the phosphatidylcholine is dipalmitoylphosphatidylcholine (DPPC).
57. The method of claim 46, wherein the phosphatidylcholine is dipalmitoylphosphatidylcholine (DPPC) and the sterol is cholesterol.
58. The method of claim 46, wherein a ratio by weight of free aminoglycoside to the aminoglycoside encapsulated in the liposome is between about 1:100 and about 100:1.
59. The method of claim 46, wherein a ratio by weight of free aminoglycoside to the aminoglycoside encapsulated in the liposome is between about 1:10 and about 10:1.
60. The method of claim 46, wherein a ratio by weight of free aminoglycoside to the aminoglycoside encapsulated in the liposome is between about 1:2 and about 2:1.
61. The method of claim 46, wherein the liposomal encapsulated amikacin comprises unilamellar and multilamellar vesicles.
62. The method of claim 47, wherein the sterol is cholesterol.
63. The method of claim 47, wherein the phosphatidylcholine is dipalmitoylphosphatidylcholine (DPPC).
64. The method of claim 47, wherein the phosphatidylcholine is dipalmitoylphosphatidylcholine (DPPC) and the sterol is cholesterol.
65. The method of claim 47, wherein a ratio by weight of free amikacin to the amikacin encapsulated in the liposome is between about 1:100 and about 100:1.
66. The method of claim 47, wherein a ratio by weight of free amikacin to the amikacin encapsulated in the liposome is between about 1:10 and about 10:1.
67. The method of claim 47, wherein a ratio by weight of free amikacin to the amikacin encapsulated in the liposome is between about 1:2 and about 2:1.
US15/875,308 2005-12-08 2018-01-19 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof Abandoned US20180153918A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/875,308 US20180153918A1 (en) 2005-12-08 2018-01-19 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US16/746,205 US20200390758A1 (en) 2005-12-08 2020-01-17 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US18/087,506 US20230133762A1 (en) 2005-12-08 2022-12-22 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US74846805P 2005-12-08 2005-12-08
US11/634,343 US8226975B2 (en) 2005-12-08 2006-12-05 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/527,213 US8632804B2 (en) 2005-12-08 2012-06-19 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/666,420 US8642075B2 (en) 2005-12-08 2012-11-01 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US14/080,922 US20140072620A1 (en) 2005-12-08 2013-11-15 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/875,308 US20180153918A1 (en) 2005-12-08 2018-01-19 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/080,922 Continuation US20140072620A1 (en) 2005-12-08 2013-11-15 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/746,205 Continuation US20200390758A1 (en) 2005-12-08 2020-01-17 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof

Publications (1)

Publication Number Publication Date
US20180153918A1 true US20180153918A1 (en) 2018-06-07

Family

ID=38123420

Family Applications (17)

Application Number Title Priority Date Filing Date
US11/634,343 Active 2028-08-15 US8226975B2 (en) 2005-12-08 2006-12-05 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/527,213 Active US8632804B2 (en) 2005-12-08 2012-06-19 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/664,181 Active US8679532B2 (en) 2005-12-08 2012-10-30 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/666,420 Active US8642075B2 (en) 2005-12-08 2012-11-01 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/675,587 Active US8673349B2 (en) 2005-12-08 2012-11-13 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/675,559 Active US8673348B2 (en) 2005-12-08 2012-11-13 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US14/080,922 Abandoned US20140072620A1 (en) 2005-12-08 2013-11-15 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US14/987,508 Active US9402845B2 (en) 2005-12-08 2016-01-04 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/066,360 Abandoned US20160184302A1 (en) 2005-12-08 2016-03-10 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/066,346 Abandoned US20160184301A1 (en) 2005-12-08 2016-03-10 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/205,925 Active US9549939B2 (en) 2005-12-08 2016-07-08 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/205,918 Active US9511082B2 (en) 2005-12-08 2016-07-08 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/241,439 Active US9549925B2 (en) 2005-12-08 2016-08-19 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/376,086 Active 2027-07-16 US10328071B2 (en) 2005-12-08 2016-12-12 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/875,308 Abandoned US20180153918A1 (en) 2005-12-08 2018-01-19 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US16/746,205 Abandoned US20200390758A1 (en) 2005-12-08 2020-01-17 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US18/087,506 Pending US20230133762A1 (en) 2005-12-08 2022-12-22 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof

Family Applications Before (14)

Application Number Title Priority Date Filing Date
US11/634,343 Active 2028-08-15 US8226975B2 (en) 2005-12-08 2006-12-05 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/527,213 Active US8632804B2 (en) 2005-12-08 2012-06-19 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/664,181 Active US8679532B2 (en) 2005-12-08 2012-10-30 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/666,420 Active US8642075B2 (en) 2005-12-08 2012-11-01 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/675,587 Active US8673349B2 (en) 2005-12-08 2012-11-13 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US13/675,559 Active US8673348B2 (en) 2005-12-08 2012-11-13 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US14/080,922 Abandoned US20140072620A1 (en) 2005-12-08 2013-11-15 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US14/987,508 Active US9402845B2 (en) 2005-12-08 2016-01-04 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/066,360 Abandoned US20160184302A1 (en) 2005-12-08 2016-03-10 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/066,346 Abandoned US20160184301A1 (en) 2005-12-08 2016-03-10 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/205,925 Active US9549939B2 (en) 2005-12-08 2016-07-08 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/205,918 Active US9511082B2 (en) 2005-12-08 2016-07-08 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/241,439 Active US9549925B2 (en) 2005-12-08 2016-08-19 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US15/376,086 Active 2027-07-16 US10328071B2 (en) 2005-12-08 2016-12-12 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/746,205 Abandoned US20200390758A1 (en) 2005-12-08 2020-01-17 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US18/087,506 Pending US20230133762A1 (en) 2005-12-08 2022-12-22 Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof

Country Status (13)

Country Link
US (17) US8226975B2 (en)
EP (4) EP3067047B1 (en)
JP (1) JP5324223B2 (en)
AU (1) AU2006322076C1 (en)
CA (3) CA2896083A1 (en)
CY (1) CY1118059T1 (en)
DK (1) DK1962805T3 (en)
ES (2) ES2594368T3 (en)
HU (1) HUE029994T2 (en)
LT (1) LT1962805T (en)
PL (1) PL1962805T3 (en)
PT (1) PT1962805T (en)
WO (1) WO2007067520A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10238675B2 (en) 2014-05-15 2019-03-26 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US10328071B2 (en) 2005-12-08 2019-06-25 Insmed Incorporated Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US10471149B2 (en) 2012-11-29 2019-11-12 Insmed Incorporated Stabilized vancomycin formulations
US11571386B2 (en) 2018-03-30 2023-02-07 Insmed Incorporated Methods for continuous manufacture of liposomal drug products

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8409846B2 (en) 1997-09-23 2013-04-02 The United States Of America As Represented By The Department Of Veteran Affairs Compositions, methods and devices for maintaining an organ
US7879351B2 (en) * 2002-10-29 2011-02-01 Transave, Inc. High delivery rates for lipid based drug formulations, and methods of treatment thereof
US7718189B2 (en) 2002-10-29 2010-05-18 Transave, Inc. Sustained release of antiinfectives
ES2686361T3 (en) * 2002-10-29 2018-10-17 Insmed Incorporated Liposomes comprising an aminoglycoside for the treatment of lung infections
EP2316419B1 (en) 2004-05-17 2012-07-11 Gilead Sciences, Inc. Aerosolized fosfomycin/tobramycin combination for the treatment of bacterial respiratory infections
US9301519B2 (en) 2004-10-07 2016-04-05 Transmedics, Inc. Systems and methods for ex-vivo organ care
US8304181B2 (en) 2004-10-07 2012-11-06 Transmedics, Inc. Method for ex-vivo organ care and for using lactate as an indication of donor organ status
ES2348736T3 (en) 2004-10-07 2010-12-13 Transmedics, Inc. SYSTEMS AND METHODS FOR EX-LIVE ORGAN CARE.
US9078428B2 (en) 2005-06-28 2015-07-14 Transmedics, Inc. Systems, methods, compositions and solutions for perfusing an organ
EP3354260B1 (en) * 2006-04-06 2020-12-09 Insmed Incorporated Methods for coacervation induced liposomal encapsulation and formulations thereof
EP3677118A1 (en) 2006-04-19 2020-07-08 Transmedics, Inc. Systems for ex vivo organ care
US8268347B1 (en) 2006-10-24 2012-09-18 Aradigm Corporation Dual action, inhaled formulations providing both an immediate and sustained release profile
US20080138397A1 (en) * 2006-10-24 2008-06-12 Aradigm Corporation Processes for taste-masking of inhaled formulations
US8071127B2 (en) * 2006-10-24 2011-12-06 Aradigm Corporation Dual action, inhaled formulations providing both an immediate and sustained release profile
US8119156B2 (en) * 2006-10-24 2012-02-21 Aradigm Corporation Dual action, inhaled formulations providing both an immediate and sustained release profile
CN101209243B (en) 2006-12-29 2010-12-08 石药集团中奇制药技术(石家庄)有限公司 Liposome medicament and preparation thereof
US9457179B2 (en) 2007-03-20 2016-10-04 Transmedics, Inc. Systems for monitoring and applying electrical currents in an organ perfusion system
US20100196455A1 (en) * 2007-05-04 2010-08-05 Transave, Inc. Compositions of Multicationic Drugs for Reducing Interactions with Polyanionic Biomolecules and Methods of Use Thereof
US9114081B2 (en) 2007-05-07 2015-08-25 Insmed Incorporated Methods of treating pulmonary disorders with liposomal amikacin formulations
US9333214B2 (en) 2007-05-07 2016-05-10 Insmed Incorporated Method for treating pulmonary disorders with liposomal amikacin formulations
US9119783B2 (en) 2007-05-07 2015-09-01 Insmed Incorporated Method of treating pulmonary disorders with liposomal amikacin formulations
US20110059062A1 (en) * 2007-07-06 2011-03-10 Michael A Pellico Use of hydrolytic and oxidative enzymes to dissolve biofilm in airway passages
US20090291132A1 (en) * 2008-01-04 2009-11-26 Brian Charles Keller Enhanced delivery of antifungal agents
US8420380B2 (en) 2008-01-31 2013-04-16 Transmedics, Inc. Systems and methods for ex vivo lung care
KR20100137439A (en) * 2008-03-04 2010-12-30 엘란 파마 인터내셔널 리미티드 Stable liquid formulations of anti-infective agents and adjusted anti-infective agent dosing regimens
WO2009137611A2 (en) * 2008-05-06 2009-11-12 Board Of Regents, The University Of Texas System Treatment of pulmonary fungal infection with voriconazole via inhalation
IT1405998B1 (en) * 2010-12-09 2014-02-06 Bionest Ltd MULTIPURPOSE GEL AGAINST THE VAGINAL DRYNESS WITH A DIRECT AND DELAYED EFFECT
AU2012242578B2 (en) * 2011-04-14 2016-07-21 Transmedics, Inc. Organ care solution for ex-vivo machine perfusion of donor lungs
AU2012250568B2 (en) * 2011-05-05 2017-06-29 Matinas Biopharma Nanotechnologies, Inc. Cochleate compositions and methods of making and using same
US9572774B2 (en) 2011-05-19 2017-02-21 Savara Inc. Dry powder vancomycin compositions and associated methods
CA2870860C (en) 2012-05-21 2021-07-27 Insmed Incorporated Systems for treating pulmonary infections
EP3060198A4 (en) 2013-10-22 2017-06-28 Aradigm Corporation Inhaled surfactant-modified liposomal formulations providing both an immediate and sustained release profile
CA2928736A1 (en) * 2013-10-30 2015-05-07 Inspirx Inc. Inhaled aerosolized immuno-chemotherapy for the treatment of mdr tb
JP2017510662A (en) 2014-04-08 2017-04-13 アラダイム コーポレーション Liposomal ciprofloxacin preparation with activity against nontuberculous mycobacteria
ES2839202T3 (en) 2014-06-02 2021-07-05 Transmedics Inc Ex vivo organ care system
KR20180004168A (en) * 2015-04-22 2018-01-10 마티나스 바이오파마 나노테크놀로지스, 인코포레이티드 Compositions and methods for the treatment of mycobacterial infections and lung diseases
US10194655B2 (en) 2015-09-09 2019-02-05 Transmedics, Inc. Aortic cannula for ex vivo organ care system
US20210369675A1 (en) * 2016-08-20 2021-12-02 John Malcolm Hall Gregg Antimicrobial drug methods of use & therapeutic compositions
KR20210003197A (en) 2018-04-23 2021-01-11 티엘씨 바이오파머슈티컬즈 인코포레이티드 Inhalable liposome sustained-release composition for use in the treatment of lung diseases
CN114040750A (en) 2019-05-14 2022-02-11 盈擘医药股份有限公司 Inhalable bronchodilator sustained-release composition for treating pulmonary diseases
EP4125810A1 (en) * 2020-03-22 2023-02-08 InspirMed Corp. Composition of antiviral agent for use in prophylactic or post-exposure treatment of infectious or respiratory diseases
EP3906946A1 (en) 2020-05-04 2021-11-10 EK Biosciences GmbH Antipathogen vesicle
CA3183397A1 (en) * 2020-06-18 2021-12-23 Daryl C. Drummond Oxazolidinone compounds, liposome compositions comprising oxazolidinone compounds and methods of use thereof
WO2023183297A1 (en) * 2022-03-21 2023-09-28 The Board Of Trustees Of The University Of Illinois Ion channel prosthetic compositions comprising lipid-coated crystals of amphotericin b

Family Cites Families (334)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3091572A (en) 1962-07-16 1963-05-28 Schering Corp Gentamycin and method of production
US3136704A (en) 1962-12-05 1964-06-09 Schering Corp Manufacture of gentamycin
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4394448A (en) 1978-02-24 1983-07-19 Szoka Jr Francis C Method of inserting DNA into living cells
GB2046092B (en) 1979-03-05 1983-11-02 Toyama Chemical Co Ltd Pharmaceutical composition containing a lysophospholid and a phospholipid
HU184141B (en) 1979-12-27 1984-07-30 Human Oltoanyagtermelo Adjuvant particles compositions containing said particles and biologically active substances adsorbed thereon and a process for the preparation thereof
US4451447A (en) 1980-03-31 1984-05-29 Bristol-Myers Company Pharmaceutical formulations
EP0069307B1 (en) 1981-07-02 1986-03-05 F. HOFFMANN-LA ROCHE & CO. Aktiengesellschaft Process for preparing liposome solutions
US4547490A (en) 1981-12-31 1985-10-15 Neomed, Inc. Synthetic whole blood and a method of making the same
US4522803A (en) 1983-02-04 1985-06-11 The Liposome Company, Inc. Stable plurilamellar vesicles, their preparation and use
US4684625A (en) * 1982-07-08 1987-08-04 Syntex (U.S.A.) Inc. Method for enhancing the anti-infective activity of muramyldipeptide derivatives
US5030453A (en) 1983-03-24 1991-07-09 The Liposome Company, Inc. Stable plurilamellar vesicles
US4588578A (en) 1983-08-08 1986-05-13 The Liposome Company, Inc. Lipid vesicles prepared in a monophase
US4981692A (en) 1983-03-24 1991-01-01 The Liposome Company, Inc. Therapeutic treatment by intramammary infusion
US5169637A (en) 1983-03-24 1992-12-08 The Liposome Company, Inc. Stable plurilamellar vesicles
US4515736A (en) 1983-05-12 1985-05-07 The Regents Of The University Of California Method for encapsulating materials into liposomes
US5059591B1 (en) 1983-05-26 2000-04-25 Liposome Co Inc Drug preparations of reduced toxicity
CA1237670A (en) 1983-05-26 1988-06-07 Andrew S. Janoff Drug preparations of reduced toxicity
US4606939A (en) 1983-06-22 1986-08-19 The Ohio State University Research Foundation Small particle formation
CA1237671A (en) 1983-08-01 1988-06-07 Michael W. Fountain Enhancement of pharmaceutical activity
GB8322178D0 (en) 1983-08-17 1983-09-21 Sterwin Ag Preparing aerosol compositions
EP0153955A1 (en) 1983-09-06 1985-09-11 Health Research, Inc. Liposome delivery method for decreasing the toxicity of an antitumor drug
US4721612A (en) 1984-04-12 1988-01-26 The Liposome Company, Inc. Steroidal liposomes
US4963367A (en) 1984-04-27 1990-10-16 Medaphore, Inc. Drug delivery compositions and methods
US4794000A (en) 1987-01-08 1988-12-27 Synthetic Blood Corporation Coacervate-based oral delivery system for medically useful compositions
US5008050A (en) 1984-06-20 1991-04-16 The Liposome Company, Inc. Extrusion technique for producing unilamellar vesicles
SE8403905D0 (en) 1984-07-30 1984-07-30 Draco Ab LIPOSOMES AND STEROID ESTERS
US5736155A (en) 1984-08-08 1998-04-07 The Liposome Company, Inc. Encapsulation of antineoplastic agents in liposomes
US5077056A (en) 1984-08-08 1991-12-31 The Liposome Company, Inc. Encapsulation of antineoplastic agents in liposomes
US4880635B1 (en) 1984-08-08 1996-07-02 Liposome Company Dehydrated liposomes
ATE78158T1 (en) * 1985-05-22 1992-08-15 Liposome Technology Inc METHOD AND SYSTEM FOR INHALATION OF LIPOSOMES.
US4975282A (en) 1985-06-26 1990-12-04 The Liposome Company, Inc. Multilamellar liposomes having improved trapping efficiencies
US5409704A (en) 1985-06-26 1995-04-25 The Liposome Company, Inc. Liposomes comprising aminoglycoside phosphates and methods of production and use
US5059421A (en) 1985-07-26 1991-10-22 The Liposome Company, Inc. Preparation of targeted liposome systems of a defined size distribution
DE3689769T2 (en) 1985-07-05 1994-07-21 Liposome Co Inc MULTILAMELLAR LIPOSOME WITH IMPROVED INCLUDING EFFECT.
JPH0665648B2 (en) 1985-09-25 1994-08-24 塩野義製薬株式会社 Stable freeze-drying formulation of platinum anticancer substance
US4861580A (en) 1985-10-15 1989-08-29 The Liposome Company, Inc. Composition using salt form of organic acid derivative of alpha-tocopheral
US5041278A (en) 1985-10-15 1991-08-20 The Liposome Company, Inc. Alpha tocopherol-based vesicles
US5041581A (en) 1985-10-18 1991-08-20 The University Of Texas System Board Of Regents Hydrophobic cis-platinum complexes efficiently incorporated into liposomes
US5023087A (en) 1986-02-10 1991-06-11 Liposome Technology, Inc. Efficient method for preparation of prolonged release liposome-based drug delivery system
US6759057B1 (en) 1986-06-12 2004-07-06 The Liposome Company, Inc. Methods and compositions using liposome-encapsulated non-steroidal anti-inflammatory drugs
US4833134A (en) 1986-08-19 1989-05-23 Takeda Chemical Industries, Ltd. Cephem compounds
US5049388A (en) 1986-11-06 1991-09-17 Research Development Foundation Small particle aerosol liposome and liposome-drug combinations for medical use
US4933121A (en) 1986-12-10 1990-06-12 Ciba Corning Diagnostics Corp. Process for forming liposomes
US5320906A (en) 1986-12-15 1994-06-14 Vestar, Inc. Delivery vehicles with amphiphile-associated active ingredient
DE3751871D1 (en) 1986-12-23 1996-09-19 Liposome Co Inc Liposomes containing guanidine aminoglycoside
US5723147A (en) 1987-02-23 1998-03-03 Depotech Corporation Multivesicular liposomes having a biologically active substance encapsulated therein in the presence of a hydrochloride
US5616334A (en) 1987-03-05 1997-04-01 The Liposome Company, Inc. Low toxicity drug-lipid systems
MX9203808A (en) 1987-03-05 1992-07-01 Liposome Co Inc HIGH DRUG CONTENT FORMULATIONS: LIPID, FROM LIPOSOMIC-ANTINEOPLASTIC AGENTS.
US4857311A (en) 1987-07-31 1989-08-15 Massachusetts Institute Of Technology Polyanhydrides with improved hydrolytic degradation properties
US4895452A (en) 1988-03-03 1990-01-23 Micro-Pak, Inc. Method and apparatus for producing lipid vesicles
MX9203504A (en) 1988-04-20 1992-07-01 Liposome Co Inc AGENT COMPLEX: HIGH PROPORTION ACTIVE LIPID.
US5269979A (en) 1988-06-08 1993-12-14 Fountain Pharmaceuticals, Inc. Method for making solvent dilution microcarriers
IL91664A (en) 1988-09-28 1993-05-13 Yissum Res Dev Co Ammonium transmembrane gradient system for efficient loading of liposomes with amphipathic drugs and their controlled release
BE1001869A3 (en) 1988-10-12 1990-04-03 Franz Legros METHOD OF PACKAGING liposomal AMINOGLUCOSIDIQUES ANTIBIOTICS IN PARTICULAR THE GENTAMYCIN.
US4952405A (en) 1988-10-20 1990-08-28 Liposome Technology, Inc. Method of treating M. avium infection
US4906476A (en) 1988-12-14 1990-03-06 Liposome Technology, Inc. Novel liposome composition for sustained release of steroidal drugs in lungs
US5006343A (en) 1988-12-29 1991-04-09 Benson Bradley J Pulmonary administration of pharmaceutically active substances
US5843473A (en) * 1989-10-20 1998-12-01 Sequus Pharmaceuticals, Inc. Method of treatment of infected tissues
US5542935A (en) 1989-12-22 1996-08-06 Imarx Pharmaceutical Corp. Therapeutic delivery systems related applications
US5580575A (en) 1989-12-22 1996-12-03 Imarx Pharmaceutical Corp. Therapeutic drug delivery systems
WO1991009616A1 (en) 1989-12-22 1991-07-11 Yale University Quinolone antibiotics encapsulated in lipid vesicles
US5820848A (en) 1990-01-12 1998-10-13 The Liposome Company, Inc. Methods of preparing interdigitation-fusion liposomes and gels which encapsulate a bioactive agent
US5279833A (en) 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
EP0527940A1 (en) 1990-05-08 1993-02-24 Liposome Technology, Inc. Direct spray-dried drug/lipid powder composition
US6623671B2 (en) 1990-10-05 2003-09-23 Royden M. Coe Liposome extrusion process
US5614216A (en) 1990-10-17 1997-03-25 The Liposome Company, Inc. Synthetic lung surfactant
IT1245761B (en) 1991-01-30 1994-10-14 Alfa Wassermann Spa PHARMACEUTICAL FORMULATIONS CONTAINING GLYCOSAMINOGLICANS ABSORBABLE ORALLY.
EP0525132B1 (en) 1991-02-14 1996-01-03 Baxter International Inc. Binding of recognizing substances to liposomes
US6629646B1 (en) 1991-04-24 2003-10-07 Aerogen, Inc. Droplet ejector with oscillating tapered aperture
ATE155695T1 (en) 1991-11-07 1997-08-15 Ritzau Pari Werk Gmbh Paul NEBULISER, PARTICULARLY FOR USE IN DEVICES FOR INHALATION THERAPY
US5770563A (en) 1991-12-06 1998-06-23 The United States Of America As Represented By The Department Of Health And Human Services Heparin- and sulfatide binding peptides from the type I repeats of human thrombospondin and conjugates thereof
WO1993012240A1 (en) 1991-12-17 1993-06-24 The Regents Of The University Of California Gene therapy for cystic fibrosis transmembrane conductance regulator activity (cftr)
US5756353A (en) 1991-12-17 1998-05-26 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol-and liposome-based delivery
US5858784A (en) 1991-12-17 1999-01-12 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol- and liposome-based delivery
US6890555B1 (en) 1992-02-05 2005-05-10 Qlt, Inc. Liposome compositions of porphyrin photosensitizers
US5334761A (en) 1992-08-28 1994-08-02 Life Technologies, Inc. Cationic lipids
US5871710A (en) 1992-09-04 1999-02-16 The General Hospital Corporation Graft co-polymer adducts of platinum (II) compounds
US5958449A (en) 1992-12-02 1999-09-28 Nexstar Pharmaceuticals, Inc. Antibiotic formulation and use for bacterial infections
AU3244393A (en) 1992-12-02 1994-06-22 Vestar, Inc. Antibiotic formulation and process
US5665383A (en) 1993-02-22 1997-09-09 Vivorx Pharmaceuticals, Inc. Methods for the preparation of immunostimulating agents for in vivo delivery
US5395619A (en) 1993-03-03 1995-03-07 Liposome Technology, Inc. Lipid-polymer conjugates and liposomes
JPH09502700A (en) 1993-04-02 1997-03-18 ザ リポソーム カンパニー、インコーポレーテッド Method for producing liposome
CA2120197A1 (en) 1993-04-02 1994-10-03 Kenji Endo Stable aqueous dispersions containing liposomes
US5759571A (en) 1993-05-11 1998-06-02 Nexstar Pharmaceuticals, Inc. Antibiotic formulation and use for drug resistant infections
US5497763A (en) 1993-05-21 1996-03-12 Aradigm Corporation Disposable package for intrapulmonary delivery of aerosolized formulations
JPH06345663A (en) 1993-06-08 1994-12-20 Sumitomo Pharmaceut Co Ltd Liposome preparation containing vancomycin
JP3479535B2 (en) 1993-07-08 2003-12-15 ザ リポソーム カンパニー、インコーポレーテッド Methods for controlling liposome particle size
CA2101241C (en) 1993-07-23 1998-12-22 Jonathan P. Wong Liposome-encapsulated ciprofloxacin
DE69409361T2 (en) 1993-11-05 1998-11-12 Amgen Inc PRODUCTION OF LIPOSOMES AND METHOD FOR ENCODING SUBSTANCES
US5766627A (en) 1993-11-16 1998-06-16 Depotech Multivescular liposomes with controlled release of encapsulated biologically active substances
JPH09506866A (en) 1993-12-14 1997-07-08 ジョーンズ ホプキンス ユニバーシティー スクール オブ メディシン Controlled release of pharmaceutically active substances for immunotherapy
ES2120471T3 (en) 1993-12-17 1998-11-01 Pari Gmbh Spezialisten Fuer Effektive Inhalation SPRAY NOZZLE.
AU710504B2 (en) 1994-03-15 1999-09-23 Brown University Research Foundation Polymeric gene delivery system
US5610198A (en) 1994-03-18 1997-03-11 The United States Of America As Represented By The Department Of Health And Human Services Anti-mycobacterial compositions and their use for the treatment of tuberculosis and related diseases
US6221335B1 (en) 1994-03-25 2001-04-24 Isotechnika, Inc. Method of using deuterated calcium channel blockers
ATE205098T1 (en) 1994-05-19 2001-09-15 Pari Gmbh DEVICE FOR DRYING AND BUFFERING AEROSOLS
US5550109A (en) 1994-05-24 1996-08-27 Magainin Pharmaceuticals Inc. Inducible defensin peptide from mammalian epithelia
US5741516A (en) 1994-06-20 1998-04-21 Inex Pharmaceuticals Corporation Sphingosomes for enhanced drug delivery
US5543152A (en) 1994-06-20 1996-08-06 Inex Pharmaceuticals Corporation Sphingosomes for enhanced drug delivery
US5993850A (en) 1994-09-13 1999-11-30 Skyepharma Inc. Preparation of multivesicular liposomes for controlled release of encapsulated biologically active substances
US5753613A (en) 1994-09-30 1998-05-19 Inex Pharmaceuticals Corporation Compositions for the introduction of polyanionic materials into cells
US5508269A (en) 1994-10-19 1996-04-16 Pathogenesis Corporation Aminoglycoside formulation for aerosolization
US6000394A (en) 1994-10-26 1999-12-14 Paul Rizau Pari-Werk Gmbh Generation of an aerosol of an exact dose
AR002009A1 (en) 1994-12-22 1998-01-07 Astra Ab PHARMACEUTICAL COMPOSITION, PROCEDURE FOR THE MANUFACTURE OF A PROLIPOSOMA POWDER AS USED IN SUCH COMPOSITION, PROCEDURE FOR LAMANUFACTURE OF SUCH COMPOSITION, USE OF SUCH PHARMACEUTICAL COMPOSITION IN THE MANUFACTURE OF A DISPOSAL MEDICINAL PRODUCT.
US5662929A (en) 1994-12-23 1997-09-02 Universite De Montreal Therapeutic liposomal formulation
US5883074A (en) 1995-02-08 1999-03-16 Microcide Pharmaceuticals, Inc. Potentiators of antibacterial agents
US5972379A (en) 1995-02-14 1999-10-26 Sequus Pharmaceuticals, Inc. Liposome composition and method for administering a quinolone
US5800833A (en) 1995-02-27 1998-09-01 University Of British Columbia Method for loading lipid vesicles
AU5557796A (en) 1995-04-18 1996-11-07 Dehlinger, Peter J. Liposome drug-loading method and composition
US5855610A (en) 1995-05-19 1999-01-05 Children's Medical Center Corporation Engineering of strong, pliable tissues
AU5799996A (en) 1995-05-26 1996-12-11 Cell Genesys, Inc. Delivery vehicles comprising stable lipid/nucleic acid compl exes
DE19520622C2 (en) 1995-06-06 2003-05-15 Pari Gmbh Device for atomizing fluids
DK0782448T3 (en) 1995-06-06 2002-07-08 Bayer Ag Non-irritating, non-sensitizing, non-ototoxic antibacterial ear preparations
US5643599A (en) 1995-06-07 1997-07-01 President And Fellows Of Harvard College Intracellular delivery of macromolecules
US6521211B1 (en) 1995-06-07 2003-02-18 Bristol-Myers Squibb Medical Imaging, Inc. Methods of imaging and treatment with targeted compositions
CA2224253A1 (en) 1995-06-09 1996-12-27 Martin J. Macphee Chitin hydrogels, methods of their production and use
US5942253A (en) 1995-10-12 1999-08-24 Immunex Corporation Prolonged release of GM-CSF
AU730969B2 (en) 1995-10-19 2001-03-22 University Of Washington Discrete-length polyethylene glycols
DE19602628C2 (en) 1996-01-25 2000-06-29 Pari Gmbh Nebulizer
GB9602969D0 (en) 1996-02-13 1996-04-10 The Technology Partnership Plc Liquid supply apparatus
US5840702A (en) 1996-03-22 1998-11-24 Uab Research Foundation Cystic fibrosis treatment
CA2250219C (en) 1996-03-28 2008-10-07 Hayat Onyuksel Materials and methods for making improved liposome compositions
US5875776A (en) 1996-04-09 1999-03-02 Vivorx Pharmaceuticals, Inc. Dry powder inhaler
US6132765A (en) 1996-04-12 2000-10-17 Uroteq Inc. Drug delivery via therapeutic hydrogels
CA2174803C (en) 1996-04-23 2000-07-11 Jonathan P. Wong Use of liposome encapsulated ciprofloxacin as an immunotherapeutic drug
DE19616573C2 (en) 1996-04-25 1999-03-04 Pari Gmbh Use of subcritical blowing agent mixtures and aerosols for the micronization of drugs with the help of dense gases
DK0910382T3 (en) 1996-04-26 2003-10-06 Genaera Corp Squalamine in combination with other anticancer agents for the treatment of tumors
GB9609779D0 (en) 1996-05-10 1996-07-17 Univ Bruxelles Freeze dried liposome encapsulated amphiphilic drug compositions and a process for the preparation thereof
US6254854B1 (en) 1996-05-24 2001-07-03 The Penn Research Foundation Porous particles for deep lung delivery
US6770291B2 (en) 1996-08-30 2004-08-03 The United States Of America As Represented By The Department Of Health And Human Services Liposome complexes for increased systemic delivery
US6503881B2 (en) 1996-08-21 2003-01-07 Micrologix Biotech Inc. Compositions and methods for treating infections using cationic peptides alone or in combination with antibiotics
WO1998007409A1 (en) 1996-08-23 1998-02-26 Sequus Pharmaceuticals, Inc. Liposomes containing a cisplatin compound
TW520297B (en) 1996-10-11 2003-02-11 Sequus Pharm Inc Fusogenic liposome composition and method
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
US5837282A (en) 1996-10-30 1998-11-17 University Of British Columbia Ionophore-mediated liposome loading
CN1245423A (en) 1996-12-30 2000-02-23 巴特勒纪念研究院 Formulation and method for treating neoplasms by inhalation
US6451784B1 (en) 1996-12-30 2002-09-17 Battellepharma, Inc. Formulation and method for treating neoplasms by inhalation
US6458373B1 (en) 1997-01-07 2002-10-01 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
DE19713636A1 (en) 1997-04-02 1998-10-08 Pari Gmbh Breath simulator
US20020039594A1 (en) 1997-05-13 2002-04-04 Evan C. Unger Solid porous matrices and methods of making and using the same
JP4142149B2 (en) 1997-07-10 2008-08-27 明治製菓株式会社 Vancomycin lyophilized formulation
DE19734022C2 (en) 1997-08-06 2000-06-21 Pari Gmbh Inhalation therapy device with a valve to limit the flow of inspiration
US6106858A (en) 1997-09-08 2000-08-22 Skyepharma, Inc. Modulation of drug loading in multivescular liposomes
CA2215716C (en) * 1997-09-17 1999-12-07 Her Majesty The Queen, In Right Of Canada, As Represented By The Ministe R Of National Defence Aerosol delivery of liposome-encapsulated fluoroquinolone
US6090407A (en) 1997-09-23 2000-07-18 Research Development Foundation Small particle liposome aerosols for delivery of anti-cancer drugs
KR20010031363A (en) 1997-10-22 2001-04-16 젠스 포니카우 Use of antifungal agents for the topical treatment of fungus-induced mucositis
US6051251A (en) 1997-11-20 2000-04-18 Alza Corporation Liposome loading method using a boronic acid compound
WO1999030686A1 (en) 1997-12-12 1999-06-24 Inex Pharmaceuticals Corp. Cationic drugs encapsulated in anionic liposomes
GB9827370D0 (en) 1998-01-16 1999-02-03 Pari Gmbh Mouthpiece for inhalation therapy units
US6468532B1 (en) 1998-01-22 2002-10-22 Genentech, Inc. Methods of treating inflammatory diseases with anti-IL-8 antibody fragment-polymer conjugates
WO1999061003A1 (en) 1998-05-27 1999-12-02 Euroceltique S.A. Drug delivery system comprising a tightly compacted solid medicament stock
US6200598B1 (en) 1998-06-18 2001-03-13 Duke University Temperature-sensitive liposomal formulation
DE19827228C2 (en) 1998-06-18 2000-07-13 Pari Gmbh Liquid atomizer device
US6509323B1 (en) 1998-07-01 2003-01-21 California Institute Of Technology Linear cyclodextrin copolymers
US6916490B1 (en) 1998-07-23 2005-07-12 UAB Research Center Controlled release of bioactive substances
WO2000009089A1 (en) 1998-08-12 2000-02-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem Liposomal bupivacaine compositions prepared using an ammonium sulfate gradient
DE19846382C1 (en) 1998-10-08 2000-07-06 Pari Gmbh Counter and its use in inhalers, nebulizers or similar metered dose inhalers
JP2002529393A (en) * 1998-11-12 2002-09-10 フランク ジー. ピルキーウィッツ, Inhalation system
ATE397488T1 (en) 1998-11-13 2008-06-15 Heriot William A DEVICE FOR PRODUCING LIPOSOMES
US6855296B1 (en) 1998-11-13 2005-02-15 Optime Therapeutics, Inc. Method and apparatus for liposome production
CH693763A5 (en) 1998-12-17 2004-01-30 Chiron Corp Use of an antibiotic for the preparation of a medicament for the treatment of severe chronic bronchitis (bronchiectasis).
US6211162B1 (en) 1998-12-30 2001-04-03 Oligos Etc. Inc. Pulmonary delivery of protonated/acidified nucleic acids
IL144756A0 (en) 1999-02-08 2002-06-30 Alza Corp Method for controlling liposome size
EP1173193A4 (en) 1999-04-02 2003-01-29 Univ Princeton Desleucyl glycopeptide antibiotics and methods of making same
US6613352B2 (en) 1999-04-13 2003-09-02 Universite De Montreal Low-rigidity liposomal formulation
US7297344B1 (en) 1999-05-27 2007-11-20 Euro-Celtique, S.A. Preparations for the promotion of wound healing in the upper respiratory tract and/or ear
AU780454B2 (en) 1999-06-03 2005-03-24 Jessie L.S. Au Methods and compositions for modulating cell proliferation and cell death
EP1190705A4 (en) 1999-06-24 2009-03-18 Kyowa Hakko Kogyo Kk Method of regulating leakage of drug encapsulated in liposomes
CA2378438C (en) 1999-07-15 2010-05-04 Inex Pharmaceuticals Corp. Methods for preparation of lipid-encapsulated therapeutic agents
US6352996B1 (en) 1999-08-03 2002-03-05 The Stehlin Foundation For Cancer Research Liposomal prodrugs comprising derivatives of camptothecin and methods of treating cancer using these prodrugs
US6174878B1 (en) 1999-08-31 2001-01-16 Alcon Laboratories, Inc. Topical use of kappa opioid agonists to treat otic pain
US6235177B1 (en) 1999-09-09 2001-05-22 Aerogen, Inc. Method for the construction of an aperture plate for dispensing liquid droplets
US6962151B1 (en) 1999-11-05 2005-11-08 Pari GmbH Spezialisten für effektive Inhalation Inhalation nebulizer
DE19953317C1 (en) 1999-11-05 2001-02-01 Pari Gmbh Aerosol generation unit for inhalation therapies, comprises a valve which allows patient's exhaled air to enter into the surroundings, but prevents direct entry of external air into the mixing chamber during the inhaling phase
US6511676B1 (en) 1999-11-05 2003-01-28 Teni Boulikas Therapy for human cancers using cisplatin and other drugs or genes encapsulated into liposomes
JP4198850B2 (en) 1999-11-29 2008-12-17 オムロンヘルスケア株式会社 Liquid spray device
WO2001039789A1 (en) 1999-12-04 2001-06-07 Research Development Foundation Carbon dioxide enhancement of inhalation therapy
DE10004465A1 (en) 2000-02-02 2001-08-16 Pari Gmbh Inhalation nebulizer, comprises a liquid reservoir with an artificial surface structure comprising protrusions and indentations
WO2001056548A2 (en) 2000-02-04 2001-08-09 Lipoxen Technologies Limited Liposomes composition produced by a dehydration-rehydration process
US6761877B2 (en) 2000-02-18 2004-07-13 Biocrystal, Ltd. Functionalized encapsulated fluorescent nanocrystals
US7100600B2 (en) 2001-03-20 2006-09-05 Aerogen, Inc. Fluid filled ampoules and methods for their use in aerosolizers
US7600511B2 (en) 2001-11-01 2009-10-13 Novartis Pharma Ag Apparatus and methods for delivery of medicament to a respiratory system
US6948491B2 (en) 2001-03-20 2005-09-27 Aerogen, Inc. Convertible fluid feed system with comformable reservoir and methods
US7971588B2 (en) 2000-05-05 2011-07-05 Novartis Ag Methods and systems for operating an aerosol generator
MXPA02010884A (en) 2000-05-05 2003-03-27 Aerogen Ireland Ltd Apparatus and methods for the delivery of medicaments to the respiratory system.
US8336545B2 (en) 2000-05-05 2012-12-25 Novartis Pharma Ag Methods and systems for operating an aerosol generator
WO2001093846A2 (en) 2000-05-23 2001-12-13 The Trustees Of Columbia University In The City Of New York Method for treating respiratory disorders associated with pulmonary elastic fiber injury comprising the use of clycosaminoglycans
US6338859B1 (en) 2000-06-29 2002-01-15 Labopharm Inc. Polymeric micelle compositions
US6521736B2 (en) 2000-09-15 2003-02-18 University Of Massachusetts Amphiphilic polymeric materials
CA2424345A1 (en) 2000-10-16 2002-04-25 Neopharm, Inc. Liposomal formulation of mitoxantrone
CN1116875C (en) 2000-10-19 2003-08-06 南京振中生物工程有限公司 Taxusol-lipid composition and its preparing process
US6497901B1 (en) 2000-11-02 2002-12-24 Royer Biomedical, Inc. Resorbable matrices for delivery of bioactive compounds
EP1203614A1 (en) 2000-11-03 2002-05-08 Polymun Scientific Immunbiologische Forschung GmbH Process and apparatus for preparing lipid vesicles
AU2002219998B2 (en) 2000-12-01 2006-03-02 Biomira, Inc. Preparation of large liposomes by infusion into peg
CN1286649C (en) 2000-12-26 2006-11-29 精工爱普生株式会社 Ink jet recording device, and ink type information setting method and cartridge therein
DE10102846B4 (en) 2001-01-23 2012-04-12 Pari Pharma Gmbh aerosol generator
CA2437555A1 (en) 2001-02-01 2002-08-08 Yiyu Zou Stabilised polymeric aerosols for pulmonary gene delivery
DE10109897A1 (en) 2001-02-21 2002-11-07 Novosom Ag Optional cationic liposomes and their use
US6546927B2 (en) 2001-03-13 2003-04-15 Aerogen, Inc. Methods and apparatus for controlling piezoelectric vibration
US6554201B2 (en) 2001-05-02 2003-04-29 Aerogen, Inc. Insert molded aerosol generator and methods
US6732944B2 (en) 2001-05-02 2004-05-11 Aerogen, Inc. Base isolated nebulizing device and methods
EP2186508A3 (en) 2001-05-18 2010-08-25 Novartis AG Methods and unit dose formulations for the inhalation administration of aminoglycoside antibiotics
US20030060451A1 (en) 2001-05-29 2003-03-27 Rajneesh Taneja Enhancement of oral bioavailability of non-emulsified formulations of prodrug esters with lecithin
DE10126808C1 (en) 2001-06-01 2002-08-14 Pari Gmbh inhalation mask
DE10126807C2 (en) 2001-06-01 2003-12-04 Pari Gmbh Inhalation therapy device with a valve to limit the flow of inspiration
EP1269993A1 (en) 2001-06-21 2003-01-02 Applied NanoSystems B.V. Delivery of small hydrophilic molecules packaged into lipid vesicles
CA2451432A1 (en) 2001-06-23 2003-01-03 Lyotropic Therapeutics, Inc. Particles with improved solubilization capacity
AU2002323151A1 (en) 2001-08-13 2003-03-03 University Of Pittsburgh Application of lipid vehicles and use for drug delivery
WO2003015707A2 (en) 2001-08-20 2003-02-27 Transave, Inc. Method for treating lung cancers
US6623723B2 (en) 2001-08-21 2003-09-23 Cellular Sciences Inc. Method for treating bronchial constriction and bronchospasm
SK1012004A3 (en) 2001-08-21 2005-04-01 Pfizer Products Inc. Use of azithromycin for the manufacture of a medicament for the treatment of respiratory infections in humans
EP1304131B1 (en) 2001-10-18 2005-06-29 PARI GmbH Spezialisten für effektive Inhalation Inhalator
ATE269735T1 (en) 2001-10-18 2004-07-15 Pari Gmbh INHALATION THERAPY DEVICE
US20030096774A1 (en) 2001-11-21 2003-05-22 Igor Gonda Compositions of nucleic acids and cationic aminoglycosides and methods of using and preparing the same
TWI324518B (en) 2001-12-19 2010-05-11 Nektar Therapeutics Pulmonary delivery of aminoglycosides
AU2003235678A1 (en) 2002-01-15 2003-07-30 Aerogen, Inc. Systems and methods for clearing aerosols from the effective anatomic dead space
ES2603067T3 (en) 2002-01-15 2017-02-23 Novartis Ag Methods and systems for operating an aerosol generator
US20040009126A1 (en) * 2002-03-05 2004-01-15 Transave, Inc. Inhalation system for prevention and treatment of intracellular infections
US20030205226A1 (en) 2002-05-02 2003-11-06 Pre Holding, Inc. Aerosol medication inhalation system
ES2572770T3 (en) 2002-05-20 2016-06-02 Novartis Ag Apparatus for providing spray for medical treatment and methods
EP2338478B1 (en) 2002-06-28 2014-07-23 Protiva Biotherapeutics Inc. Method for producing liposomes
US20040014028A1 (en) * 2002-07-19 2004-01-22 Wolf Biotech Method for determination of protective epitopes for vaccination, diagnosis and vaccine quality control
MXPA05001312A (en) 2002-08-02 2005-08-03 Transave Inc Platinum aggregates and process for producing the same.
ATE463304T1 (en) 2002-08-02 2010-04-15 Pari Pharma Gmbh DEVICE FOR GENERATING LIQUID DROPS
AU2003268087A1 (en) * 2002-08-23 2004-03-11 Ian Ma Liposomal gemcitabine compositions for better drug delivery
DE10239321B3 (en) 2002-08-27 2004-04-08 Pari GmbH Spezialisten für effektive Inhalation Aerosol therapy device
KR100489701B1 (en) 2002-10-09 2005-05-16 주식회사 태평양 Submicron-liposome containing highly concentrated triterpenoid and method for preparing thereof
ES2686361T3 (en) 2002-10-29 2018-10-17 Insmed Incorporated Liposomes comprising an aminoglycoside for the treatment of lung infections
US7879351B2 (en) 2002-10-29 2011-02-01 Transave, Inc. High delivery rates for lipid based drug formulations, and methods of treatment thereof
US7718189B2 (en) 2002-10-29 2010-05-18 Transave, Inc. Sustained release of antiinfectives
DE10250625A1 (en) 2002-10-30 2004-05-19 Pari GmbH Spezialisten für effektive Inhalation Inhalation therapy device
EP1565165A2 (en) 2002-11-26 2005-08-24 Gilead Sciences, Inc. Liposomal formulations
DE10257381B4 (en) 2002-12-09 2006-09-14 Pari GmbH Spezialisten für effektive Inhalation Inhalation therapy device
US7968115B2 (en) 2004-03-05 2011-06-28 Board Of Regents, The University Of Texas System Liposomal curcumin for treatment of cancer
CN1767831B (en) 2003-04-08 2014-12-10 普罗热尼奇制药公司 Pharmaceutical formulations containing methylnaltrexone
US6900184B2 (en) 2003-04-14 2005-05-31 Wyeth Holdings Corporation Compositions containing pipercillin and tazobactam useful for injection
DE10320143A1 (en) 2003-05-06 2004-12-16 Pari GmbH Spezialisten für effektive Inhalation Nebulizer connection device for respirators or the like
US8058493B2 (en) 2003-05-21 2011-11-15 Baker Hughes Incorporated Removing amines from hydrocarbon streams
WO2004110493A2 (en) 2003-05-30 2004-12-23 Alza Corporation Method of pulmonary administration of an agent
US8616195B2 (en) 2003-07-18 2013-12-31 Novartis Ag Nebuliser for the production of aerosolized medication
GB2388581A (en) 2003-08-22 2003-11-19 Danisco Coated aqueous beads
DE10345950A1 (en) 2003-10-02 2005-05-19 Pari GmbH Spezialisten für effektive Inhalation Inhalation therapy device with valve
DE10347994A1 (en) 2003-10-15 2005-06-16 Pari GmbH Spezialisten für effektive Inhalation Aqueous aerosol preparation
DE10348237A1 (en) 2003-10-16 2005-05-19 Pari GmbH Spezialisten für effektive Inhalation Inhalation therapy device with a jet nebulizer
US20050214224A1 (en) * 2003-11-04 2005-09-29 Nektar Therapeutics Lipid formulations for spontaneous drug encapsulation
AU2004312090B2 (en) 2003-12-31 2008-08-14 The Board Of Regents, The University Of Texas System Pharmaceutical composition for thrombin peptide derivatives
US7452524B2 (en) 2004-01-27 2008-11-18 Gilead Sciences, Inc. Method for improvement of tolerance for therapeutically effective agents delivered by inhalation
US7556799B2 (en) 2004-03-30 2009-07-07 Relypsa, Inc. Ion binding polymers and uses thereof
DE102004016985B4 (en) 2004-04-07 2010-07-22 Pari Pharma Gmbh Aerosol generating device and inhalation device
JP4452799B2 (en) 2004-07-14 2010-04-21 独立行政法人産業技術総合研究所 Method for producing liposome using coacervate
EP1809254A2 (en) 2004-10-28 2007-07-25 Alza Corporation Lyophilized liposome formulations and method
US8337815B2 (en) 2004-12-23 2012-12-25 Discovery Laboratories, Inc. Pulmonary surfactant formulations
DE102005006372B4 (en) 2005-02-11 2007-11-29 Pari GmbH Spezialisten für effektive Inhalation Inhalation therapy device and method for its operation
DE102005006374B3 (en) 2005-02-11 2006-07-20 Pari GmbH Spezialisten für effektive Inhalation Aerosol production device, comprises a circular membrane for atomizing liquid, piezoelectric actuator coupled to the membrane, flexible platinum substrate, electrical lines, and reinforcement area
DE102005006375B4 (en) 2005-02-11 2007-10-11 Pari GmbH Spezialisten für effektive Inhalation Aerosol generating device for inhalation therapy devices
JP2006263054A (en) 2005-03-23 2006-10-05 Konica Minolta Sensing Inc Acquisition method of respiratory disease related analysis data, oxymeter system, its operation program, oxymeter and oxygen supply system
EP1712220A1 (en) 2005-04-15 2006-10-18 PARI GmbH Spezialisten für effektive Inhalation Pharmaceutical aerosol composition
BRPI0611198B1 (en) 2005-05-25 2018-02-06 Aerogen, Inc. VIBRATION SYSTEMS AND METHODS
DE102005024779B4 (en) 2005-05-31 2008-02-21 Pari GmbH Spezialisten für effektive Inhalation Breath-controlled inhalation therapy device
DE102005029498B4 (en) 2005-06-24 2007-08-30 Pari GmbH Spezialisten für effektive Inhalation Inhalation therapy device
USD656604S1 (en) 2005-06-28 2012-03-27 Pari Gmbh Part for inhalation therapy nebuliser
DE102005034403B3 (en) 2005-07-22 2007-02-22 Airbus Deutschland Gmbh Guide means for a device for the production of fiber preforms in the TFP process for composite components
US9005654B2 (en) 2005-07-27 2015-04-14 Protiva Biotherapeutics, Inc. Systems and methods for manufacturing liposomes
ATE439802T1 (en) 2005-09-07 2009-09-15 Koninkl Philips Electronics Nv SYSTEM AND METHOD FOR INDUCTIVELY MEASURING THE BIOIMPEDANCE OF A CONDUCTIVE TISSUE
US20070065367A1 (en) 2005-09-20 2007-03-22 Rany Condos Method of treating pulmonary disease with interferons
KR100705981B1 (en) 2005-10-12 2007-04-10 주식회사 리제론 Compositions comprising human growth hormone for preventing hair loss or stimulating hair sprouting
US20070105757A1 (en) 2005-10-31 2007-05-10 May Thomas B Vancomycin formulations having reduced amount of histamine
DE102006051512A1 (en) 2005-12-06 2007-06-14 Pari GmbH Spezialisten für effektive Inhalation Pharmaceutical drug compositions with cyclosporin
CA2896083A1 (en) 2005-12-08 2007-06-14 Insmed Incorporated Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
DE102006001113B3 (en) 2006-01-09 2007-06-28 Pari GmbH Spezialisten für effektive Inhalation Aerosol therapy device comprises an atomizer, an aerosol generator, a nosepiece for delivering aerosol to one nostril, a device for creating flow resistance in the other nostril, and a connector that imparts pressure fluctuations
US8263645B2 (en) 2006-02-03 2012-09-11 Pari Pharma Gmbh Disodium cromoglycate compositions and methods for administering same
DE102006006183A1 (en) 2006-02-10 2007-08-16 Pari GmbH Spezialisten für effektive Inhalation Inhalation therapy device for use in premature babies and toddlers
US7958887B2 (en) 2006-03-10 2011-06-14 Aradigm Corporation Nozzle pore configuration for intrapulmonary delivery of aerosolized formulations
DE102006012174A1 (en) 2006-03-16 2007-09-20 Pari GmbH Spezialisten für effektive Inhalation Inhalation therapy devices compressor
EP3354260B1 (en) 2006-04-06 2020-12-09 Insmed Incorporated Methods for coacervation induced liposomal encapsulation and formulations thereof
DE102006017002B3 (en) 2006-04-11 2007-01-11 Pari GmbH Spezialisten für effektive Inhalation Nebulizer for inhalation therapy comprises compressed gas outlet openings arranged in a row to which are assigned fluid outlet openings forming a nozzle with the gas outlet openings
USD583928S1 (en) 2006-04-27 2008-12-30 Pari Pharama Gmbh Nebulizer
US20080131497A1 (en) 2006-09-28 2008-06-05 Perkins Walter R Formulations of DNase and Methods of Use Thereof
US8119156B2 (en) 2006-10-24 2012-02-21 Aradigm Corporation Dual action, inhaled formulations providing both an immediate and sustained release profile
US8268347B1 (en) 2006-10-24 2012-09-18 Aradigm Corporation Dual action, inhaled formulations providing both an immediate and sustained release profile
US8071127B2 (en) 2006-10-24 2011-12-06 Aradigm Corporation Dual action, inhaled formulations providing both an immediate and sustained release profile
US20080108104A1 (en) 2006-11-02 2008-05-08 Colorado State University Research Foundation Identification of bacterial species and subspecies using lipids
EP1927373B1 (en) 2006-11-30 2012-08-22 PARI Pharma GmbH Inhalation nebulizer
CN102648916A (en) 2007-02-09 2012-08-29 联合治疗公司 Treprostinil treatment for interstitial lung disease and asthma
DE502007002037D1 (en) 2007-04-11 2009-12-31 Pari Gmbh Aerosol therapy device
US20100196455A1 (en) 2007-05-04 2010-08-05 Transave, Inc. Compositions of Multicationic Drugs for Reducing Interactions with Polyanionic Biomolecules and Methods of Use Thereof
US9119783B2 (en) 2007-05-07 2015-09-01 Insmed Incorporated Method of treating pulmonary disorders with liposomal amikacin formulations
US9114081B2 (en) 2007-05-07 2015-08-25 Insmed Incorporated Methods of treating pulmonary disorders with liposomal amikacin formulations
WO2008137917A1 (en) 2007-05-07 2008-11-13 Transave, Inc. Method of treating bacterial infections with antibacterial formulations
US9333214B2 (en) 2007-05-07 2016-05-10 Insmed Incorporated Method for treating pulmonary disorders with liposomal amikacin formulations
US7931521B1 (en) 2007-05-16 2011-04-26 Sheila K Griffin Adjustable bra strap
UA27298U (en) 2007-06-13 2007-10-25 Method for preventing pleural empyema after pneumonectomy
UA27804U (en) 2007-07-26 2007-11-12 Method for preventing respiratory complications after surgery in lungs and pleura
EP2030644A1 (en) 2007-08-31 2009-03-04 PARI Pharma GmbH Aerosols for sinunasal drug delivery
NZ562236A (en) 2007-10-05 2010-04-30 Univ Otago Detection of volatile compounds as markers for Mycobacteria tuberculosis
JP5855829B2 (en) 2007-10-23 2016-02-09 インスメッド, インコーポレイテッド Liposomal vancomycin preparation
DE102007056462B4 (en) 2007-11-23 2011-10-27 Pari Pharma Gmbh Disposable ampoule for a device for generating aerosols
US10201587B2 (en) 2008-04-08 2019-02-12 Melinta Therapeutics, Inc. Methods of inhibiting and treating biofilms using glycopeptide antibiotics
DE102008022987A1 (en) 2008-05-09 2009-11-12 Pari Pharma Gmbh Nebulizer for respirators and ventilator with such a nebulizer
ES2475065T3 (en) 2008-10-09 2014-07-10 Tekmira Pharmaceuticals Corporation Enhanced amino acids and methods for nucleic acid administration
JP2012522009A (en) 2009-03-26 2012-09-20 パルマトリックス,インコーポレイテッド Methods for treating and preventing pneumonia and ventilator-associated tracheobronchitis
ES2457442T3 (en) 2009-05-08 2014-04-25 Pari Pharma Gmbh Concentrated pharmaceutical formulations of mast cell stabilizers
DE102009026636B4 (en) 2009-06-02 2011-04-14 Pari Pharma Gmbh A method of welding a membrane to a carrier in the manufacture of a membrane nebulizer
US9149588B2 (en) 2009-07-17 2015-10-06 Nektar Therapeutics Systems and methods for driving sealed nebulizers
EA023860B1 (en) 2009-07-17 2016-07-29 Нектар Терапьютикс Negatively biased sealed nebulizers systems and methods
EP2457065A1 (en) 2009-07-22 2012-05-30 Koninklijke Philips Electronics N.V. Thermal flow sensor integrated circuit with low response time and high sensitivity
US20130034534A1 (en) 2009-09-29 2013-02-07 Philipp Kroneberg Method for treatment of patients with cystic fibrosis
WO2011049960A2 (en) 2009-10-21 2011-04-28 Otonomy, Inc. Compositions and methods for the treatment of sinonasal disorders
US8536220B2 (en) 2010-01-26 2013-09-17 Murray Fulgham Supplement composition and method of use
WO2011133617A1 (en) 2010-04-23 2011-10-27 The Board Of Trustees Of The University Of Illinois Nano-hybrid delivery system for sequential utilization of passive and active targeting
CN103052392A (en) 2010-07-12 2013-04-17 赛利亚医药公司 Treatment of lung infections by administration of tobramycin by aerolisation
AU2010359346B2 (en) 2010-08-20 2015-01-29 Dr. Reddy's Laboratories Sa Phospholipid depot
WO2012047674A2 (en) 2010-09-27 2012-04-12 Microdose Therapeutx, Inc. Methods and compositions for disease treatment using inhalation
EP3470057B1 (en) 2010-09-29 2021-11-03 Pulmatrix Operating Company, Inc. Cationic dry powders comprising magnesium salt
EP2457609A1 (en) 2010-11-24 2012-05-30 PARI Pharma GmbH Aerosol generator
WO2012106382A1 (en) 2011-01-31 2012-08-09 Genoa Pharmaceuticals, Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
SG195038A1 (en) 2011-05-19 2013-12-30 Savara Inc Dry powder vancomycin compositions and associated methods
EP3597644B1 (en) 2011-10-18 2021-09-29 Dicerna Pharmaceuticals, Inc. Amine cationic lipids and uses thereof
US20140308304A1 (en) 2011-12-07 2014-10-16 Alnylam Pharmaceuticals, Inc. Lipids for the delivery of active agents
WO2013093891A1 (en) 2011-12-22 2013-06-27 Nuvo Research Gmbh Liposomal chlorite or chlorate compositions
CA2870860C (en) 2012-05-21 2021-07-27 Insmed Incorporated Systems for treating pulmonary infections
US20150246137A1 (en) 2012-09-27 2015-09-03 The University Of North Carolina At Chapel Hill Lipid coated nanoparticles containing agents having low aqueous and lipid solubilities and methods thereof
RU2675859C2 (en) 2012-11-29 2018-12-25 Инсмед Инкорпорейтед Stabilised vancomycin formulations
US20160193148A1 (en) 2013-08-01 2016-07-07 University Of Georgia Research Foundation, Inc. Liposomal formulations for the treatment of bacterial infections
EP3060198A4 (en) 2013-10-22 2017-06-28 Aradigm Corporation Inhaled surfactant-modified liposomal formulations providing both an immediate and sustained release profile
JP2017510662A (en) 2014-04-08 2017-04-13 アラダイム コーポレーション Liposomal ciprofloxacin preparation with activity against nontuberculous mycobacteria
US20160120806A1 (en) 2014-04-08 2016-05-05 Aradigm Corporation Nanocrystals formed in a microenvironment
AU2015244399B2 (en) 2014-04-08 2019-10-03 Aradigm Corporation Liposomes that form drug nanocrystals after freeze-thaw
ES2926985T3 (en) 2014-05-15 2022-10-31 Insmed Inc Methods for treating nontuberculous mycobacterial lung infections
US9402345B2 (en) 2014-07-23 2016-08-02 Cnh Industrial America Llc Cotton harvester
WO2017008076A1 (en) 2015-07-09 2017-01-12 Insmed Incorporated Compositions and methods for treating lung diseases and lung injury
US20170165374A1 (en) 2015-11-18 2017-06-15 Insmed, Inc. Compositions and methods for treating bacterial infections

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10328071B2 (en) 2005-12-08 2019-06-25 Insmed Incorporated Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof
US10471149B2 (en) 2012-11-29 2019-11-12 Insmed Incorporated Stabilized vancomycin formulations
US10238675B2 (en) 2014-05-15 2019-03-26 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US10251900B2 (en) 2014-05-15 2019-04-09 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US10398719B2 (en) 2014-05-15 2019-09-03 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US10588918B2 (en) 2014-05-15 2020-03-17 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US10751355B2 (en) 2014-05-15 2020-08-25 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US10828314B2 (en) 2014-05-15 2020-11-10 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US11395830B2 (en) 2014-05-15 2022-07-26 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US11446318B2 (en) 2014-05-15 2022-09-20 Insmed Incorporated Methods for treating pulmonary non-tuberculous mycobacterial infections
US11571386B2 (en) 2018-03-30 2023-02-07 Insmed Incorporated Methods for continuous manufacture of liposomal drug products

Also Published As

Publication number Publication date
US9549925B2 (en) 2017-01-24
PT1962805T (en) 2016-10-05
US20130028960A1 (en) 2013-01-31
US9511082B2 (en) 2016-12-06
AU2006322076C1 (en) 2013-11-14
JP2009518411A (en) 2009-05-07
EP3067046A1 (en) 2016-09-14
EP3067046B1 (en) 2020-03-25
US9549939B2 (en) 2017-01-24
US8673349B2 (en) 2014-03-18
EP3718532A1 (en) 2020-10-07
US20070196461A1 (en) 2007-08-23
US8226975B2 (en) 2012-07-24
US9402845B2 (en) 2016-08-02
JP5324223B2 (en) 2013-10-23
US20130052260A1 (en) 2013-02-28
EP3067047A1 (en) 2016-09-14
PL1962805T3 (en) 2017-01-31
US8673348B2 (en) 2014-03-18
CA2631872C (en) 2014-04-01
US20160113927A1 (en) 2016-04-28
CA2896083A1 (en) 2007-06-14
US20160317563A1 (en) 2016-11-03
US20130071468A1 (en) 2013-03-21
US20160184301A1 (en) 2016-06-30
CY1118059T1 (en) 2017-06-28
US8642075B2 (en) 2014-02-04
LT1962805T (en) 2016-10-25
US20160184302A1 (en) 2016-06-30
AU2006322076B2 (en) 2013-07-11
US8632804B2 (en) 2014-01-21
WO2007067520A3 (en) 2007-12-06
US20230133762A1 (en) 2023-05-04
US20170087155A1 (en) 2017-03-30
HUE029994T2 (en) 2017-04-28
US8679532B2 (en) 2014-03-25
US20160354371A1 (en) 2016-12-08
AU2006322076A1 (en) 2007-06-14
CA2838111A1 (en) 2007-06-14
EP1962805B1 (en) 2016-07-06
US20200390758A1 (en) 2020-12-17
US20140072620A1 (en) 2014-03-13
CA2838111C (en) 2016-01-19
US20160317564A1 (en) 2016-11-03
EP1962805A2 (en) 2008-09-03
EP1962805A4 (en) 2013-01-02
CA2631872A1 (en) 2007-06-14
US10328071B2 (en) 2019-06-25
ES2798263T3 (en) 2020-12-10
WO2007067520A2 (en) 2007-06-14
DK1962805T3 (en) 2016-09-26
US20130071469A1 (en) 2013-03-21
EP3067047B1 (en) 2022-04-20
US20130064883A1 (en) 2013-03-14
ES2594368T3 (en) 2016-12-19

Similar Documents

Publication Publication Date Title
US20230133762A1 (en) Lipid-based compositions of antiinfectives for treating pulmonary infections and methods of use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: TRANSAVE, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WEERS, JEFF;REEL/FRAME:044733/0287

Effective date: 20070126

Owner name: TRANSAVE, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WEERS, JEFF;REEL/FRAME:044733/0297

Effective date: 20100506

Owner name: TRANSAVE, LLC, NEW JERSEY

Free format text: MERGER;ASSIGNOR:TRANSAVE, INC.;REEL/FRAME:044733/0292

Effective date: 20101201

Owner name: INSMED INCORPORATED, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TRANSAVE, LLC;REEL/FRAME:044733/0301

Effective date: 20120615

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