US20200383908A1 - Cromolyn compositions and methods thereof - Google Patents

Cromolyn compositions and methods thereof Download PDF

Info

Publication number
US20200383908A1
US20200383908A1 US16/733,667 US202016733667A US2020383908A1 US 20200383908 A1 US20200383908 A1 US 20200383908A1 US 202016733667 A US202016733667 A US 202016733667A US 2020383908 A1 US2020383908 A1 US 2020383908A1
Authority
US
United States
Prior art keywords
cromolyn
solution
microns
pharmaceutically acceptable
disease
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/733,667
Inventor
David R. Elmaleh
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.)
General Hospital Corp
Original Assignee
General Hospital Corp
Aztherapies 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
Priority claimed from PCT/US2014/039118 external-priority patent/WO2015002703A1/en
Application filed by General Hospital Corp, Aztherapies Inc filed Critical General Hospital Corp
Priority to US16/733,667 priority Critical patent/US20200383908A1/en
Assigned to AZ THERAPIES reassignment AZ THERAPIES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ELMALEH, DAVID R.
Assigned to ELMALEH, DAVID R. reassignment ELMALEH, DAVID R. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AZ THERAPIES, INC.
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ELMALEH, DAVID R.
Priority to US17/108,895 priority patent/US11013686B2/en
Publication of US20200383908A1 publication Critical patent/US20200383908A1/en
Priority to US17/972,772 priority patent/US20230248646A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0075Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a dry powder inhaler [DPI], e.g. comprising micronized drug mixed with lactose carrier particles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • 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
    • A61M15/00Inhalators
    • A61M15/0085Inhalators 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
    • A61M16/00Devices for influencing the respiratory system of patients by gas treatment, e.g. mouth-to-mouth respiration; Tracheal tubes
    • A61M16/10Preparation of respiratory gases or vapours
    • A61M16/14Preparation of respiratory gases or vapours by mixing different fluids, one of them being in a liquid phase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61M2202/00Special media to be introduced, removed or treated
    • A61M2202/06Solids
    • A61M2202/064Powder

Definitions

  • the invention relates generally to cromolyn compositions and methods thereof. Specifically, the invention relates to effectively delivering cromolyn particles to a patient in need thereof for treating various diseases.
  • Cromolyn also known as cromoglicic acid, cromoglycate, or cromoglicate
  • cromolyn sodium also known as disodium cromoglycate or DSCG
  • Cromolyn demonstrates poor oral absorption. Delivery of cromolyn via inhalation has proven inefficient and difficult due, at least in part, to the hygroscopic nature of cromolyn sodium. For example, micronized powders containing cromolyn sodium particles spontaneously absorb water, forming clumps that impair efficient delivery of the cromolyn powder. See Keller et al. Expert Opin. Drug Deliv. 8, 1-17 (2011).
  • the present invention provides improved compositions and methods for delivering cromolyn via inhalation, efficiently and consistently over a range of inspiratory flow rates.
  • the invention is directed to a method of delivering cromolyn to a patient in need thereof.
  • patients include patients in need of systemic delivery of cromolyn, e.g., to the brain or other non-lung tissues.
  • the patient has an amyloid-associated condition.
  • Patients in need of cromolyn also include patients in need of pulmonary delivery of cromolyn, for lung or airway related conditions.
  • the patient has an inflammatory or allergic lung disease, such as asthma.
  • the method comprises administering to the patient via oral inhalation a pharmaceutically acceptable salt or ester of cromolyn in the form of a powder comprising particles of the pharmaceutically acceptable salt or ester of cromolyn.
  • the majority of particles may have a diameter of about 2 to about 5 microns.
  • the powder is administered using a device that deposits (a) at least 1.5 mg and (b) at least 20% by weight of the administered amount of pharmaceutically acceptable salt or ester of cromolyn to Stage 4 and higher of a Next Generation Pharmaceutical Impactor (NGf) cascade impactor device at a flow rate of, for example, 20-90 L/min.
  • NGf Next Generation Pharmaceutical Impactor
  • the invention also provides a method of delivering cromolyn to a patient in need thereof, comprising administering to the patient via oral inhalation a pharmaceutically acceptable salt or ester of cromolyn in the form of a powder, under conditions such that (a) at least 1.5 mg and (b) at least 20% by weight of the administered amount of the pharmaceutically acceptable salt or ester of cromolyn is delivered to the lower airways of the patient.
  • a pharmaceutically acceptable salt or ester of cromolyn in the form of a powder, under conditions such that (a) at least 1.5 mg and (b) at least 20% by weight of the administered amount of the pharmaceutically acceptable salt or ester of cromolyn is delivered to the lower airways of the patient.
  • the term “lower airways” refers to the region of the airways/lung that corresponds to Stage 4 and higher of a Next Generation Pharmaceutical Impactor (NGI) cascade impactor device.
  • NTI Next Generation Pharmaceutical Impactor
  • the invention also provides a method of delivering cromolyn to a patient in need thereof, comprising administering to the patient via oral inhalation a pharmaceutically acceptable salt or ester of cromolyn in the form of a powder comprising particles of the pharmaceutically acceptable salt or ester of cromolyn using a dry powder inhaler (DPI) device comprising a chamber comprising a piezoelectric vibrator or an ultrasonic vibrator for deaggregating a powder and an air flow passageway in which the deaggregated powder is picked up and carried for oral inhalation by a patient.
  • DPI dry powder inhaler
  • the majority of the particles of the pharmaceutically acceptable salt or ester of cromolyn may have a diameter of about 2 to about 5 microns.
  • the pharmaceutically acceptable salt or ester of cromolyn is administered to the patient.
  • the pharmaceutically acceptable salt or ester of cromolyn is administered with one or more pharmaceutically acceptable excipients.
  • the powder comprises about 0.1% to about 80%, for example, about 40% to about 80% by weight of an excipient.
  • the excipient is a monosaccharide, a disaccharide, an oligosaccharide, a polysaccharide, or a polyalcohol, such as lactose, mannitol, or sorbitol.
  • the invention provides a method of treating an amyloid-associated condition in a patient in need thereof.
  • the method comprises administering to the patient via oral inhalation an amount of powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn via pulmonary delivery, the majority of particles having a diameter of about 2 to about 5 microns, using a device that deposits (a) at least 1.5 mg and (b) at least 20% by weight of the administered amount of pharmaceutically acceptable salt or ester of cromolyn to Stage 4 and higher of a Next Generation Pharmaceutical Impactor (NGI) cascade impactor device at a flow rate of, for example, 20-90 L/min (e.g., for about 4 seconds).
  • NTI Next Generation Pharmaceutical Impactor
  • the invention further provides a method of treating inflammatory or allergic lung diseases in a patient in need thereof.
  • the method comprises administering to the patient via oral inhalation an amount of powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn, the majority of particles having a diameter of about 2 to about 5 microns, at a frequency of 1 or 2 times daily, each dose comprising about 3 mg to about 20 mg of a pharmaceutically acceptable salt or ester of cromolyn.
  • the dose and/or frequency of delivery according to such methods are reduced compared to the conventional dosages and frequencies.
  • the powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn is administered to a patient using an active dry powder inhaler, such as a dry powder inhaler comprising a chamber comprising a piezoelectric vibrator or an ultrasonic vibrator for deaggregating a dry powder and an air flow passageway in which the deaggregated powder is picked up and carried for oral inhalation by a patient.
  • an active dry powder inhaler such as a dry powder inhaler comprising a chamber comprising a piezoelectric vibrator or an ultrasonic vibrator for deaggregating a dry powder and an air flow passageway in which the deaggregated powder is picked up and carried for oral inhalation by a patient.
  • the invention is directed to a method for treating an amyloid-associated condition in a patient in need thereof, comprising administering to the patient via oral inhalation an amount of liquid particles of a solution comprising a pharmaceutically acceptable salt or ester of cromolyn using a nebulizer.
  • the particles have a mass median aerodynamic diameter (MMAD) of about 0.5 to about 15 microns. In some embodiments, the majority of particles have a diameter of about 2 microns to about 5 microns.
  • MMAD mass median aerodynamic diameter
  • the invention further includes use of a pharmaceutically acceptable salt or ester of cromolyn in the preparation of a medicament for treating inflammatory or allergic lung diseases in a patient in need thereof in an amount from about 3 mg to about 20 mg (e.g., from about 16.1 mg to about 19.9 mg).
  • the medicament is administered via oral inhalation at a frequency of 1 or 2 times daily, and the pharmaceutically acceptable salt or ester of cromolyn is in the form of a powder comprising particles of pharmaceutically acceptable salt or ester of cromolyn optionally having a diameter of about 0.5 to about 15 microns, preferably an average particle size of about 5 microns or less.
  • the invention further contemplates use of a pharmaceutically acceptable salt or ester of cromolyn in the preparation of a medicament for treating an amyloid-associated condition in a patient in need thereof.
  • the medicament is administered to the patient via pulmonary delivery or oral inhalation.
  • the pharmaceutically acceptable salt or ester of cromolyn is in the form of a powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn, wherein the majority of particles having a diameter of about 2 to about 5 microns.
  • the medicament is optionally delivered using a dry powder inhaler device as described herein.
  • the pharmaceutically acceptable salt or ester of cromolyn is in solution and administered using a nebulizer.
  • the invention provides a blister pack for delivering cromolyn to a patient in need thereof.
  • the blister pack comprises blisters containing about 3 mg to about 20 mg (e.g., about 16.1 mg to about 19.9 mg) of a pharmaceutically acceptable salt or ester of cromolyn.
  • the invention also is directed to a kit comprising a blister pack as described herein and a dry powder inhaler (DPI) device.
  • the device is an active dry powder inhaler, such as a dry powder inhaler device comprising a chamber comprising a piezoelectric vibrator for deaggregating a dry powder and an air flow passageway in which the deaggregated powder is picked up and carried for oral inhalation by a patient.
  • the kit further comprises ibuprofen tablets.
  • the invention includes, as an additional aspect, all embodiments of the invention narrower in scope in any way than the variations specifically mentioned above. With respect to aspects of the invention described as a genus, all individual species are individually considered separate aspects of the invention. With respect to elements described as a selection within a range, it should be understood that all discrete subunits within the range are contemplated as an embodiment of the invention. Ranges may be expressed herein as from “about” or “approximately” one particular value and/or to “about” or “approximately” another particular value. When such a range is expressed, another embodiment according to the invention includes from the one particular value and/or to the other particular value. Similarly, when particular values are expressed as approximations, but use of antecedents such as “about,” “at least about,” or “less than about,” it will be understood that the particular value forms another embodiment.
  • the invention provides improved methods and compositions for the efficient and consistent delivery of cromolyn via inhalation. Such methods preferably deliver a consistent amount of drug over a wide range of patient inspiratory flow rates. According to some aspects of the inventive method, a large portion of an administered amount of cromolyn is delivered to regions of the lungs that mediate transport into systemic circulation (the bronchi, bronchioles, and alveoli), e.g. the lower airways. Thus, the inventive method can provide an effective means for delivering cromolyn systemically, i.e., into the blood stream (and, by extension, to other non-lung regions of the body, such as the brain).
  • the enhanced delivery efficiency associated with the inventive method allows administration of lower doses of cromolyn and/or less frequent administration of cromolyn, to achieve a desired biological response in any condition requiring lung delivery or systemic delivery.
  • Advantages include improved therapeutic efficacy at conventional doses, or maintained/improved therapeutic efficacy at lower doses and/or lower frequencies of administration, leading to improved ease of use, higher patient compliance, and improved therapeutic benefit, as well as cost savings associated with using reduced amounts of drug.
  • many drug packaging systems containing groups of individual delivery units, such as blister packs or capsules have maximum capacities for a single dose of drug contained within individual delivery units. More efficient delivery of cromolyn also advantageously facilitates the packaging of a therapeutically effective dose of cromolyn in each individual delivery unit.
  • Advantages include delivery of a higher dose per single administration, delivery of uniform doses in a multiple administration regimen (i.e., little variability between doses), and/or use of a single individual delivery unit per administration rather than multiple individual delivery units per administration.
  • the invention provides a method of delivering cromolyn to a patient in need thereof.
  • Such patients include patients in need of lung or systemic delivery of cromolyn, and include patients with amyloid-associated conditions as well as patients with an inflammatory or allergic lung disease such as asthma.
  • cromolyn also known as cromoglicic acid, cromoglycate, cromoglicate, or 5,5′-(2-hydroxypropane-1,3-diyl)bis(oxy)bis(4-oxo-4H-chromene-2-carboxylic acid)
  • cromolyn also known as cromoglicic acid, cromoglycate, cromoglicate, or 5,5′-(2-hydroxypropane-1,3-diyl)bis(oxy)bis(4-oxo-4H-chromene-2-carboxylic acid
  • cromolyn also known as cromoglicic acid, cromoglycate, cromoglicate, or 5,5′-(2-hydroxypropane-1
  • Pharmaceutically acceptable salts are well known to those skilled in the art and include pharmaceutically acceptable inorganic and organic base addition salts, which may be formed with metals or amines, such as alkali and alkaline earth metals or organic amines.
  • Pharmaceutically acceptable salts of compounds may also be prepared with a pharmaceutically acceptable cation. These salts can be prepared in situ in the administration vehicle or the dosage form manufacturing process, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation, with ammonia, or with a pharmaceutically-acceptable organic primary, secondary or tertiary amine.
  • Suitable pharmaceutically acceptable cations are well known to those skilled in the art and include alkaline, alkaline earth, ammonium and quaternary ammonium cations.
  • metals used as cations are lithium, sodium, potassium, magnesium, ammonium, calcium, aluminum, or ferric, and the like.
  • suitable amines include ethylamine, diethylamine, piperazine, isopropylamine, trimethylamine, histidine, N,N′-dibenzylethylenediamine, chloroprocaine, choline, ethanolamine, diethanolamine, dicyclohexylamine, ethylenediamine, N-methylglucamine, and procaine.
  • Suitable esters of cromolyn include, but are not limited to, carboxylate esters of one or both of the carboxylic acids of cromolyn, such as aliphatic esters (e.g., methyl esters, ethyl esters, propyl esters, butyl esters such as t-butyl esters, and pentyl esters), aryl esters (e.g., phenyl esters and benzyl esters), and combinations thereof.
  • the pharmaceutically acceptable salt of cromolyn is disodium cromoglycate.
  • the pharmaceutically acceptable salt or ester of cromolyn is administered via inhalation, generally via oral inhalation, however, nasal inhalation or a combination of oral and nasal inhalation can also be used.
  • administration via inhalation as described herein delivers cromolyn to the lungs of the patient, depositing the pharmaceutically acceptable salt or ester of cromolyn onto surfaces of the lung that allow absorption into the blood stream (e.g., the bronchi, bronchioles, and alveoli), e.g. the lower airways.
  • the particles of the cromolyn salt or ester have a median particle diameter (D 50 ) of less than about 5 microns, less than about 4.8 microns, less than about 4.5 microns, less than about 4.2 microns, less than about 4 microns, less than about 3.8 microns, less than about 3.5 microns, less than about 3.2 microns, less than about 3 microns, less than about 2.8 microns, less than about 2.5 microns, less than about 2 microns, less than about 1.8 microns, less than about 1.5 microns, less than about 1.2 microns, less than about 1 micron, about 0.1 microns to about 5 microns, about 0.5 microns to about 5 microns, about 0.8 microns to about 5 microns, about 1 micron to about 5 microns, about 1.2 microns to about 5 microns, about 1.5 microns to about 5 microns, about 1.8 microns to about 5 microns, about 2
  • the particles of the cromolyn salt or ester optionally have a D 90 of less than 15 microns, less than 14 microns, less than 13 microns, less than 12 microns, less than 11 microns, less than 10 microns, less than 9.5 microns, less than 9 microns, less than 8.5 microns, less than 8 microns, less than 7.5 microns, less than 7 microns, less than 6.5 microns, less than 6 microns, less than 5.5 microns, less than 5 microns, less than 4.5 microns, less than 4 microns, less than 3.9 microns, less than 3.8 microns, less than 3.7 microns, less than 3.6 microns, about 3 microns to about 15 microns, about 5 microns to about 13 microns, about 7 microns to about 11 microns, and/or about 9 microns to about 10 microns.
  • the term D 50 refers to a diameter at which 50% of a sample's mass is comprised of smaller particles.
  • D 90 refers to a diameter at which 90% of a sample's mass is comprised of smaller particles.
  • the D 50 is also known as the “mass median diameter” as it divides the sample equally by mass.
  • the particles of the cromolyn salt or ester have a mass median aerodynamic diameter (MMAD) of about 0.5 to about 15 microns, about 0.5 to about 10 microns, about 1 to about 5 microns, about 1 to about 4 microns, about 1 to about 3.5 microns, about 1 to about 3 microns, about 1 to about 2.5 microns, and/or about 1 to about 2 microns.
  • MMAD mass median aerodynamic diameter
  • the term “aerodynamic diameter” refers to the diameter of a sphere of unit density that reaches the same velocity in an air stream as a non-spherical particle of arbitrary density.
  • the term “mass median aerodynamic diameter” refers to the aerodynamic diameter at which 50% of the particles by mass are larger and 50% are smaller. Methods of determining aerodynamic diameter are known in the art and described in, e.g., Chow et al., “Particle Engineering for Pulmonary Drug Delivery,” Pharm. Res., 24 (3), 411-437 (2007).
  • the particles of the cromolyn salt or ester may have a geometric standard deviation (GSD) of about 1.3 to about 2.5, about 1.4 to about 2.4, about 1.5 to about 2.3, about 1.6 to about 2.2, about 1.7 to about 2.1, and/or about 1.8 to about 2.
  • GSD geometric standard deviation
  • the powder optionally comprises one or more pharmaceutically acceptable excipients.
  • Suitable excipients are well tolerated by pulmonary tissue, and include, but are not limited to, monosaccharides, disaccharides, oligosaccharides, polysaccharides, polyalcohols, and combinations thereof.
  • Exemplary excipients include, but are not limited to, lactose, mannitol, sorbitol, and combinations thereof.
  • the pharmaceutically acceptable excipient(s), when present, are included in the powder in a total amount of about 0.1% to about 80% by weight, about 1% to about 80% by weight, about 5% to about 80% by weight, about 10% to about 80% by weight, about 15% to about 80% by weight, about 20% to about 80% by weight, about 25% to about 80% by weight, about 30% to about 80% by weight, about 35% to about 80% by weight, about 40% to about 80% by weight, about 20 to about 75% by weight, about 20% to about 70% by weight, about 20% to about 65% by weight, about 20% to about 60% by weight, about 25% to about 55% by weight, about 30% to about 50% by weight, about 35% to about 45% by weight, and/or about 40% by weight.
  • the pharmaceutically acceptable excipients can be included in an anhydrous form or as a hydrate, such as a monohydrate or higher-order hydrate.
  • the excipient(s) optionally have a particle size of about 250 microns or less, such as about 10 microns to about 150 microns, although other excipient particle sizes may be used in the context of the invention.
  • the amount of powder that is administered to a patient comprises about 3 mg to about 20 mg of the pharmaceutically acceptable salt or ester of cromolyn, for example, about 3 mg to about 19 mg, about 3 mg to about 18 mg, about 3 mg to about 17 mg, about 3 mg to about 16 mg, about 3 mg to about 15 mg, about 3 mg to about 14 mg, about 3 mg to about 13 mg, about 3 mg to about 12 mg, about 3 mg to about 11 mg, about 3 mg to about 10 mg, about 3 mg to about 9 mg, about 3 mg to about 8 mg, about 3 mg to about 7 mg, about 4 mg to about 20 mg, about 6 mg to about 19 mg, about 8 mg to about 18 mg, about 10 mg to about 18 mg, about 12 mg to about 18 mg, about 14 mg to about 18 mg, about 16 mg to about 18 mg, about 17 mg to about 18 mg, about 16.1 mg
  • the amount of powder is optionally administered as a single dose or administration, which may be inhaled in a single breath or span multiple breaths in the course of the single administration.
  • the single dose or administration is administered from a single individual delivery unit, such as a single blister or single capsule.
  • This single dose may be administered repeatedly to the patient at any interval over the course of a treatment period.
  • a single dose of pharmaceutically acceptable salt or ester of cromolyn e.g., about 16.1 mg to about 19.9 mg
  • Treatment periods include at least 1, 2, 3, 4, 5, 6, or 7 days, or at least 1, 2, 3, or 4 weeks, or at least 1, 2, 3, 4, 5 or 6 months, or a year or more.
  • administering the powder involves suspending the powder into a gas (such as air or oxygen), thereby forming an aerosol containing the powder and the gas. Concurrently with, or subsequent to, suspension formation, the powder suspension is inhaled by a patient. In some embodiments, the powder is suspended in a gas stream being inhaled by a patient such that administering comprises suspending the powder into an inhaled gas stream.
  • a gas such as air or oxygen
  • Suspending the powder into a gas may be carried out by any means, including vibration.
  • Vibrating the powder generally aerosolizes at least a portion (i.e., at least about 10%, at least about 30%, at least about 50%, at least about 75%, and/or at least about 90%) of the powder.
  • vibrating the powder generally involves suspending at least a portion of the powder in the gas that is in the immediate vicinity of the powder.
  • the powder comprising particles of cromolyn and optional pharmaceutically acceptable excipient(s) can include aggregates, for example, aggregates between two or more cromolyn particles and/or between cromolyn particles and excipient particles.
  • vibrating the powder deagglomerates particle aggregates, advantageously providing smaller particles that are more readily delivered regions of the lungs allowing transport into the blood stream (e.g., the bronchi, bronchioles, and alveoli), e.g. the lower airways.
  • the vibrator is vibrated such that the resulting vibrations generate synthetic jets that aerosolize and/or deaggregate the powder.
  • Methods for forming synthetic jets are described, for example, in U.S. Pat. No. 7,318,434, which is incorporated herein by reference in its entirety.
  • administering the powder involves vibrating the powder at high frequency, for example, a frequency of about 10 kHz to about 50 kHz, about 15 kHz to about 40 kHz, and/or about 20 kHz to about 30 kHz.
  • the amount of powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn is administered using a device that deposits (a) at least 1.5 mg and (b) at least 20% by weight of the administered amount of pharmaceutically acceptable salt or ester of cromolyn to Stage 4 and higher of a Next Generation Pharmaceutical Impactor (NGI) cascade impactor device at a flow rate of, for example, 20-90 L/min for about 1 to about 10 seconds (e.g., for about 4 seconds).
  • NTI Next Generation Pharmaceutical Impactor
  • the device deposits (a) at least 1 mg, at least 1.5 mg, at least 1.8 mg, at least 2 mg, at least 2.2 mg, at least 2.5 mg, at least 2.8 mg, at least 3 mg, at least 3.2 mg, at least 3.5 mg, at least 3.8 mg, at least 4 mg, at least 4.2 mg, at least 4.5 mg, at least 4.8 mg, at least 5 mg, at least 5.2 mg, at least 5.5 mg, at least 5.8 mg, at least 6 mg, at least 6.2 mg, at least 6.5 mg, at least 6.8 mg, at least 7 mg, at least 7.2 mg, at least 7.5 mg, at least 7.8 mg, at least 8 mg, at least 8.2 mg, at least 8.5 mg, at least 8.8 mg, at least 9 mg, at least 9.2 mg, at least 9.5 mg, at least 9.8 mg, at least 10 mg, about 1.3 mg to about 7 mg, about 1.5 mg to about 3 mg, about 1.5 mg to about 5 mg, about 1.5 mg to about 7 mg, about 1 mg to about 4
  • the term “administered amount” in the phrase “% by weight of the administered amount” refers to the amount of pharmaceutically acceptable salt or ester of cromolyn present in the individual delivery unit (e.g., blister or capsule or other unit dose container) connected to the device.
  • the device delivers at least about 4 to about 5 mg of cromolyn salt or ester to Stage 4 and higher of an NGI cascade impactor as described herein from a capsule or blister comprising, for example about 17.1 mg, of cromolyn salt or ester with or without added excipients.
  • NGI cascade impactors are useful for studying aerodynamic size distribution of aerosols and simulating delivery to different regions of the lung.
  • the respiratory tract forms a particle size-selective system wherein progressively finer particles pass through mouth, larynx and larger airways to be deposited in the peripheral lung (e.g., the alveolar spaces).
  • cascade impactors include several “stages” allowing passage of progressively finer particles through the system.
  • An aerosol stream carrying particles passes through each stage at a constant air flow (e.g., 20-90 L/min).
  • Particles of similar aerodynamic size deposit at a particular stage; smaller particles are deposited at further stages of the NGI cascade impactor than larger particles.
  • Stages 4 and higher of the NGI cascade impactor trap particles having a mass median aerodynamic diameter (MMAD) of about 5 microns or less.
  • the NGI cascade impactor is generally operated under conditions of low relative humidity, such as 20% relative humidity or less. Deposition in the central (bronchial) region of the lung generally peaks with particles having an MMAD of about 4 to about 6 microns, and deposition in the peripheral (alveolar) region of the lung generally peaks with particles having an MMAD of about 2 to about 4 microns.
  • Stages 4 and higher of the NGI cascade impactor at a flow rate of 70-90 L/min (e.g., for about 4 seconds of total actuation) simulate the regions of the lung providing significant systemic absorption of a medicament into the bloodstream.
  • NGI cascade impactor devices are available from MSP Corporation, Shoreview, Minn.
  • the inventive methods for delivering cromolyn allow consistent delivery of the active agent over a wide range of flow rates.
  • a consistent amount of powder is delivered to a patient over an inhalation flow rate of 20 L/min to 100 L/min, or 20 to 80 L/min, 20 to 60 L/min, 40-80 L/min, 60-80 L/min, 60-90 L/min, 70-90 L/min, or 75-85 L/min.
  • Consistent amount is meant a relative standard deviation (RSD) of 10% or less (e.g., 5% or less, 3% or less and/or 2.5% or less) in the amount powder that is received by the patient.
  • RSS relative standard deviation
  • the devices for administering the powder described herein include, but are not limited to, a dry powder inhaler (DPI) device, a metered dose inhaler (MDI) device, and a dry powder nebulizer (DPN) device.
  • DPI dry powder inhaler
  • MDI metered dose inhaler
  • DPN dry powder nebulizer
  • Suitable devices typically administer a metered dose or a predetermined dose. Such doses generally are administered over a set period of time or a set volume of air.
  • the device functions independent of the patient's inspiratory flow rate.
  • the device delivers a consistent amount of cromolyn over a wide range of flow rates, such as an inhalation flow rate of 20 L/min to 100 L/min, or 20 to 80 L/min, 20 to 60 L/min, 40-80 L/min, 60-80 L/min, 60-90 L/min, 70-90 L/min, or 75-85 L/min.
  • the device delivers a consistent amount of cromolyn at an inhalation flow rate of 80 L/min.
  • the device is a dry powder inhaler comprising a chamber in fluid communication with an air flow passageway.
  • a vibrator e.g., a piezoelectric vibrator or an ultrasonic vibrator, suitable for aerosolizing and/or deaggregating a dry powder.
  • the inhaler optionally includes a lever for activating the piezoelectric or ultrasonic vibrator and for cooperating with a means for puncturing a blister or opening means for opening a blister.
  • powder is delivered from the inhaler by receiving a pack (e.g., capsule or blister) containing the powder in an operating position proximal to the piezoelectric or ultrasonic vibrator.
  • a pack e.g., capsule or blister
  • the patient places the mouthpiece of the inhaler into the mouth, forms a seal with his/her lips around the mouthpiece, and withdraws air through the air flow passageway while pressing the lever.
  • the pack Upon activation by the lever or other triggering event, the pack is broken, the piezoelectric or ultrasonic vibrator is activated, and the deaggregated powder is picked up into the inspiratory air flow of the patient and carried through the air flow passageway for inhalation by the patient.
  • the inhaler optionally includes an inhalation flow rate detector which detects the movement of air through the air flow passageway.
  • the event that triggers release of the powder includes movement of air through the air flow passageway above a threshold rate.
  • the powder is delivered in a period of time of, for example, about one second to about five seconds of total inhalation time, for example, about one second to about two seconds.
  • the inhalation period can be monitored using a visual indicator, e.g., flashing or changing the color of a light emitting diode, or an audible indicator.
  • the lever, inhalation flow rate, and piezoelectric or ultrasonic vibrator functions are monitored and recorded in memory, such as a flash drive or other computer memory in direct contact with the inhaler or removed from the inhaler but in signal communication with the inhaler.
  • the functional parameters of the device may be transmitted from the device to a remote location, recorded in memory, and made available to healthcare providers via a wired or wireless communication network.
  • the information also optionally may be delivered to point of care devices for monitoring treatment.
  • the recorded performance information allows health care practitioners to monitor the use of the device, which can be particularly beneficial when a patient's ability to operate the device diminishes due to declining cognitive and/or physical health.
  • the invention also includes a method comprising administering cromolyn to a patient using a nebulizer, preferably in a method for treating an amyloid-associated condition in a patient in need thereof.
  • the method comprises administering liquid particles of a solution comprising a pharmaceutically acceptable salt or ester of cromolyn using a nebulizer via inhalation (e.g., oral inhalation) by the patient.
  • solution particles are droplets of solution in which the pharmaceutically acceptable salt or ester of cromolyn and any optional excipients are dissolved.
  • the pharmaceutically acceptable salt or ester of cromolyn can be present in the solution at a concentration of about 1 mg/mL to about 100 mg/mL, for example, about 5 mg/mL to about 50 mg/mL, and/or about 10 mg/mL to 20 mg/mL.
  • excipient(s) can be present in the solution at a concentration of about 1 mg/mL to about 100 mg/mL, for example, about 5 mg/mL to about 50 mg/mL, and/or about 10 mg/mL to 20 mg/mL.
  • the solution particles i.e., droplets
  • MMAD mass median aerodynamic diameter
  • the solution particles have a mass median aerodynamic diameter (MMAD) of about 1 micron to about 10 microns, for example, about 1 micron to 8 micron, about 1 micron to 4 micron, about 1 micron to about 3.5 microns, about 1 micron to about 3 microns, about 1 micron to about 2.5 microns, about 1 micron to about 2 microns, about 2 microns to about 4 microns, and/or about 2.5 microns to about 3.5 microns.
  • MMAD mass median aerodynamic diameter
  • the solution particles also optionally comprise a median diameter of less than about 2 microns, less than about 1.8 microns, less than about 1.5 microns, less than about 1.2 microns, less than about 1 micron, about 0.5 microns to about 2 microns, about 0.8 microns to about 2 microns, about 1 micron to about 2 microns, about 1.2 microns to about 2 microns, about 1.5 microns to about 2 microns, and/or about 1.8 microns to about 2 microns.
  • Excipients, dosage, target regions of the lung, delivery amounts and efficiencies, and methods of estimating delivery to target regions of the lung are described above.
  • Suitable devices for administering cromolyn as a solution include, but are not limited to, a jet, ultrasonic or electronic nebulizers.
  • nebulizer devices form aerosolized particles of cromolyn (e.g., a pharmaceutically acceptable salt or ester of cromolyn, such as cromolyn sodium) from a pharmaceutically acceptable solution (e.g., a hypotonic or isotonic solution).
  • the nebulizer device optionally includes a liquid reservoir separated from a mouthpiece or face mask by a vibrating mesh, a piezoelectric element, or a compressed gas atomizer element.
  • a solution comprising a pharmaceutically acceptable salt or ester of cromolyn is present in the liquid reservoir, and the device produces a mist of particles for oral inhalation.
  • the cromolyn solution is optionally delivered to the patient via inhalation (oral, nasal, or a combination thereof) over the course of about ten minutes or less (e.g., two minutes).
  • the nebulizer may have any of the features of dry powder administration devices described herein.
  • the device deposits/delivers at least 1 mg, at least 1.5 mg, at least 1.8 mg, at least 2 mg, at least 2.2 mg, at least 2.5 mg, at least 2.8 mg, at least 3 mg, at least 3.2 mg, at least 3.5 mg, at least 3.8 mg, at least 4 mg, at least 4.2 mg, at least 4.5 mg, at least 4.8 mg, at least 5 mg, at least 5.2 mg, at least 5.5 mg, 5.8 mg, at least 6 mg, at least 6.2 mg, at least 6.5 mg, at least 7.8 mg, at least 7 mg, at least 7.2 mg, at least 7.5 mg, 7.8 mg, at least 8 mg, at least 8.2 mg, at least 8.5 mg, at least 8.8 mg, at least 9 mg, at least 9.2 mg, at least 9.5 mg, at least 9.8 mg, at least 10 mg, about 1.3 mg to about 7 mg, about 1.5 mg to about 3 mg, about 1.5 mg to about
  • the device deposits/delivers at least 20%, at least 30%, at least 35%, at least 40%, at least 45%, about 30% to about 75%, about 30% to about 65%, about 30% to about 60%, about 30% to about 55%, and/or about 30% to about 50% by weight of the administered amount of pharmaceutically acceptable salt or ester of cromolyn (as defined above) to Stage 4 and higher of an NGI cascade impactor device at a flow rate of, for example, about 20 L to about 90 L/min (e.g., for about 10 seconds or less).
  • Nebulizers also are described in U.S. Pat. No. 8,263,645 and U.S. Patent Application Publication Nos. 2007/0193577, 2009/0118249, and 2012/0118991, which are incorporated by reference in their entireties.
  • any of the methods described herein include a method of treating an amyloid-associated condition in a patient in need thereof.
  • the method comprises administering to the patient via inhalation an amount of powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn as described herein.
  • the method comprises administering to the patient via inhalation an amount of particles of a solution comprising a pharmaceutically acceptable salt or ester of cromolyn using a nebulizer.
  • disodium cromolyn penetrates the blood-brain barrier to bind beta-amyloid peptides and inhibit association of amyloid peptides and/or polymerization into oligomers and higher order aggregates.
  • the inhibition of beta-amyloid association and/or polymerization is believed to arrest amyloid-mediated impairment of neurons and may enhance removal of oligomers and/or aggregates from the brain, thereby slowing, stopping or reducing amyloid accumulation in the brain.
  • amyloid-associated condition is meant a disease or pathological disorder associated with abnormal amyloid aggregation and/or accumulation. See Eisenberg and Jucker, Cell, 148, 1188-1203 (2012). Amyloid accumulation has been linked to a wide variety of disorders ranging from neurodegenerative disorders to systemic amyloidoses to arthritis.
  • amyloid-associated conditions include, but are not limited to, Alzheimer's disease (beta amyloid), dementia, diabetes mellitus type 2 (IAPP), Parkinson's disease (alpha-synuclein), idiopathetic myeloma, amyloid polyneuropathy, amyloid cardiomyopathy, systemic senile amyloidosis, transmissible spongiform encephalopathy (e.g., Creutzfeldt-Jakob disease) (PrPSc), Down's syndrome, Huntington's Disease (Huntingtin), medullary carcinoma of the thyroid (calcitonin), atherosclerosis (apolipoprotein), rheumatoid arthritis (serum amyloid A), aortic medial amyloid (Medin), prolactinomas (prolactin), familial amyloid polyneuropathy (FAP) (transthyretin), frontemporal lobar degeneration-tau (Tau), Familial British and Danish
  • amyloid-associated conditions include head injuries (traumatic brain injury); amyloid- ⁇ plaques have been found in patients within hours following traumatic brain injury. See Johnson et al., Nature Reviews Neuroscience, 11, 361-370 (2010).
  • treating and “treatment” refers to any reduction in the severity and/or onset of signs or symptoms associated with an ailment, such as an amyloid-associated condition or an inflammatory or allergic lung disease (described below). Accordingly, “treating” and “treatment” includes therapeutic and prophylactic measures that reduce any of the signs or symptoms.
  • a subject such as a human patient. For example, a reduction in signs such as detectable amyloid plaques can be beneficial.
  • an increase in soluble amyloid levels in the cerebrospinal fluid from abnormally low levels to normal, age-appropriate levels can also be beneficial.
  • the quality of life of a patient is improved by reducing to any degree the severity of symptoms in a subject and/or delaying the appearance of symptoms.
  • the method in one aspect is performed as soon as possible after it has been determined that a subject is at risk for an amyloid-associated condition or as soon as possible after the amyloid-associated condition manifests in the subject.
  • prognosis may be improved by initiating treatment as soon as possible after the occurrence of the head injury.
  • Amyloid deposits can be detected ex vivo using, e.g., fluorescent dyes, circular dichroism, and X-ray diffraction.
  • fluorescent dyes e.g., fluorescent dyes, circular dichroism, and X-ray diffraction.
  • amyloids in biopsied tissue typically demonstrate green birefringence when stained with Congo red and viewed under a polarizing microscope.
  • Amyloid plaques can be detected in vivo using, e.g., magnetic resonance imaging (MRI) (see, e.g., Baltes et al., Methods Mol Biol., 711, 511-33 (2011)) and positron emission tomography (PET) (Kepe et al., Methods Enzymol., 412, 144-60 (2006)).
  • MRI magnetic resonance imaging
  • PET positron emission tomography
  • Many of the neurodegenerative diseases referenced herein also are diagnosed by conducting mental status and neuropsychological tests (assessing, e.g., memory and problem solving) and blood tests (e.g., blood tests for Alzheimer's disease biomarker signatures). See Burnham et al., “A blood-based predictor for neocortical A ⁇ burden in Alzheimer's disease: results from the AIBL study,” Molecular Psychiatry (April 2013).
  • the invention provides methods of treating inflammatory or allergic lung diseases in a patient in need thereof, comprising administering to the patient via inhalation an amount of powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn as described herein.
  • the dose and/or frequency of administration according to such methods are reduced compared to the conventional dosages and frequencies.
  • Inflammatory or allergic lung diseases include asthma, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), pulmonary fibrosis, cystic fibrosis.
  • COPD chronic obstructive pulmonary disease
  • ARDS acute respiratory distress syndrome
  • pulmonary fibrosis cystic fibrosis.
  • Asthma is a condition associated with inflammation of airway passages in the lungs and airway hyperresponsiveness (AHR). Asthma also is marked by excess mucus production. Symptoms range from minor (e.g., slight shortness of breath) to severe (wheezing, inability to breath, and/or chest tightness) and vary from person to person. During an asthma attack, the lining of airways swell, thereby constricting the passage and reducing airflow to and from the lungs.
  • Asthma is caused or triggered by, for example, infection, allergens, chemical substances and fumes, pollutants, medications, physical exertion, stress, and food additives. Asthma is classified into four general categories: mild intermittent (mild symptoms up to two days/week), mild persistent (symptoms more than twice/week, but not daily, and one or two nighttime episodes/month), moderate persistent (daily symptoms and three or four nighttime symptoms/month), and severe persistent (symptoms throughout most days and frequently at night).
  • treating refers to any reduction in the severity and/or onset of symptoms associated with inflammatory or allergic lung diseases (e.g., asthma) and, as such, includes therapeutic and prophylactic measures.
  • treatment may result in a reduced number and/or severity of asthmatic attacks in a patient prone to allergy or airway hyperresponsiveness.
  • the method in one aspect is performed as soon as possible after it has been determined that a subject is at risk for inflammatory or allergic lung diseases (e.g., allergy or airway hyperresponsiveness is diagnosed) or as soon as possible after an inflammatory or allergic lung disease (e.g., asthma) manifests in the subject.
  • the pharmaceutically acceptable salt or ester of cromolyn is optionally administered with one or more additional medicaments.
  • additional anti-amyloid agents or anti-inflammatory agents can be administered.
  • additional anti-asthma agents, or anti-inflammation agents, or other agents that are used to treat airway hyperresponsiveness can be administered.
  • Additional medicaments may be provided in any dosage form, including solid dosage forms (e.g., tablets, capsules and powders) and liquid dosage forms (e.g., solutions, suspensions, emulsions, syrups and elixirs).
  • the additional medicaments may be administered by any known route of administration, including oral (e.g., ingestion or inhalation), injection (e.g., intravenous, intraarterial, intramuscular, subcutaneous, intradermal, intraarticular, intrathecal, epidural, intracerebral, or intraperitoneal), buccal, rectal, topical, transdermal, intranasal, via the pulmonary route, via inhalation, or intraophthalmic.
  • the additional medicaments may be administered concurrently with or sequentially (i.e. before or after) with the pharmaceutically acceptable salt or ester of cromolyn.
  • Additional medicaments include, for example, Levodopa (Sinemet), anticholinergics, Eldepryl, steroids, antihistamines, long-acting or short-acting beta-agonists, immunomodulators (e.g., Omalizumab), and Theophylline.
  • the additional medicament can be a cholinesterase inhibitor (e.g., Donepezil (Aricept®), rivastigmine (Exelon®), galantamine (Razadyne®), or Tacrine (Cognex®)), a NMDA receptor antagonist (e.g., memantine (Namenda®)), a gamma secretase inhibitor (e.g., LY451039 (Semagacestat, Eli Lily)), a metal ionophore (e.g., PBT2 (Prana)), a statin, and/or an endocannabinoid (e.g., arachidonoylethanolamine, tetrahydrocannabinol, 2-arachidonoyl glycerol, 2-arachidonyl glyceryl ether, N-arachidonoyl-dopamine, or virodhamine).
  • a cholinesterase inhibitor e.g.,
  • non-steroidal anti-inflammatory drugs include, but are not limited to, ibuprofen, acetylsalicylic acid, diflunisal, salsalate, dexibuprofen, naproxen, fenoprofen, ketoprofen, dexketoprofen, flurbiprofen, oxaprozin, loxoprofen, indomethacin, tolmetin, sulindac, etodolac, ketorolac, diclofenac, nabumetone, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam, isoxicam, mefenamic acid, meclofenamic acid, flufenamic acid, tolfenamic acid, celecoxib, licofelone, hyperforin, and figwort.
  • the non-steroidal anti-inflammatory drug is administered orally (via ingestion or inhalation).
  • the non-steroidal anti-inflammatory drug e.g., ibuprofen
  • the non-steroidal anti-inflammatory drug can be administered in an amount of about 5 mg to about 80 mg per day, for example, about 5 mg to about 60 mg per day, about 5 mg to about 50 mg per day, about 5 mg to about 40 mg per day, about 5 mg to about 30 mg per day, about 5 mg to about 20 mg per day, and/or about 5 mg to about 15 mg per day.
  • the non-steroidal anti-inflammatory drug e.g., ibuprofen
  • ibuprofen may be administered in a once-daily dose of about 5 mg to 20 mg, preferably in a once-daily dose of about 10 mg.
  • the invention further provides a group of individual delivery units, for example, a blister pack comprising blisters, containing a pharmaceutically acceptable salt or ester of cromolyn.
  • Blister packs are known in the art, and generally comprise a solid support comprising a plurality of spaced bubbles or wells (collectively referred to herein as “blisters”) for carrying a predetermined amount of medicament.
  • a film or membrane seals the wells, and is susceptible to puncture or release from the solid support to make the medicament available for delivery.
  • the specific shape, proportions, and dimensions of the blister pack and the individual blisters can be adjusted for use in a particular delivery device.
  • the blister pack is optionally provided as a coil or a circular (e.g., carousel) cartridge for insertion in a dry powder inhaler, and the blisters are shaped as inverted cones or domes. Additionally, the number of blisters (corresponding to the number of doses) may be varied.
  • the blister pack is composed of a material that protects the contents of the blisters from exposure to the environment and is compatible for use with an inhalation device for delivering cromolyn to a patent.
  • Suitable materials include, but are not limited to, PVC (polyvinyl chloride), PVC/PVDC (polyvinylidene chloride) blends, PE (polyethylene), PP (polypropylene), polystyrene, cellophane, polyester (e.g. a polyester terephthalate), paper, polyamide, PET (polyethylene terephthalate), COC (cyclic olefin copolymer), metallic (e.g., aluminum) foil and any blend thereof.
  • PVC polyvinyl chloride
  • PVC/PVDC polyvinylidene chloride
  • PE polyethylene
  • PP polypropylene
  • polystyrene cellophane
  • polyester e.g. a polyester terephthalate
  • paper e.g.
  • Blister packs are further described in, for example, U.S. Pat. Nos. 5,497,763; 7,080,644; 7,828,150; 7,931,022; and 8,291,900, and International Patent Publication Nos. WO 1999/23180 and WO 1989/01348 (all of which are hereby incorporated by reference in their entirety, and particularly with respect to their respective descriptions of blister packs).
  • blisters have a maximum capacity of about 15 to 20 mg of ingredients, which includes both the active ingredient (i.e. cromolyn, or its pharmaceutically acceptable salt or ester) and pharmaceutically acceptable excipients.
  • the blisters of the blister pack contain about 3 mg to about 20 mg of a pharmaceutically acceptable salt or ester of cromolyn, for example, 3 mg to about 19 mg, 3 mg to about 18 mg, 3 mg to about 17 mg, about 3 mg to about 15 mg, about 3 mg to about 14 mg, about 3 mg to about 13 mg, about 3 mg to about 12 mg, about 3 mg to about 11 mg, about 3 mg to about 10 mg, about 3 mg to about 9 mg, about 3 mg to about 8 mg, about 3 mg to about 7 mg, about 4 mg to about 20 mg, about 6 mg to about 18 mg, about 8 mg to about 17.5 mg, or about 16.1 mg to about 19.9 mg of the pharmaceutically acceptable salt or ester of cromolyn.
  • additional amounts of pharmaceutically acceptable excipients are included in the blisters with the cromolyn.
  • a capsule is used instead of a blister. Capsules for inhaler devices are well known in the art.
  • the pharmaceutically acceptable salt or ester of cromolyn is provided in a solid dosage form, preferably a powder containing particles of the pharmaceutically acceptable salt or ester of cromolyn as described herein.
  • the powder optionally comprises one or more pharmaceutically acceptable excipients, as described above.
  • the pharmaceutically acceptable excipients when present, are included in the powder in a total amount of about 0.1% to about 80% by weight, about 1% to about 80% by weight, about 5% to about 80% by weight, about 10% to about 80% by weight, about 15% to about 80% by weight, about 20% to about 80% by weight, about 25% to about 80% by weight, about 30% to about 80% by weight, about 35% to about 80% by weight, about 40% to about 80% by weight, about 20 to about 75% by weight, about 20% to about 70/o by weight, about 20% to about 65% by weight, about 20% to about 60% by weight, about 25% to about 55% by weight, about 30% to about 50% by weight, about 35% to about 45% by weight, and/or about 40% by weight.
  • the cromolyn is provided in liquid solution form.
  • the invention further provides a kit comprising a blister pack, such as the blister pack described herein, and a dry powder inhaler (DPI) device.
  • a kit comprises a capsule pack and a dry powder inhaler (DPI) device.
  • the device is an active dry powder inhaler device, such as a dry powder inhaler device that comprises a chamber comprising a piezoelectric vibrator for deaggregating a dry powder and an air flow passageway in which the deaggregated powder is picked up and carried for inhalation by a patient.
  • the kit optionally includes one or more additional medicaments, such as a non-steroidal anti-inflammatory drug (e.g., ibuprofen, acetylsalicylic acid, diflunisal, salsalate, dexibuprofen, naproxen, fenoprofen, ketoprofen, dexketoprofen, flurbiprofen, oxaprozin, loxoprofen, indomethacin, tolmetin, sulindac, etodolac, ketorolac, diclofenac, nabumetone, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam, isoxicam, mefenamic acid, meclofenamic acid, flufenamic acid, tolfenamic acid, celecoxib, licofelone, hyperforin, and figwort).
  • a non-steroidal anti-inflammatory drug e.g.,
  • NMI Next Generation Pharmaceutical Impactor cascade impactor device
  • the passive inhaler device included an active particle dispersion mechanism (ACTIVEMESH, Aespira) involving breath-driven beating of a mesh package containing the powder to be delivered.
  • ACTIVEMESH active particle dispersion mechanism
  • a blend containing disodium cromoglycate (DSCG) (Cambrex) and Lactohale LH300 lactose (Friesland Foods Domo) was prepared by blending in an Alpine Picoline high shear mixer (module Picomix) (Hosokawa Alpine, Augsburg, Germany) at a speed of 4000 rpm for 3 minutes.
  • the blend contained 80:20 (wt./wt.) DSCG:lactose.
  • Neat DSCG also was tested.
  • the samples (neat cromolyn or cromolyn:lactose blend) were loaded into the passive dry powder inhaler device and tested using an NGI cascade impactor device at a flow rate of approximately 100 L/min for 2.4 seconds.
  • the results of testing are provided in Table 1.
  • the passive inhaler device delivered only 3% of the initial dose of neat DSCG and only 6% of the initial dose of blended cromolyn to Stage 4 and higher of the NGI cascade impactor device.
  • cromolyn Delivery of cromolyn by a PROHALER pre-metered multidose passive dry powder inhaler device (Aptar) was assessed.
  • the PROHALER inhaler was initially expected to more efficiently deliver cromolyn because the device creates rapidly fluctuating airflow/shear and turbulence to deagglomerate the particles emitted from the blister prior to oral inhalation by the patient.
  • the PROHALER inhaler also included breath-triggered opening of a dose and a blister cartridge system.
  • DSCG disodium cromoglycate
  • DMV-Fonterra Excipients lactose monohydrate
  • the blends were filled into blisterstrips, which were then assembled into a PROHALER inhaler device and tested using an NGI device at a flow rate of approximately 35 L/min.
  • the results of testing are provided in Table 2.
  • the emitted dose represents the total quantity of DSCG fired into the impactor.
  • a fine particle dose with particles less than 3 ⁇ m in size corresponds approximately to the amount of cromolyn delivered to Stage 4 and higher of the NGI device.
  • the PROHALER inhaler device delivered only about 4.4% of the initial dose of blended cromolyn (50:50 blend) and only about 9.6% of the initial dose of blended cromolyn (20:80 blend) to Stage 4 and higher of the NGI cascade impactor device.
  • TWISTER inhaler device a capsule-based dry powder inhaler. Compared to blisters, capsules typically have much higher drug loading capacities.
  • a blend containing disodium cromoglycate (DSCG) and lactose was prepared.
  • the blend was assembled into a TWISTER inhaler device and tested using an NGI cascade impactor device at a flow rate of approximately 30 L/min.
  • the results of testing are provided in Table 3.
  • the TWISTER inhaler device delivered only about 24% of the initial dose of blended cromolyn to Stage 4 and higher of the NGI cascade impactor device.
  • the inhaler device included injection molded plastic components, electronics, a battery, and drug filled blisters.
  • the inhaler included a lever arm, upon actuation of which the blister was placed in contact with a piezoelectric vibrator within the device and then pierced by needles.
  • the inhaler also included an airflow sensor which automatically turned on the piezoelectric vibrator after a minimum inspiratory flow rate was exceeded. Activation of the piezoelectric vibrator deaggregated the powder particles and aerosolized them out of the blister and into the inspiratory airstream.
  • the inhaler included visual feedback at the start of dosing and as confirmation of successful completion of dosing, thereby facilitating improved patient compliance. Similar devices have been shown to deliver a consistent amount of a drug at each of the different flow rates tested, ranging from 20 L/min to 60 L/min.
  • a blend containing disodium cromoglycate (DSCG) (Cambrex) and lactose monohydrate (DFE Pharma) (grade: lactohale LH201) was prepared by blending at a speed of 150 rpm for 15 minutes.
  • the blend contained 60:40 (wt/wt.) DSCG:lactose.
  • Neat DSCG also was tested.
  • the samples (neat cromolyn or cromolyn:lactose blend) were filled into blisters, which were then assembled into the inhaler device and tested using an NGI device at a flow rate of approximately 30 L/min for 2 seconds.
  • the results of testing are provided in Table 4.
  • the fine particle dose is the amount of cromolyn recovered below a diameter of 5 ⁇ m.
  • the fine particle fraction ⁇ 5 ⁇ m is provided as a percentage of the amount of cromolyn recovered from the NGI cascade impactor device (i.e., the delivered dose).
  • the active inhaler device delivered 42% of the initial dose of neat DSCG and 43% of the initial dose of blended cromolyn to Stage 4 and higher of the NGI cascade impactor device.
  • the results for the active dry powder inhaler device demonstrate delivery of over 1.5 mg and over 30% of the administered dose to Stages 4 and higher of the NGI cascade impactor device.
  • cromolyn An FDA-approved route of administration for cromolyn is oral inhalation using a capsule-based dry powder inhaler, with 20 mg cromolyn loaded per capsule. Studies have shown that with high inspiratory rates, the inhaled cromolyn is delivered to the human lung, with 10-15% of the inhaled drug-delivered dose absorbed into the bloodstream.
  • Cromolyn is supplied by Cambrex Proforma (Milan, Italy) and used to formulate a 17.1 mg dose of cromolyn for inhalation using inhalation grade excipients.
  • Cromolyn was micronized at Cambrex to achieve an average particle size of 5 microns or less. It is important for particles to be less than 10 microns, with the majority of particles falling between 2 and 5 microns, since this is necessary for successful deposition to the secondary bronchi of the respiratory tract following inhalation.
  • Cromolyn capsules for inhalation were manufactured by Pharmaterials, Ltd (Reading, UK). Initial characterization of the manufactured Cromolyn was performed by Pharmaterials and was found to be crystalline and hygroscopic material with particle size distribution suitable for inhalation (d50 ⁇ 5 ⁇ m and d90 ⁇ 10 ⁇ m). Therefore, there was a need to use hydrophobic excipient in the powder formulation to improve product performance and stability. Addition of hydrophobic excipient offers inherent resistance of dry powder inhalation formulations to negative effect of moisture to such formulations. Magnesium stearate was chosen as suitable hydrophobic excipient since it is commercially used in dry powder inhaler (DPI) products.
  • DPI dry powder inhaler
  • ALZT-OP1a (Cromolyn) Formulation ALZT-OP1a Composition Placebo Drug Product Quality mg/ mg/ Component Standard Function % w/w capsule % w/w capsule Cromolyn sodium USP Active — — 58.0 17.1 a (micronized) Lactose NF Diluent 98.0 44.1 40.0 12.8 monohydrate Magnesium NF Stabilizer 2.0 0.9 2.0 0.6 stearate (micronized) Hydroxypropyl In-house Encapsulation NA NA NA NA methylcellulose capsule b Total 100% 45 100% 32 USP: United States Pharmacopeia; NF: National Formulary a Weight of cromolyn sodium, USP per capsules is 17.1 mg on an anhydrous basis (18.6 mg per capsule on as-is basis). b Hydroxypropyl methylcellulose capsule functions only to meter and deliver the drug product through the dry powder inhaler and is not ingested during administration.
  • the AZHALER DPI (239700001AB-Rs01) is a monodose inhaler device manufactured by Plastiape S.p.a (Italy) for use with ALZT-OP1a (cromolyn). It is a single-dose device used to deliver the inhalation powder to the patient's lung in form of an aerosol.
  • the inhaler consists of a container, a valve, and a mouthpiece.
  • the low resistance model 80 L airflow resistance
  • These device reaches a pressure drop of 4 kPa at c.80 L/min, and is suitable for use on a wide range of patient population i.e. including elderly, children and patients with severe respiratory impairment
  • the primary objective of the Phase I open-labeled study was to determine the PK of cromolyn in plasma and CSF, following administration of a single 17.1 mg inhaled dose of cromolyn.
  • the study also evaluated the PK of cromolyn in plasma and CSF, following administration of 34.2 mg of cromolyn (administration of two consecutive inhaled doses of 17.1 mg taken not more than two minutes apart).
  • the secondary objective of the study was to evaluate the safety and tolerability of a single or double dose of cromolyn following administration.
  • the study population consisted of 24 normal, healthy volunteers between the ages of 55-75 in good general health, without respiratory disease, confirmed by pulmonary function testing.
  • PK profile of cromolyn in plasma was characterized by a rapid absorption phase, with an average C max of 46.7 ⁇ 33.0 ng/ml (range 14.0-133.0 ng/ml) at 22.8 ⁇ 16.6 min (range 6-60 min) after single dose (17.1 mg) inhalation and an average C max of 96.8 ⁇ 46.2 ng/ml (range 36.1-236.0 ng/ml) at 22.2 ⁇ 19.4 min (range 6-60 min) after double dose (34.2 mg) inhalation.
  • the average apparent t % of cromolyn in plasma was 1.756 ⁇ 0.9 h (range 0.6-3.7 h) following single dose (17.1 mg) inhalation and 1.91 ⁇ 0.7 h (range 0.7-3.8 h) following double dose (34.2 mg) inhalation, indicating moderate clearance.
  • the average AUC 0-inf of cromolyn in plasma increased with the dose increase from 195.71 ⁇ 97.33 h*ng/ml (range 93.3-287.0 h*ng/ml) to 284.55 ⁇ 91.29 h*ng/ml (range 154.8-443.3 h*ng/ml) following single (17.1 mg) and double (34.2 mg) dose inhalation, respectively.
  • Cromolyn plasma PK parameters are summarized in Table 6.
  • cromolyn in CSF were indicative of drug delivery to the brain.
  • the average C max in CSF during the observed time interval of up to 4 hours was 0.24 ⁇ 0.077 ng/ml (range 0.2-0.4 ng/ml) at 3.72 ⁇ 0.704 h after single dose (17.1 mg) inhalation and 0.34 ⁇ 0.171 ng/ml (range 0.2-0.6 ng/ml) at 3.45 ⁇ 0.952 h after double dose (34.2 mg) inhalation.
  • the observation period (0-4 hours) for the CSF samples was too short to allow for determination of t 1/2 .
  • the cromolyn AUC in plasma was 675 and 809 times higher than in CSF 2 hours after single (17.1 mg) and double (34.2 mg) dose inhalation and 232 and 299 times higher 4 hours after single and double dose inhalation, respectively.
  • This Phase I study is designed as a randomized, double-blinded, placebo-controlled study for subjects with evidence of early AD.
  • Group I arm which will consist of Cromolyn for inhalation
  • Group I placebo arm which will consist of inhaled placebo
  • a minimum of 200 evaluable subjects will be randomized to receive one of two possible treatment assignments containing active study drug or placebo.
  • Study drugs are dispensed as 12 or 24-week supplies at Day 1, Weeks 12, 24, and 48.
  • Cromolyn powder blend or placebo will be filled into capsules for use with an AZHALER dry powder inhaler device.
  • Each capsule will contain 17.1 mg of the active product ingredient (cromolyn) and inhalation grade lactose monohydrate as well as magnesium stearate as excipients, or placebo (lactose alone).
  • the once-daily cromolyn dose to be tested in this study is about 21%, the dose from the four times daily approved dose level (80 mg cromolyn total per day [17.1/80]) for the treatment of asthma.
  • the total duration of subject participation is expected to be approximately 74 weeks or 7 visits. All subjects are required to have an initial screening visit (Day ⁇ 21 to Day ⁇ 1) to determine eligibility and a baseline visit (Day 1) to confirm eligibility prior to treatment start, and then asked to complete 72 weeks of daily dosing that requires them to return to the clinic at weeks 4, 12, 24, 48 and 72.
  • the primary endpoint of this study is a significant stabilization/improvement in cognitive and functional performance, as measured by the Clinical Dementia Rating test (CDR-SB), from baseline to Week 72, for subjects in the cromolyn treatment group compared to the placebo group.
  • CDR-SB Clinical Dementia Rating test
  • the primary efficacy variable will be analyzed using a mixed-effect model for repeated measures (MMRM) approach.
  • the model will include the fixed effects treatment, visit, treatment by visit interaction, region (US, Europe, Asia/ Pacific) and the covariates APOe4 (one or more alleles, no alleles), anti-dementia drug use (absent, present), baseline CDR-SB and age assuming an unstructured correlation matrix.
  • CDR Clinical Dementia Rating
  • the CDR is a clinical staging instrument for dementia. It characterizes six domains of cognitive and functional performance: Memory, Orientation, Judgment & Problem Solving, Community Affairs, Home & Hobbies, and Personal Care. The necessary information to make each rating is obtained through a semi-structured interview of the patient and a reliable informant or collateral source (e.g., a family member).
  • the CDR questionnaire provides descriptive anchors that guide the clinician in making appropriate ratings based on interview data and clinical judgment. The CDR is conducted at Screening and at Weeks 12, 24, 48, and 72.
  • CDR Global Clinical Dementia Rating
  • the Washington University Global CDR score (5 stages: 0, 0.5, 1, 2, 3) will be used as an inclusion criterion to screen and select a subject population consistent with early AD. Subjects are required to have a Global CDR score of 0.5 (mildly impaired), with the Memory Box score being at least 0.5. Each global score will be calculated using the Washington University online algorithm.
  • CDR-SB Clinical Dementia Rating-Sum of Boxes
  • the CDR-SB score (score range 0-18) will serve as the primary endpoint for the study and will be used for study group comparisons, monitoring disease progression, and evaluating treatment effect between active and placebo groups.
  • a CDR-SB score of 0 represents normal functioning and a score of 18 represents severe impairment.
  • the CDR-SB is expected to provide a more detailed quantitative general index than the Global CDR in patients with mild dementia.
  • the CDR-SB is conducted at Screening and at Weeks 12, 24, 48, and 72.
  • MMSE Mini-Mental State Examination
  • the MMSE is a brief examination intended to evaluate an adult participant's level of cognitive functioning. The test is performed in following areas: orientation in time and place, learning and immediate recall, mental control and concentration, short-term recall, naming ability, language expression, verbal comprehension, writing comprehension, writing ability, and visual-spatial coordination. Scores range from 0 (maximum cognitive deficit) to 30 (no cognitive deficit). The MMSE is conducted at Screening and at Weeks 12, 24, 48, and 72.
  • WMS-III The Wechsler Memory Scale-Third Edition
  • the Logical Memory II subtest will be used to assess delayed memory function.
  • the WMS-III Logical Memory I (Immediate Recall) and Logical Memory II (Delayed Recall) will both be administered at Screening (only), but only the Logical Memory II score will be used as an inclusion criterion.
  • LM H scores acceptable for inclusion will be categorized by subjects' years of education.
  • Logical Memory II will be administered 30 minutes after the administration of Logical Memory I.
  • the cromolyn monotherapy administered daily at the dose of 17.1 mg to patients having early stage AD can be as effective in stabilization/improvement of cognitive and functional performance as the combination therapy of cromolyn and ibuprofen.

Abstract

Methods of delivering cromolyn to a patient in need thereof, methods of treating amyloid-associated conditions and inflammatory or allergic lung diseases, and packs and kits comprising cromolyn are described.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part application of the U.S. patent application Ser. No. 14/893,487, filed Nov. 23, 2015, which is a national phase application of PCT international application PCT/US2014/039118, filed May 22, 2014, which claims priority to and the benefit of U.S. Provisional Application 61/826,798, filed May 23, 2013, both of which are incorporated by reference herein in their entirety.
  • FIELD OF THE INVENTION
  • The invention relates generally to cromolyn compositions and methods thereof. Specifically, the invention relates to effectively delivering cromolyn particles to a patient in need thereof for treating various diseases.
  • BACKGROUND OF THE INVENTION
  • Cromolyn (also known as cromoglicic acid, cromoglycate, or cromoglicate) has been approved previously for use in asthma. Its approved form is available as a disodium salt form, cromolyn sodium (also known as disodium cromoglycate or DSCG). Cromolyn demonstrates poor oral absorption. Delivery of cromolyn via inhalation has proven inefficient and difficult due, at least in part, to the hygroscopic nature of cromolyn sodium. For example, micronized powders containing cromolyn sodium particles spontaneously absorb water, forming clumps that impair efficient delivery of the cromolyn powder. See Keller et al. Expert Opin. Drug Deliv. 8, 1-17 (2011). Additionally, the performance and the efficiency of previously used inhalers are highly dependent upon a patient's inspiratory flow rate, leading to a wide variability in the amount of cromolyn sodium that is delivered to a patient. See Richards et al., Journal of Pharmacology and Experimental Therapeutics, 241, 1028-1032 (1987).
  • The present invention provides improved compositions and methods for delivering cromolyn via inhalation, efficiently and consistently over a range of inspiratory flow rates.
  • SUMMARY OF THE INVENTION
  • The invention is directed to a method of delivering cromolyn to a patient in need thereof. Such patients include patients in need of systemic delivery of cromolyn, e.g., to the brain or other non-lung tissues. In exemplary embodiments, the patient has an amyloid-associated condition. Patients in need of cromolyn also include patients in need of pulmonary delivery of cromolyn, for lung or airway related conditions. In exemplary embodiments, the patient has an inflammatory or allergic lung disease, such as asthma.
  • The method comprises administering to the patient via oral inhalation a pharmaceutically acceptable salt or ester of cromolyn in the form of a powder comprising particles of the pharmaceutically acceptable salt or ester of cromolyn. The majority of particles may have a diameter of about 2 to about 5 microns. The powder is administered using a device that deposits (a) at least 1.5 mg and (b) at least 20% by weight of the administered amount of pharmaceutically acceptable salt or ester of cromolyn to Stage 4 and higher of a Next Generation Pharmaceutical Impactor (NGf) cascade impactor device at a flow rate of, for example, 20-90 L/min.
  • In related methods, the invention also provides a method of delivering cromolyn to a patient in need thereof, comprising administering to the patient via oral inhalation a pharmaceutically acceptable salt or ester of cromolyn in the form of a powder, under conditions such that (a) at least 1.5 mg and (b) at least 20% by weight of the administered amount of the pharmaceutically acceptable salt or ester of cromolyn is delivered to the lower airways of the patient. As used herein, the term “lower airways” refers to the region of the airways/lung that corresponds to Stage 4 and higher of a Next Generation Pharmaceutical Impactor (NGI) cascade impactor device.
  • The invention also provides a method of delivering cromolyn to a patient in need thereof, comprising administering to the patient via oral inhalation a pharmaceutically acceptable salt or ester of cromolyn in the form of a powder comprising particles of the pharmaceutically acceptable salt or ester of cromolyn using a dry powder inhaler (DPI) device comprising a chamber comprising a piezoelectric vibrator or an ultrasonic vibrator for deaggregating a powder and an air flow passageway in which the deaggregated powder is picked up and carried for oral inhalation by a patient. The majority of the particles of the pharmaceutically acceptable salt or ester of cromolyn may have a diameter of about 2 to about 5 microns.
  • In various embodiments, about 3 mg to about 20 mg (e.g., about 16.1 mg to about 19.9 mg) of the pharmaceutically acceptable salt or ester of cromolyn is administered to the patient. Optionally the pharmaceutically acceptable salt or ester of cromolyn is administered with one or more pharmaceutically acceptable excipients. In various embodiments, the powder comprises about 0.1% to about 80%, for example, about 40% to about 80% by weight of an excipient. In various embodiments, the excipient is a monosaccharide, a disaccharide, an oligosaccharide, a polysaccharide, or a polyalcohol, such as lactose, mannitol, or sorbitol.
  • Additionally, the invention provides a method of treating an amyloid-associated condition in a patient in need thereof. The method comprises administering to the patient via oral inhalation an amount of powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn via pulmonary delivery, the majority of particles having a diameter of about 2 to about 5 microns, using a device that deposits (a) at least 1.5 mg and (b) at least 20% by weight of the administered amount of pharmaceutically acceptable salt or ester of cromolyn to Stage 4 and higher of a Next Generation Pharmaceutical Impactor (NGI) cascade impactor device at a flow rate of, for example, 20-90 L/min (e.g., for about 4 seconds).
  • The invention further provides a method of treating inflammatory or allergic lung diseases in a patient in need thereof. The method comprises administering to the patient via oral inhalation an amount of powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn, the majority of particles having a diameter of about 2 to about 5 microns, at a frequency of 1 or 2 times daily, each dose comprising about 3 mg to about 20 mg of a pharmaceutically acceptable salt or ester of cromolyn. The dose and/or frequency of delivery according to such methods are reduced compared to the conventional dosages and frequencies.
  • In various aspects of the invention, the powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn is administered to a patient using an active dry powder inhaler, such as a dry powder inhaler comprising a chamber comprising a piezoelectric vibrator or an ultrasonic vibrator for deaggregating a dry powder and an air flow passageway in which the deaggregated powder is picked up and carried for oral inhalation by a patient.
  • In various aspects, the invention is directed to a method for treating an amyloid-associated condition in a patient in need thereof, comprising administering to the patient via oral inhalation an amount of liquid particles of a solution comprising a pharmaceutically acceptable salt or ester of cromolyn using a nebulizer. The particles have a mass median aerodynamic diameter (MMAD) of about 0.5 to about 15 microns. In some embodiments, the majority of particles have a diameter of about 2 microns to about 5 microns.
  • The invention further includes use of a pharmaceutically acceptable salt or ester of cromolyn in the preparation of a medicament for treating inflammatory or allergic lung diseases in a patient in need thereof in an amount from about 3 mg to about 20 mg (e.g., from about 16.1 mg to about 19.9 mg). The medicament is administered via oral inhalation at a frequency of 1 or 2 times daily, and the pharmaceutically acceptable salt or ester of cromolyn is in the form of a powder comprising particles of pharmaceutically acceptable salt or ester of cromolyn optionally having a diameter of about 0.5 to about 15 microns, preferably an average particle size of about 5 microns or less. The invention further contemplates use of a pharmaceutically acceptable salt or ester of cromolyn in the preparation of a medicament for treating an amyloid-associated condition in a patient in need thereof. The medicament is administered to the patient via pulmonary delivery or oral inhalation. In some embodiments, the pharmaceutically acceptable salt or ester of cromolyn is in the form of a powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn, wherein the majority of particles having a diameter of about 2 to about 5 microns. The medicament is optionally delivered using a dry powder inhaler device as described herein. Alternatively, the pharmaceutically acceptable salt or ester of cromolyn is in solution and administered using a nebulizer.
  • Additionally, the invention provides a blister pack for delivering cromolyn to a patient in need thereof. The blister pack comprises blisters containing about 3 mg to about 20 mg (e.g., about 16.1 mg to about 19.9 mg) of a pharmaceutically acceptable salt or ester of cromolyn. The invention also is directed to a kit comprising a blister pack as described herein and a dry powder inhaler (DPI) device. In some embodiments, the device is an active dry powder inhaler, such as a dry powder inhaler device comprising a chamber comprising a piezoelectric vibrator for deaggregating a dry powder and an air flow passageway in which the deaggregated powder is picked up and carried for oral inhalation by a patient. In some embodiments, the kit further comprises ibuprofen tablets.
  • Further aspects of the invention may become apparent to those skilled in the art from a review of the following detailed description, taken in conjunction with the appended claims. While the invention is susceptible of embodiments in various forms, described hereinafter are specific embodiments of the invention with the understanding that the disclosure is illustrative, and is not intended to limit the invention to specific embodiments described herein. The entire document is intended to be related as a unified disclosure, and it should be understood that all combinations of features described herein are contemplated, even if the combination of features are not found together in the same sentence, or paragraph, or section of this document. For example, where embodiments concerning a method of delivering cromolyn are described, embodiments involving methods of therapy, kits, and the like that have the same properties and features are specifically contemplated, and the reverse also is true.
  • In addition to the foregoing, the invention includes, as an additional aspect, all embodiments of the invention narrower in scope in any way than the variations specifically mentioned above. With respect to aspects of the invention described as a genus, all individual species are individually considered separate aspects of the invention. With respect to elements described as a selection within a range, it should be understood that all discrete subunits within the range are contemplated as an embodiment of the invention. Ranges may be expressed herein as from “about” or “approximately” one particular value and/or to “about” or “approximately” another particular value. When such a range is expressed, another embodiment according to the invention includes from the one particular value and/or to the other particular value. Similarly, when particular values are expressed as approximations, but use of antecedents such as “about,” “at least about,” or “less than about,” it will be understood that the particular value forms another embodiment.
  • With respect to aspects of the invention described or claimed with “a” or “an,” it should be understood that these terms mean “one or more” unless context unambiguously requires a more restricted meaning. The term “or” should be understood to encompass items in the alternative or together, unless context unambiguously requires otherwise. If aspects of the invention are described as “comprising” a feature, embodiments also are contemplated “consisting of” or “consisting essentially of” the feature.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention provides improved methods and compositions for the efficient and consistent delivery of cromolyn via inhalation. Such methods preferably deliver a consistent amount of drug over a wide range of patient inspiratory flow rates. According to some aspects of the inventive method, a large portion of an administered amount of cromolyn is delivered to regions of the lungs that mediate transport into systemic circulation (the bronchi, bronchioles, and alveoli), e.g. the lower airways. Thus, the inventive method can provide an effective means for delivering cromolyn systemically, i.e., into the blood stream (and, by extension, to other non-lung regions of the body, such as the brain). The enhanced delivery efficiency associated with the inventive method allows administration of lower doses of cromolyn and/or less frequent administration of cromolyn, to achieve a desired biological response in any condition requiring lung delivery or systemic delivery. Advantages include improved therapeutic efficacy at conventional doses, or maintained/improved therapeutic efficacy at lower doses and/or lower frequencies of administration, leading to improved ease of use, higher patient compliance, and improved therapeutic benefit, as well as cost savings associated with using reduced amounts of drug. Additionally, many drug packaging systems containing groups of individual delivery units, such as blister packs or capsules, have maximum capacities for a single dose of drug contained within individual delivery units. More efficient delivery of cromolyn also advantageously facilitates the packaging of a therapeutically effective dose of cromolyn in each individual delivery unit. Advantages include delivery of a higher dose per single administration, delivery of uniform doses in a multiple administration regimen (i.e., little variability between doses), and/or use of a single individual delivery unit per administration rather than multiple individual delivery units per administration.
  • The invention is described in further detail below. Section headings are for convenience of reading and not intended to be limiting per se.
  • Powder Administration
  • In one aspect, the invention provides a method of delivering cromolyn to a patient in need thereof. Such patients include patients in need of lung or systemic delivery of cromolyn, and include patients with amyloid-associated conditions as well as patients with an inflammatory or allergic lung disease such as asthma. In any of the embodiments described herein, cromolyn (also known as cromoglicic acid, cromoglycate, cromoglicate, or 5,5′-(2-hydroxypropane-1,3-diyl)bis(oxy)bis(4-oxo-4H-chromene-2-carboxylic acid)) can be administered as a powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn.
  • Pharmaceutically acceptable salts are well known to those skilled in the art and include pharmaceutically acceptable inorganic and organic base addition salts, which may be formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Pharmaceutically acceptable salts of compounds may also be prepared with a pharmaceutically acceptable cation. These salts can be prepared in situ in the administration vehicle or the dosage form manufacturing process, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically-acceptable metal cation, with ammonia, or with a pharmaceutically-acceptable organic primary, secondary or tertiary amine. Suitable pharmaceutically acceptable cations are well known to those skilled in the art and include alkaline, alkaline earth, ammonium and quaternary ammonium cations. Examples of metals used as cations are lithium, sodium, potassium, magnesium, ammonium, calcium, aluminum, or ferric, and the like. Examples of suitable amines include ethylamine, diethylamine, piperazine, isopropylamine, trimethylamine, histidine, N,N′-dibenzylethylenediamine, chloroprocaine, choline, ethanolamine, diethanolamine, dicyclohexylamine, ethylenediamine, N-methylglucamine, and procaine. Suitable esters of cromolyn include, but are not limited to, carboxylate esters of one or both of the carboxylic acids of cromolyn, such as aliphatic esters (e.g., methyl esters, ethyl esters, propyl esters, butyl esters such as t-butyl esters, and pentyl esters), aryl esters (e.g., phenyl esters and benzyl esters), and combinations thereof. In one example, the pharmaceutically acceptable salt of cromolyn is disodium cromoglycate.
  • The pharmaceutically acceptable salt or ester of cromolyn is administered via inhalation, generally via oral inhalation, however, nasal inhalation or a combination of oral and nasal inhalation can also be used. When systemic delivery is desired, administration via inhalation as described herein delivers cromolyn to the lungs of the patient, depositing the pharmaceutically acceptable salt or ester of cromolyn onto surfaces of the lung that allow absorption into the blood stream (e.g., the bronchi, bronchioles, and alveoli), e.g. the lower airways.
  • In one embodiment, the particles of the cromolyn salt or ester have a median particle diameter (D50) of less than about 5 microns, less than about 4.8 microns, less than about 4.5 microns, less than about 4.2 microns, less than about 4 microns, less than about 3.8 microns, less than about 3.5 microns, less than about 3.2 microns, less than about 3 microns, less than about 2.8 microns, less than about 2.5 microns, less than about 2 microns, less than about 1.8 microns, less than about 1.5 microns, less than about 1.2 microns, less than about 1 micron, about 0.1 microns to about 5 microns, about 0.5 microns to about 5 microns, about 0.8 microns to about 5 microns, about 1 micron to about 5 microns, about 1.2 microns to about 5 microns, about 1.5 microns to about 5 microns, about 1.8 microns to about 5 microns, about 2 microns to about 5 microns, about 1.2 microns to about 5 microns, about 2.5 micron to about 5 microns, about 2.8 microns to about 5 microns, about 3 microns to about 5 microns, about 3.2 microns to about 5 microns, 3.5 micron to about 5 microns, about 3.8 microns to about 5 microns, about 4 microns to about 5 microns, about 4.2 microns to about 5 microns, 4.5 micron to about 5 microns, and/or about 4.8 to about 5 microns. The particles of the cromolyn salt or ester optionally have a D90 of less than 15 microns, less than 14 microns, less than 13 microns, less than 12 microns, less than 11 microns, less than 10 microns, less than 9.5 microns, less than 9 microns, less than 8.5 microns, less than 8 microns, less than 7.5 microns, less than 7 microns, less than 6.5 microns, less than 6 microns, less than 5.5 microns, less than 5 microns, less than 4.5 microns, less than 4 microns, less than 3.9 microns, less than 3.8 microns, less than 3.7 microns, less than 3.6 microns, about 3 microns to about 15 microns, about 5 microns to about 13 microns, about 7 microns to about 11 microns, and/or about 9 microns to about 10 microns.
  • As used herein, the term D50 refers to a diameter at which 50% of a sample's mass is comprised of smaller particles. As used herein, the term D90 refers to a diameter at which 90% of a sample's mass is comprised of smaller particles. The D50 is also known as the “mass median diameter” as it divides the sample equally by mass.
  • In another embodiment, the particles of the cromolyn salt or ester have a mass median aerodynamic diameter (MMAD) of about 0.5 to about 15 microns, about 0.5 to about 10 microns, about 1 to about 5 microns, about 1 to about 4 microns, about 1 to about 3.5 microns, about 1 to about 3 microns, about 1 to about 2.5 microns, and/or about 1 to about 2 microns. Particles of a desired size are obtained by any method, such as a method known to one of skill in the art, including micronization or milling. As used herein, the term “aerodynamic diameter” refers to the diameter of a sphere of unit density that reaches the same velocity in an air stream as a non-spherical particle of arbitrary density. As used herein, the term “mass median aerodynamic diameter” refers to the aerodynamic diameter at which 50% of the particles by mass are larger and 50% are smaller. Methods of determining aerodynamic diameter are known in the art and described in, e.g., Chow et al., “Particle Engineering for Pulmonary Drug Delivery,” Pharm. Res., 24 (3), 411-437 (2007).
  • The particles of the cromolyn salt or ester may have a geometric standard deviation (GSD) of about 1.3 to about 2.5, about 1.4 to about 2.4, about 1.5 to about 2.3, about 1.6 to about 2.2, about 1.7 to about 2.1, and/or about 1.8 to about 2. As used herein, the term “geometric standard deviation” refers to the spread of an aerodynamic particle size distribution. GSD can be calculated as follows: GSD=(d84/d16)1/2, where d84 and d16 represent the diameters at which 84% and 16% of the aerosol mass are contained, respectively, in diameters less than these diameters.
  • The powder optionally comprises one or more pharmaceutically acceptable excipients. Suitable excipients are well tolerated by pulmonary tissue, and include, but are not limited to, monosaccharides, disaccharides, oligosaccharides, polysaccharides, polyalcohols, and combinations thereof. Exemplary excipients include, but are not limited to, lactose, mannitol, sorbitol, and combinations thereof. The pharmaceutically acceptable excipient(s), when present, are included in the powder in a total amount of about 0.1% to about 80% by weight, about 1% to about 80% by weight, about 5% to about 80% by weight, about 10% to about 80% by weight, about 15% to about 80% by weight, about 20% to about 80% by weight, about 25% to about 80% by weight, about 30% to about 80% by weight, about 35% to about 80% by weight, about 40% to about 80% by weight, about 20 to about 75% by weight, about 20% to about 70% by weight, about 20% to about 65% by weight, about 20% to about 60% by weight, about 25% to about 55% by weight, about 30% to about 50% by weight, about 35% to about 45% by weight, and/or about 40% by weight. The pharmaceutically acceptable excipients can be included in an anhydrous form or as a hydrate, such as a monohydrate or higher-order hydrate. The excipient(s) optionally have a particle size of about 250 microns or less, such as about 10 microns to about 150 microns, although other excipient particle sizes may be used in the context of the invention.
  • A particular administration regimen for a particular patient will depend on several factors, such as the condition being treated, co-therapies administered, the size of the patient, frequency of administration, etc. In various embodiments, the amount of powder that is administered to a patient comprises about 3 mg to about 20 mg of the pharmaceutically acceptable salt or ester of cromolyn, for example, about 3 mg to about 19 mg, about 3 mg to about 18 mg, about 3 mg to about 17 mg, about 3 mg to about 16 mg, about 3 mg to about 15 mg, about 3 mg to about 14 mg, about 3 mg to about 13 mg, about 3 mg to about 12 mg, about 3 mg to about 11 mg, about 3 mg to about 10 mg, about 3 mg to about 9 mg, about 3 mg to about 8 mg, about 3 mg to about 7 mg, about 4 mg to about 20 mg, about 6 mg to about 19 mg, about 8 mg to about 18 mg, about 10 mg to about 18 mg, about 12 mg to about 18 mg, about 14 mg to about 18 mg, about 16 mg to about 18 mg, about 17 mg to about 18 mg, about 16.1 mg to about 19.9 mg, or 16.1 mg to 19.9 mg of the pharmaceutically acceptable salt or ester of cromolyn. The amount of powder is optionally administered as a single dose or administration, which may be inhaled in a single breath or span multiple breaths in the course of the single administration. Optionally, the single dose or administration is administered from a single individual delivery unit, such as a single blister or single capsule. This single dose (single administration) may be administered repeatedly to the patient at any interval over the course of a treatment period. For example, a single dose of pharmaceutically acceptable salt or ester of cromolyn (e.g., about 16.1 mg to about 19.9 mg) is administered to a patient once a day, twice a day, or three times a day for a treatment period. Examples of treatment periods include at least 1, 2, 3, 4, 5, 6, or 7 days, or at least 1, 2, 3, or 4 weeks, or at least 1, 2, 3, 4, 5 or 6 months, or a year or more.
  • Generally, administering the powder involves suspending the powder into a gas (such as air or oxygen), thereby forming an aerosol containing the powder and the gas. Concurrently with, or subsequent to, suspension formation, the powder suspension is inhaled by a patient. In some embodiments, the powder is suspended in a gas stream being inhaled by a patient such that administering comprises suspending the powder into an inhaled gas stream.
  • Suspending the powder into a gas (such as air or oxygen) may be carried out by any means, including vibration. Vibrating the powder generally aerosolizes at least a portion (i.e., at least about 10%, at least about 30%, at least about 50%, at least about 75%, and/or at least about 90%) of the powder. For example, vibrating the powder generally involves suspending at least a portion of the powder in the gas that is in the immediate vicinity of the powder. Additionally, the powder comprising particles of cromolyn and optional pharmaceutically acceptable excipient(s) can include aggregates, for example, aggregates between two or more cromolyn particles and/or between cromolyn particles and excipient particles. Without intending to be bound by any particular theory, vibrating the powder deagglomerates particle aggregates, advantageously providing smaller particles that are more readily delivered regions of the lungs allowing transport into the blood stream (e.g., the bronchi, bronchioles, and alveoli), e.g. the lower airways. Optionally, the vibrator is vibrated such that the resulting vibrations generate synthetic jets that aerosolize and/or deaggregate the powder. Methods for forming synthetic jets are described, for example, in U.S. Pat. No. 7,318,434, which is incorporated herein by reference in its entirety. Preferably, administering the powder involves vibrating the powder at high frequency, for example, a frequency of about 10 kHz to about 50 kHz, about 15 kHz to about 40 kHz, and/or about 20 kHz to about 30 kHz.
  • In some embodiments of the invention, the amount of powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn is administered using a device that deposits (a) at least 1.5 mg and (b) at least 20% by weight of the administered amount of pharmaceutically acceptable salt or ester of cromolyn to Stage 4 and higher of a Next Generation Pharmaceutical Impactor (NGI) cascade impactor device at a flow rate of, for example, 20-90 L/min for about 1 to about 10 seconds (e.g., for about 4 seconds). In exemplary embodiments, the device deposits (a) at least 1 mg, at least 1.5 mg, at least 1.8 mg, at least 2 mg, at least 2.2 mg, at least 2.5 mg, at least 2.8 mg, at least 3 mg, at least 3.2 mg, at least 3.5 mg, at least 3.8 mg, at least 4 mg, at least 4.2 mg, at least 4.5 mg, at least 4.8 mg, at least 5 mg, at least 5.2 mg, at least 5.5 mg, at least 5.8 mg, at least 6 mg, at least 6.2 mg, at least 6.5 mg, at least 6.8 mg, at least 7 mg, at least 7.2 mg, at least 7.5 mg, at least 7.8 mg, at least 8 mg, at least 8.2 mg, at least 8.5 mg, at least 8.8 mg, at least 9 mg, at least 9.2 mg, at least 9.5 mg, at least 9.8 mg, at least 10 mg, about 1.3 mg to about 7 mg, about 1.5 mg to about 3 mg, about 1.5 mg to about 5 mg, about 1.5 mg to about 7 mg, about 1 mg to about 4 mg, about 1 mg to about 5 mg, about 1 mg to about 7 mg, about 1.8 mg to about 5.8 mg, about 1.5 mg to about 5.5 mg, and/or about 2 mg to about 5 mg and (b) at least 10%, at least 20%, at least 30%, at least 35%, at least 40%, at least 45%, about 30% to about 75%, about 30% to about 65%, about 30% to about 60%, about 30% to about 55%, and/or about 30% to about 50% by weight of the administered amount of pharmaceutically acceptable salt or ester of cromolyn to Stage 4 and higher of an NGI cascade impactor device at a flow rate of, for example, 20 L to 90 L/min for about 10 seconds or less. As used herein, the term “administered amount” in the phrase “% by weight of the administered amount” refers to the amount of pharmaceutically acceptable salt or ester of cromolyn present in the individual delivery unit (e.g., blister or capsule or other unit dose container) connected to the device. Thus, in one illustrative embodiment, the device delivers at least about 4 to about 5 mg of cromolyn salt or ester to Stage 4 and higher of an NGI cascade impactor as described herein from a capsule or blister comprising, for example about 17.1 mg, of cromolyn salt or ester with or without added excipients. NGI cascade impactors are useful for studying aerodynamic size distribution of aerosols and simulating delivery to different regions of the lung. The respiratory tract forms a particle size-selective system wherein progressively finer particles pass through mouth, larynx and larger airways to be deposited in the peripheral lung (e.g., the alveolar spaces). Similarly, cascade impactors include several “stages” allowing passage of progressively finer particles through the system. An aerosol stream carrying particles passes through each stage at a constant air flow (e.g., 20-90 L/min). Particles of similar aerodynamic size deposit at a particular stage; smaller particles are deposited at further stages of the NGI cascade impactor than larger particles. In one example, at a flow rate of 70-90 L/min (e.g., for about 4 seconds of total actuation), Stages 4 and higher of the NGI cascade impactor trap particles having a mass median aerodynamic diameter (MMAD) of about 5 microns or less. The NGI cascade impactor is generally operated under conditions of low relative humidity, such as 20% relative humidity or less. Deposition in the central (bronchial) region of the lung generally peaks with particles having an MMAD of about 4 to about 6 microns, and deposition in the peripheral (alveolar) region of the lung generally peaks with particles having an MMAD of about 2 to about 4 microns. See Mitchell and Nagel, “Particle Size Analysis of Aerosols from Medicinal Inhalers,” KONA, 22, 32-65 (2004). Thus, Stages 4 and higher of the NGI cascade impactor at a flow rate of 70-90 L/min (e.g., for about 4 seconds of total actuation) simulate the regions of the lung providing significant systemic absorption of a medicament into the bloodstream. NGI cascade impactor devices are available from MSP Corporation, Shoreview, Minn.
  • Advantageously, the inventive methods for delivering cromolyn allow consistent delivery of the active agent over a wide range of flow rates. For example, a consistent amount of powder is delivered to a patient over an inhalation flow rate of 20 L/min to 100 L/min, or 20 to 80 L/min, 20 to 60 L/min, 40-80 L/min, 60-80 L/min, 60-90 L/min, 70-90 L/min, or 75-85 L/min. By “consistent amount” is meant a relative standard deviation (RSD) of 10% or less (e.g., 5% or less, 3% or less and/or 2.5% or less) in the amount powder that is received by the patient.
  • Devices for Powder Administration
  • The devices for administering the powder described herein include, but are not limited to, a dry powder inhaler (DPI) device, a metered dose inhaler (MDI) device, and a dry powder nebulizer (DPN) device. Suitable devices typically administer a metered dose or a predetermined dose. Such doses generally are administered over a set period of time or a set volume of air. In various embodiments, the device functions independent of the patient's inspiratory flow rate. Thus, for example, the device delivers a consistent amount of cromolyn over a wide range of flow rates, such as an inhalation flow rate of 20 L/min to 100 L/min, or 20 to 80 L/min, 20 to 60 L/min, 40-80 L/min, 60-80 L/min, 60-90 L/min, 70-90 L/min, or 75-85 L/min. In a particular embodiment, the device delivers a consistent amount of cromolyn at an inhalation flow rate of 80 L/min.
  • Optionally, the device is a dry powder inhaler comprising a chamber in fluid communication with an air flow passageway. Included in the chamber is a vibrator, e.g., a piezoelectric vibrator or an ultrasonic vibrator, suitable for aerosolizing and/or deaggregating a dry powder. The inhaler optionally includes a lever for activating the piezoelectric or ultrasonic vibrator and for cooperating with a means for puncturing a blister or opening means for opening a blister.
  • In one embodiment, powder is delivered from the inhaler by receiving a pack (e.g., capsule or blister) containing the powder in an operating position proximal to the piezoelectric or ultrasonic vibrator. The patient places the mouthpiece of the inhaler into the mouth, forms a seal with his/her lips around the mouthpiece, and withdraws air through the air flow passageway while pressing the lever. Upon activation by the lever or other triggering event, the pack is broken, the piezoelectric or ultrasonic vibrator is activated, and the deaggregated powder is picked up into the inspiratory air flow of the patient and carried through the air flow passageway for inhalation by the patient.
  • The inhaler optionally includes an inhalation flow rate detector which detects the movement of air through the air flow passageway. In some embodiments, when the inhaler includes an inhalation flow rate detector, the event that triggers release of the powder includes movement of air through the air flow passageway above a threshold rate.
  • In some embodiments, the powder is delivered in a period of time of, for example, about one second to about five seconds of total inhalation time, for example, about one second to about two seconds. The inhalation period can be monitored using a visual indicator, e.g., flashing or changing the color of a light emitting diode, or an audible indicator.
  • Optionally, the lever, inhalation flow rate, and piezoelectric or ultrasonic vibrator functions are monitored and recorded in memory, such as a flash drive or other computer memory in direct contact with the inhaler or removed from the inhaler but in signal communication with the inhaler. For example, the functional parameters of the device may be transmitted from the device to a remote location, recorded in memory, and made available to healthcare providers via a wired or wireless communication network. The information also optionally may be delivered to point of care devices for monitoring treatment. The recorded performance information allows health care practitioners to monitor the use of the device, which can be particularly beneficial when a patient's ability to operate the device diminishes due to declining cognitive and/or physical health.
  • Devices also are described in U.S. Pat. Nos. 6,026,809, 7,318,434, 7,334,577, 7,779,837, 8,322,338, and 8,371,294, U.S. Patent Application Publication Nos. 2009/0090361, 2010/0294278, 2012/0077786, International Application Publication No. WO 2005/076872, and European Patent No. 0591136 BI, which are incorporated by reference in their entireties.
  • Solution Administration Using a Nebulizer
  • In various embodiments, the invention also includes a method comprising administering cromolyn to a patient using a nebulizer, preferably in a method for treating an amyloid-associated condition in a patient in need thereof. The method comprises administering liquid particles of a solution comprising a pharmaceutically acceptable salt or ester of cromolyn using a nebulizer via inhalation (e.g., oral inhalation) by the patient. These solution particles are droplets of solution in which the pharmaceutically acceptable salt or ester of cromolyn and any optional excipients are dissolved. The pharmaceutically acceptable salt or ester of cromolyn can be present in the solution at a concentration of about 1 mg/mL to about 100 mg/mL, for example, about 5 mg/mL to about 50 mg/mL, and/or about 10 mg/mL to 20 mg/mL. When included, excipient(s) can be present in the solution at a concentration of about 1 mg/mL to about 100 mg/mL, for example, about 5 mg/mL to about 50 mg/mL, and/or about 10 mg/mL to 20 mg/mL. The solution particles (i.e., droplets) have a mass median aerodynamic diameter (MMAD) of about 1 micron to about 10 microns, for example, about 1 micron to 8 micron, about 1 micron to 4 micron, about 1 micron to about 3.5 microns, about 1 micron to about 3 microns, about 1 micron to about 2.5 microns, about 1 micron to about 2 microns, about 2 microns to about 4 microns, and/or about 2.5 microns to about 3.5 microns. The solution particles (i.e., droplets) also optionally comprise a median diameter of less than about 2 microns, less than about 1.8 microns, less than about 1.5 microns, less than about 1.2 microns, less than about 1 micron, about 0.5 microns to about 2 microns, about 0.8 microns to about 2 microns, about 1 micron to about 2 microns, about 1.2 microns to about 2 microns, about 1.5 microns to about 2 microns, and/or about 1.8 microns to about 2 microns. Excipients, dosage, target regions of the lung, delivery amounts and efficiencies, and methods of estimating delivery to target regions of the lung are described above.
  • Suitable devices for administering cromolyn as a solution include, but are not limited to, a jet, ultrasonic or electronic nebulizers. Such nebulizer devices form aerosolized particles of cromolyn (e.g., a pharmaceutically acceptable salt or ester of cromolyn, such as cromolyn sodium) from a pharmaceutically acceptable solution (e.g., a hypotonic or isotonic solution). The nebulizer device optionally includes a liquid reservoir separated from a mouthpiece or face mask by a vibrating mesh, a piezoelectric element, or a compressed gas atomizer element. In such arrangements, a solution comprising a pharmaceutically acceptable salt or ester of cromolyn is present in the liquid reservoir, and the device produces a mist of particles for oral inhalation. When using a nebulizer, the cromolyn solution is optionally delivered to the patient via inhalation (oral, nasal, or a combination thereof) over the course of about ten minutes or less (e.g., two minutes).
  • The nebulizer may have any of the features of dry powder administration devices described herein. In exemplary embodiments, the device deposits/delivers at least 1 mg, at least 1.5 mg, at least 1.8 mg, at least 2 mg, at least 2.2 mg, at least 2.5 mg, at least 2.8 mg, at least 3 mg, at least 3.2 mg, at least 3.5 mg, at least 3.8 mg, at least 4 mg, at least 4.2 mg, at least 4.5 mg, at least 4.8 mg, at least 5 mg, at least 5.2 mg, at least 5.5 mg, 5.8 mg, at least 6 mg, at least 6.2 mg, at least 6.5 mg, at least 7.8 mg, at least 7 mg, at least 7.2 mg, at least 7.5 mg, 7.8 mg, at least 8 mg, at least 8.2 mg, at least 8.5 mg, at least 8.8 mg, at least 9 mg, at least 9.2 mg, at least 9.5 mg, at least 9.8 mg, at least 10 mg, about 1.3 mg to about 7 mg, about 1.5 mg to about 3 mg, about 1.5 mg to about 5 mg, about 1.5 mg to about 7 mg, about 1 mg to about 4 mg, about 1 mg to about 5 mg, about 1 mg to about 7 mg, about 1.8 mg to about 5.8 mg, about 1.5 mg to about 5.5 mg, and/or about 2 mg to about 5 mg of pharmaceutically acceptable salt or ester of cromolyn to Stage 4 and higher of an NGI cascade impactor device at a flow rate of, for example, 20 L to 90 L/min for about 10 seconds or less. In exemplary embodiments, the device deposits/delivers at least 20%, at least 30%, at least 35%, at least 40%, at least 45%, about 30% to about 75%, about 30% to about 65%, about 30% to about 60%, about 30% to about 55%, and/or about 30% to about 50% by weight of the administered amount of pharmaceutically acceptable salt or ester of cromolyn (as defined above) to Stage 4 and higher of an NGI cascade impactor device at a flow rate of, for example, about 20 L to about 90 L/min (e.g., for about 10 seconds or less).
  • Nebulizers also are described in U.S. Pat. No. 8,263,645 and U.S. Patent Application Publication Nos. 2007/0193577, 2009/0118249, and 2012/0118991, which are incorporated by reference in their entireties.
  • Amyloid-Associated Conditions
  • Any of the methods described herein include a method of treating an amyloid-associated condition in a patient in need thereof. In various example embodiments, the method comprises administering to the patient via inhalation an amount of powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn as described herein. As described above, in alternative embodiments, the method comprises administering to the patient via inhalation an amount of particles of a solution comprising a pharmaceutically acceptable salt or ester of cromolyn using a nebulizer.
  • Without wishing to be bound by a particular theory, disodium cromolyn penetrates the blood-brain barrier to bind beta-amyloid peptides and inhibit association of amyloid peptides and/or polymerization into oligomers and higher order aggregates. The inhibition of beta-amyloid association and/or polymerization is believed to arrest amyloid-mediated impairment of neurons and may enhance removal of oligomers and/or aggregates from the brain, thereby slowing, stopping or reducing amyloid accumulation in the brain.
  • By “amyloid-associated condition” is meant a disease or pathological disorder associated with abnormal amyloid aggregation and/or accumulation. See Eisenberg and Jucker, Cell, 148, 1188-1203 (2012). Amyloid accumulation has been linked to a wide variety of disorders ranging from neurodegenerative disorders to systemic amyloidoses to arthritis. Examples of amyloid-associated conditions include, but are not limited to, Alzheimer's disease (beta amyloid), dementia, diabetes mellitus type 2 (IAPP), Parkinson's disease (alpha-synuclein), idiopathetic myeloma, amyloid polyneuropathy, amyloid cardiomyopathy, systemic senile amyloidosis, transmissible spongiform encephalopathy (e.g., Creutzfeldt-Jakob disease) (PrPSc), Down's syndrome, Huntington's Disease (Huntingtin), medullary carcinoma of the thyroid (calcitonin), atherosclerosis (apolipoprotein), rheumatoid arthritis (serum amyloid A), aortic medial amyloid (Medin), prolactinomas (prolactin), familial amyloid polyneuropathy (FAP) (transthyretin), frontemporal lobar degeneration-tau (Tau), Familial British and Danish dementia (BriPP), hereditary non-neuropathic systemic amyloidosis (lysozyme), dialysis-related amyloidosis (beta 2 microglobulin), Finnish amyloidosis (Gelsolin), atrial amyloidosis (atrial natriuretic factor), transthyretin familial amyloidosis (TTR), hereditary fibrinogen alpha-chain amyloidosis (fibrinogen alpha-chain), motor neuron diseases, lattice corneal dystrophy (keratoepithelin), amyotrophic lateral sclerosis (SOD1), and cerebral amyloid angiopathy (beta amyloid, cystatin). See, e.g., Koo et al., PNAS, 96(18), 9989-9990 (1999); Eisenberg and Jucker, supra. Other examples of amyloid-associated conditions include head injuries (traumatic brain injury); amyloid-β plaques have been found in patients within hours following traumatic brain injury. See Johnson et al., Nature Reviews Neuroscience, 11, 361-370 (2010).
  • As used herein, “treating” and “treatment” refers to any reduction in the severity and/or onset of signs or symptoms associated with an ailment, such as an amyloid-associated condition or an inflammatory or allergic lung disease (described below). Accordingly, “treating” and “treatment” includes therapeutic and prophylactic measures that reduce any of the signs or symptoms. One of ordinary skill in the art will appreciate that any degree of protection from, or amelioration of, an amyloid-associated condition or symptom associated therewith is beneficial to a subject, such as a human patient. For example, a reduction in signs such as detectable amyloid plaques can be beneficial. In another example, an increase in soluble amyloid levels in the cerebrospinal fluid from abnormally low levels to normal, age-appropriate levels can also be beneficial. The quality of life of a patient is improved by reducing to any degree the severity of symptoms in a subject and/or delaying the appearance of symptoms. Accordingly, the method in one aspect is performed as soon as possible after it has been determined that a subject is at risk for an amyloid-associated condition or as soon as possible after the amyloid-associated condition manifests in the subject. For example, due to the rapid development of amyloid-8 plaques in traumatic brain injury patients, prognosis may be improved by initiating treatment as soon as possible after the occurrence of the head injury.
  • Detection of a risk, onset, or presence of amyloid-associated condition is performed using any of a number techniques. Amyloid deposits can be detected ex vivo using, e.g., fluorescent dyes, circular dichroism, and X-ray diffraction. For example, amyloids in biopsied tissue typically demonstrate green birefringence when stained with Congo red and viewed under a polarizing microscope. Amyloid plaques can be detected in vivo using, e.g., magnetic resonance imaging (MRI) (see, e.g., Baltes et al., Methods Mol Biol., 711, 511-33 (2011)) and positron emission tomography (PET) (Kepe et al., Methods Enzymol., 412, 144-60 (2006)). Many of the neurodegenerative diseases referenced herein also are diagnosed by conducting mental status and neuropsychological tests (assessing, e.g., memory and problem solving) and blood tests (e.g., blood tests for Alzheimer's disease biomarker signatures). See Burnham et al., “A blood-based predictor for neocortical Aβ burden in Alzheimer's disease: results from the AIBL study,” Molecular Psychiatry (April 2013).
  • Inflammatory or Allergic Lung Diseases
  • The invention provides methods of treating inflammatory or allergic lung diseases in a patient in need thereof, comprising administering to the patient via inhalation an amount of powder comprising particles of a pharmaceutically acceptable salt or ester of cromolyn as described herein. The dose and/or frequency of administration according to such methods are reduced compared to the conventional dosages and frequencies.
  • Inflammatory or allergic lung diseases include asthma, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), pulmonary fibrosis, cystic fibrosis. Asthma, for example, is a condition associated with inflammation of airway passages in the lungs and airway hyperresponsiveness (AHR). Asthma also is marked by excess mucus production. Symptoms range from minor (e.g., slight shortness of breath) to severe (wheezing, inability to breath, and/or chest tightness) and vary from person to person. During an asthma attack, the lining of airways swell, thereby constricting the passage and reducing airflow to and from the lungs. Asthma is caused or triggered by, for example, infection, allergens, chemical substances and fumes, pollutants, medications, physical exertion, stress, and food additives. Asthma is classified into four general categories: mild intermittent (mild symptoms up to two days/week), mild persistent (symptoms more than twice/week, but not daily, and one or two nighttime episodes/month), moderate persistent (daily symptoms and three or four nighttime symptoms/month), and severe persistent (symptoms throughout most days and frequently at night).
  • As described herein, “treating” and “treatment” refers to any reduction in the severity and/or onset of symptoms associated with inflammatory or allergic lung diseases (e.g., asthma) and, as such, includes therapeutic and prophylactic measures. For example, treatment may result in a reduced number and/or severity of asthmatic attacks in a patient prone to allergy or airway hyperresponsiveness. The method in one aspect is performed as soon as possible after it has been determined that a subject is at risk for inflammatory or allergic lung diseases (e.g., allergy or airway hyperresponsiveness is diagnosed) or as soon as possible after an inflammatory or allergic lung disease (e.g., asthma) manifests in the subject.
  • Combination Therapies
  • The pharmaceutically acceptable salt or ester of cromolyn is optionally administered with one or more additional medicaments. For example for amyloid-associated conditions, additional anti-amyloid agents or anti-inflammatory agents can be administered. For inflammatory or allergic lung diseases, additional anti-asthma agents, or anti-inflammation agents, or other agents that are used to treat airway hyperresponsiveness can be administered.
  • Additional medicaments may be provided in any dosage form, including solid dosage forms (e.g., tablets, capsules and powders) and liquid dosage forms (e.g., solutions, suspensions, emulsions, syrups and elixirs). The additional medicaments may be administered by any known route of administration, including oral (e.g., ingestion or inhalation), injection (e.g., intravenous, intraarterial, intramuscular, subcutaneous, intradermal, intraarticular, intrathecal, epidural, intracerebral, or intraperitoneal), buccal, rectal, topical, transdermal, intranasal, via the pulmonary route, via inhalation, or intraophthalmic. The additional medicaments may be administered concurrently with or sequentially (i.e. before or after) with the pharmaceutically acceptable salt or ester of cromolyn.
  • Additional medicaments include, for example, Levodopa (Sinemet), anticholinergics, Eldepryl, steroids, antihistamines, long-acting or short-acting beta-agonists, immunomodulators (e.g., Omalizumab), and Theophylline.
  • In the case of amyloid-associated conditions, the additional medicament can be a cholinesterase inhibitor (e.g., Donepezil (Aricept®), rivastigmine (Exelon®), galantamine (Razadyne®), or Tacrine (Cognex®)), a NMDA receptor antagonist (e.g., memantine (Namenda®)), a gamma secretase inhibitor (e.g., LY451039 (Semagacestat, Eli Lily)), a metal ionophore (e.g., PBT2 (Prana)), a statin, and/or an endocannabinoid (e.g., arachidonoylethanolamine, tetrahydrocannabinol, 2-arachidonoyl glycerol, 2-arachidonyl glyceryl ether, N-arachidonoyl-dopamine, or virodhamine). Examples of non-steroidal anti-inflammatory drugs, include, but are not limited to, ibuprofen, acetylsalicylic acid, diflunisal, salsalate, dexibuprofen, naproxen, fenoprofen, ketoprofen, dexketoprofen, flurbiprofen, oxaprozin, loxoprofen, indomethacin, tolmetin, sulindac, etodolac, ketorolac, diclofenac, nabumetone, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam, isoxicam, mefenamic acid, meclofenamic acid, flufenamic acid, tolfenamic acid, celecoxib, licofelone, hyperforin, and figwort. Preferably, the non-steroidal anti-inflammatory drug is administered orally (via ingestion or inhalation). The non-steroidal anti-inflammatory drug (e.g., ibuprofen) can be administered in an amount of about 5 mg to about 80 mg per day, for example, about 5 mg to about 60 mg per day, about 5 mg to about 50 mg per day, about 5 mg to about 40 mg per day, about 5 mg to about 30 mg per day, about 5 mg to about 20 mg per day, and/or about 5 mg to about 15 mg per day. The non-steroidal anti-inflammatory drug (e.g., ibuprofen) may be administered 1 to 4 times per day, such as 1 to 2 times per day. For example, ibuprofen may be administered in a once-daily dose of about 5 mg to 20 mg, preferably in a once-daily dose of about 10 mg.
  • Blister Packs and Kits
  • The invention further provides a group of individual delivery units, for example, a blister pack comprising blisters, containing a pharmaceutically acceptable salt or ester of cromolyn. Blister packs are known in the art, and generally comprise a solid support comprising a plurality of spaced bubbles or wells (collectively referred to herein as “blisters”) for carrying a predetermined amount of medicament. A film or membrane seals the wells, and is susceptible to puncture or release from the solid support to make the medicament available for delivery. The specific shape, proportions, and dimensions of the blister pack and the individual blisters can be adjusted for use in a particular delivery device. For example, the blister pack is optionally provided as a coil or a circular (e.g., carousel) cartridge for insertion in a dry powder inhaler, and the blisters are shaped as inverted cones or domes. Additionally, the number of blisters (corresponding to the number of doses) may be varied.
  • The blister pack is composed of a material that protects the contents of the blisters from exposure to the environment and is compatible for use with an inhalation device for delivering cromolyn to a patent. Suitable materials include, but are not limited to, PVC (polyvinyl chloride), PVC/PVDC (polyvinylidene chloride) blends, PE (polyethylene), PP (polypropylene), polystyrene, cellophane, polyester (e.g. a polyester terephthalate), paper, polyamide, PET (polyethylene terephthalate), COC (cyclic olefin copolymer), metallic (e.g., aluminum) foil and any blend thereof. Different materials may be layered to form individual blisters or the blister pack, if desired. Blister packs are further described in, for example, U.S. Pat. Nos. 5,497,763; 7,080,644; 7,828,150; 7,931,022; and 8,291,900, and International Patent Publication Nos. WO 1999/23180 and WO 1989/01348 (all of which are hereby incorporated by reference in their entirety, and particularly with respect to their respective descriptions of blister packs).
  • Generally, blisters have a maximum capacity of about 15 to 20 mg of ingredients, which includes both the active ingredient (i.e. cromolyn, or its pharmaceutically acceptable salt or ester) and pharmaceutically acceptable excipients.
  • In the context of the invention, the blisters of the blister pack contain about 3 mg to about 20 mg of a pharmaceutically acceptable salt or ester of cromolyn, for example, 3 mg to about 19 mg, 3 mg to about 18 mg, 3 mg to about 17 mg, about 3 mg to about 15 mg, about 3 mg to about 14 mg, about 3 mg to about 13 mg, about 3 mg to about 12 mg, about 3 mg to about 11 mg, about 3 mg to about 10 mg, about 3 mg to about 9 mg, about 3 mg to about 8 mg, about 3 mg to about 7 mg, about 4 mg to about 20 mg, about 6 mg to about 18 mg, about 8 mg to about 17.5 mg, or about 16.1 mg to about 19.9 mg of the pharmaceutically acceptable salt or ester of cromolyn. Optionally, additional amounts of pharmaceutically acceptable excipients are included in the blisters with the cromolyn. In some embodiments, a capsule is used instead of a blister. Capsules for inhaler devices are well known in the art.
  • The pharmaceutically acceptable salt or ester of cromolyn is provided in a solid dosage form, preferably a powder containing particles of the pharmaceutically acceptable salt or ester of cromolyn as described herein. The powder optionally comprises one or more pharmaceutically acceptable excipients, as described above. The pharmaceutically acceptable excipients, when present, are included in the powder in a total amount of about 0.1% to about 80% by weight, about 1% to about 80% by weight, about 5% to about 80% by weight, about 10% to about 80% by weight, about 15% to about 80% by weight, about 20% to about 80% by weight, about 25% to about 80% by weight, about 30% to about 80% by weight, about 35% to about 80% by weight, about 40% to about 80% by weight, about 20 to about 75% by weight, about 20% to about 70/o by weight, about 20% to about 65% by weight, about 20% to about 60% by weight, about 25% to about 55% by weight, about 30% to about 50% by weight, about 35% to about 45% by weight, and/or about 40% by weight.
  • Alternatively, in some embodiments herein, the cromolyn is provided in liquid solution form.
  • In one example, the invention further provides a kit comprising a blister pack, such as the blister pack described herein, and a dry powder inhaler (DPI) device. In some embodiments, a kit comprises a capsule pack and a dry powder inhaler (DPI) device. In various embodiments, the device is an active dry powder inhaler device, such as a dry powder inhaler device that comprises a chamber comprising a piezoelectric vibrator for deaggregating a dry powder and an air flow passageway in which the deaggregated powder is picked up and carried for inhalation by a patient. The kit optionally includes one or more additional medicaments, such as a non-steroidal anti-inflammatory drug (e.g., ibuprofen, acetylsalicylic acid, diflunisal, salsalate, dexibuprofen, naproxen, fenoprofen, ketoprofen, dexketoprofen, flurbiprofen, oxaprozin, loxoprofen, indomethacin, tolmetin, sulindac, etodolac, ketorolac, diclofenac, nabumetone, piroxicam, meloxicam, tenoxicam, droxicam, lornoxicam, isoxicam, mefenamic acid, meclofenamic acid, flufenamic acid, tolfenamic acid, celecoxib, licofelone, hyperforin, and figwort). Such additional medicaments may be provided in any known dosage form, including solid dosage forms (e.g., tablets, capsules and powders) and liquid dosage forms (e.g., solutions, suspensions, emulsions, syrups and elixirs).
  • EXAMPLES Example 1
  • A Next Generation Pharmaceutical Impactor (NGI) cascade impactor device (MSP Corporation, Shoreview, Minn., US) was used to assess delivery of cromolyn by four different inhaler devices.
  • Delivery of cromolyn by a single-use passive dry powder inhaler device was assessed. The passive inhaler device included an active particle dispersion mechanism (ACTIVEMESH, Aespira) involving breath-driven beating of a mesh package containing the powder to be delivered.
  • A blend containing disodium cromoglycate (DSCG) (Cambrex) and Lactohale LH300 lactose (Friesland Foods Domo) was prepared by blending in an Alpine Picoline high shear mixer (module Picomix) (Hosokawa Alpine, Augsburg, Germany) at a speed of 4000 rpm for 3 minutes. The blend contained 80:20 (wt./wt.) DSCG:lactose. Neat DSCG also was tested. The samples (neat cromolyn or cromolyn:lactose blend) were loaded into the passive dry powder inhaler device and tested using an NGI cascade impactor device at a flow rate of approximately 100 L/min for 2.4 seconds.
  • The results of testing are provided in Table 1. The passive inhaler device delivered only 3% of the initial dose of neat DSCG and only 6% of the initial dose of blended cromolyn to Stage 4 and higher of the NGI cascade impactor device.
  • TABLE 1
    Initial Fine Particle Stages 4 to 8
    DSCG:lactose DSCG dose Dose <5 μm [μg] [μg] MMAD
    [wt./wt.] [mg] (% of initial dose) (% of initial dose) [μm]
    100:0 5 430 (9%)  150 (3%) >4
     80:20 4 400 (10%) 240 (6%) >4
  • Delivery of cromolyn by a PROHALER pre-metered multidose passive dry powder inhaler device (Aptar) was assessed. The PROHALER inhaler was initially expected to more efficiently deliver cromolyn because the device creates rapidly fluctuating airflow/shear and turbulence to deagglomerate the particles emitted from the blister prior to oral inhalation by the patient. The PROHALER inhaler also included breath-triggered opening of a dose and a blister cartridge system.
  • Two blends containing disodium cromoglycate (DSCG) (Cambrex) and lactose monohydrate (DMV-Fonterra Excipients) (grade: lactohale 200 with 13% of fines) were prepared by blending at a speed of 90 rpm for 60 minutes. The blends differed in the relative amounts of DSCG and lactose, and contained either 50:50 (wt./wt.) DSCG:lactose or 20:80 (wt/wt.) DSCG:lactose. The blends were filled into blisterstrips, which were then assembled into a PROHALER inhaler device and tested using an NGI device at a flow rate of approximately 35 L/min.
  • The results of testing are provided in Table 2. The emitted dose represents the total quantity of DSCG fired into the impactor. The fine particle fraction is calculated as follows: fine particle fraction=100%*fine particle dose/emitted dose. A fine particle dose with particles less than 3 μm in size corresponds approximately to the amount of cromolyn delivered to Stage 4 and higher of the NGI device. Thus, the PROHALER inhaler device delivered only about 4.4% of the initial dose of blended cromolyn (50:50 blend) and only about 9.6% of the initial dose of blended cromolyn (20:80 blend) to Stage 4 and higher of the NGI cascade impactor device.
  • TABLE 2
    Initial Emitted Fine Particle Fine Particle
    DSCG:lactose DSCG dose dose [mg] Dose <3 μm [μg] Fraction <3 μm MMAD
    [wt./wt] [mg] (% of initial dose) (% of initial dose) [%] [μm]
    50:50 3.9 2.4 (61%) 170 (4.4%) 8 3.9
    20:80 1.6 1.1 (69%) 154 (9.6%) 15 3.1
  • In addition, delivery of cromolyn by a TWISTER inhaler device (Aptar) was assessed. The TWISTER inhaler device is a capsule-based dry powder inhaler. Compared to blisters, capsules typically have much higher drug loading capacities.
  • A blend containing disodium cromoglycate (DSCG) and lactose was prepared. The blend was assembled into a TWISTER inhaler device and tested using an NGI cascade impactor device at a flow rate of approximately 30 L/min.
  • The results of testing are provided in Table 3. The TWISTER inhaler device delivered only about 24% of the initial dose of blended cromolyn to Stage 4 and higher of the NGI cascade impactor device.
  • TABLE 3
    Initial Emitted Fine Particle Fine Particle
    DSCG:lactose DSCG dose dose [mg] Dose <3 μm [μg] Fraction <3 μm MMAD
    [wt./wt.] [mg] (% of initial dose) (% of initial dose) [%] [μm]
    20:80 4.4 3.6 (82%) 1065 (24%) 29 >4
  • Delivery of cromolyn by an active dry powder inhaler device also was assessed. The inhaler device included injection molded plastic components, electronics, a battery, and drug filled blisters. The inhaler included a lever arm, upon actuation of which the blister was placed in contact with a piezoelectric vibrator within the device and then pierced by needles. The inhaler also included an airflow sensor which automatically turned on the piezoelectric vibrator after a minimum inspiratory flow rate was exceeded. Activation of the piezoelectric vibrator deaggregated the powder particles and aerosolized them out of the blister and into the inspiratory airstream. The inhaler included visual feedback at the start of dosing and as confirmation of successful completion of dosing, thereby facilitating improved patient compliance. Similar devices have been shown to deliver a consistent amount of a drug at each of the different flow rates tested, ranging from 20 L/min to 60 L/min.
  • A blend containing disodium cromoglycate (DSCG) (Cambrex) and lactose monohydrate (DFE Pharma) (grade: lactohale LH201) was prepared by blending at a speed of 150 rpm for 15 minutes. The blend contained 60:40 (wt/wt.) DSCG:lactose. Neat DSCG also was tested. The samples (neat cromolyn or cromolyn:lactose blend) were filled into blisters, which were then assembled into the inhaler device and tested using an NGI device at a flow rate of approximately 30 L/min for 2 seconds.
  • The results of testing are provided in Table 4. The fine particle dose is the amount of cromolyn recovered below a diameter of 5 μm. The fine particle fraction <5 μm is provided as a percentage of the amount of cromolyn recovered from the NGI cascade impactor device (i.e., the delivered dose). The active inhaler device delivered 42% of the initial dose of neat DSCG and 43% of the initial dose of blended cromolyn to Stage 4 and higher of the NGI cascade impactor device. Thus, in contrast to the results provided in Tables 1 to 3, the results for the active dry powder inhaler device demonstrate delivery of over 1.5 mg and over 30% of the administered dose to Stages 4 and higher of the NGI cascade impactor device.
  • TABLE 4
    Fine Fine Particle Stages 4 to 8
    Initial Particle Fraction <5 μm [μg]
    DSCG:lactose DSCG dose Dose <5 μm [% of delivered (% of initial MMAD
    blend [mg] [μg] dose] dose) [μg]
    100:0 5 2321 59 2098 (42%) 2.6
     60:40 5.8 2858 55 2505 (43%) 2.8
  • Example 2 Pharmacokinetic Study of Cromolyn
  • Materials and Methods
  • Route of Administration.
  • An FDA-approved route of administration for cromolyn is oral inhalation using a capsule-based dry powder inhaler, with 20 mg cromolyn loaded per capsule. Studies have shown that with high inspiratory rates, the inhaled cromolyn is delivered to the human lung, with 10-15% of the inhaled drug-delivered dose absorbed into the bloodstream.
  • Cromolyn Preparation.
  • Cromolyn is supplied by Cambrex Proforma (Milan, Italy) and used to formulate a 17.1 mg dose of cromolyn for inhalation using inhalation grade excipients.
  • Cromolyn was micronized at Cambrex to achieve an average particle size of 5 microns or less. It is important for particles to be less than 10 microns, with the majority of particles falling between 2 and 5 microns, since this is necessary for successful deposition to the secondary bronchi of the respiratory tract following inhalation.
  • Cromolyn capsules for inhalation were manufactured by Pharmaterials, Ltd (Reading, UK). Initial characterization of the manufactured Cromolyn was performed by Pharmaterials and was found to be crystalline and hygroscopic material with particle size distribution suitable for inhalation (d50<5 μm and d90<10 μm). Therefore, there was a need to use hydrophobic excipient in the powder formulation to improve product performance and stability. Addition of hydrophobic excipient offers inherent resistance of dry powder inhalation formulations to negative effect of moisture to such formulations. Magnesium stearate was chosen as suitable hydrophobic excipient since it is commercially used in dry powder inhaler (DPI) products. Additionally, its safety profile is well studied and demonstrated for use in inhalation products. Lactose monohydrate was additionally used as diluent for this case. Each manufactured capsule of ALZT-OP1a contains 17.1 mg of cromolyn. The formulations (Cromolyn and Placebo) are shown in Table 5.
  • TABLE 5
    ALZT-OP1a (Cromolyn) Formulation
    ALZT-OP1a Composition
    Placebo Drug Product
    Quality mg/ mg/
    Component Standard Function % w/w capsule % w/w capsule
    Cromolyn sodium USP Active 58.0 17.1a
    (micronized)
    Lactose NF Diluent 98.0 44.1 40.0 12.8
    monohydrate
    Magnesium NF Stabilizer 2.0 0.9 2.0 0.6
    stearate
    (micronized)
    Hydroxypropyl In-house Encapsulation NA NA NA NA
    methylcellulose
    capsuleb
    Total 100% 45 100% 32
    USP: United States Pharmacopeia;
    NF: National Formulary
    aWeight of cromolyn sodium, USP per capsules is 17.1 mg on an anhydrous basis (18.6 mg per capsule on as-is basis).
    bHydroxypropyl methylcellulose capsule functions only to meter and deliver the drug product through the dry powder inhaler and is not ingested during administration.
  • AZHALER.
  • The AZHALER DPI (239700001AB-Rs01) is a monodose inhaler device manufactured by Plastiape S.p.a (Italy) for use with ALZT-OP1a (cromolyn). It is a single-dose device used to deliver the inhalation powder to the patient's lung in form of an aerosol. The inhaler consists of a container, a valve, and a mouthpiece. In this study the low resistance model (80 L airflow resistance) was used. These device reaches a pressure drop of 4 kPa at c.80 L/min, and is suitable for use on a wide range of patient population i.e. including elderly, children and patients with severe respiratory impairment
  • Dosage.
  • With AZHALER, the results show about 4-5 mg cromolyn (in the impactor fractions with <3 micron size particles needed for systemic uptake) per 17.1 mg of API (NGI stage 4-MOC see Table 1)
      • 4-5 ee-3 g/512 g/mol=7.8-9.8 micromoles of cromolyn plasma levels
  • Assuming 0.2-1% uptake in brain from plasma=16-98 nanomoles divided by/0.5 L brain=11-66 nM cromolyn/L in brain (per day). Therefore, 17.1 mg cromolyn inhaled with AZHALER device is estimated to result in 11-66 nM concentrations in the brain.
  • Study Design.
  • The primary objective of the Phase I open-labeled study was to determine the PK of cromolyn in plasma and CSF, following administration of a single 17.1 mg inhaled dose of cromolyn. The study also evaluated the PK of cromolyn in plasma and CSF, following administration of 34.2 mg of cromolyn (administration of two consecutive inhaled doses of 17.1 mg taken not more than two minutes apart). The secondary objective of the study was to evaluate the safety and tolerability of a single or double dose of cromolyn following administration. The study population consisted of 24 normal, healthy volunteers between the ages of 55-75 in good general health, without respiratory disease, confirmed by pulmonary function testing.
  • Results
  • The results indicated that the PK profile of cromolyn in plasma was characterized by a rapid absorption phase, with an average Cmax of 46.7±33.0 ng/ml (range 14.0-133.0 ng/ml) at 22.8±16.6 min (range 6-60 min) after single dose (17.1 mg) inhalation and an average Cmax of 96.8±46.2 ng/ml (range 36.1-236.0 ng/ml) at 22.2±19.4 min (range 6-60 min) after double dose (34.2 mg) inhalation. The average apparent t % of cromolyn in plasma was 1.756±0.9 h (range 0.6-3.7 h) following single dose (17.1 mg) inhalation and 1.91±0.7 h (range 0.7-3.8 h) following double dose (34.2 mg) inhalation, indicating moderate clearance. The average AUC0-inf of cromolyn in plasma increased with the dose increase from 195.71±97.33 h*ng/ml (range 93.3-287.0 h*ng/ml) to 284.55±91.29 h*ng/ml (range 154.8-443.3 h*ng/ml) following single (17.1 mg) and double (34.2 mg) dose inhalation, respectively.
  • Cromolyn plasma PK parameters are summarized in Table 6.
  • TABLE 6
    Plasma Pharmacokinetic Parameters for Cromolyn
    AUC0-inf T0.5 Tmax Cmax Cl/F Vd/F
    Dose (h*ng/ml) (h) (h) (ug/L) (mL/h) (mL)
    17.1 mg
    Mean (SD) 195.71 1.75 0.38 46.69 108532.36 137487.60
    (97.33) (0.847) (0.276) (32.965) (65822.400) (5916.345)
    Median 206.92 1.56 0.25 36.20 82640.18 134426.90
    Min, Max 93.3, 287.0 0.6, 3.7 0.1, 1.0 14.0, 133.0 59593.2, 133728.6,
    183363.7 144307.3
    34.2 mg
    Mean (SD) 284.55 1.91 0.37 96.75 132178.06 330386.32
    (91.29) (0.695) (0.323) (46.217) (42254.442) (133203.378)
    Median 262.31 1.80 0.23 76.50 130382.20 263533.00
  • Detectable levels of cromolyn in CSF were indicative of drug delivery to the brain. The average Cmax in CSF during the observed time interval of up to 4 hours was 0.24±0.077 ng/ml (range 0.2-0.4 ng/ml) at 3.72±0.704 h after single dose (17.1 mg) inhalation and 0.34±0.171 ng/ml (range 0.2-0.6 ng/ml) at 3.45±0.952 h after double dose (34.2 mg) inhalation. The observation period (0-4 hours) for the CSF samples was too short to allow for determination of t1/2. The cromolyn AUC in plasma was 675 and 809 times higher than in CSF 2 hours after single (17.1 mg) and double (34.2 mg) dose inhalation and 232 and 299 times higher 4 hours after single and double dose inhalation, respectively.
  • These results indicate that inhaled cromolyn is transported via the deep lung to the blood, and then to the brain and CSF. The concentration of cromolyn in CSF increased up to the 4 hours of the lumber puncture.
  • Example 3 A Phase III Safety and Efficacy Study of ALZT-OP1 in Subjects with Evidence of Early Alzheimer's Disease Study Design.
  • Study Subjects.
  • This Phase I study is designed as a randomized, double-blinded, placebo-controlled study for subjects with evidence of early AD.
  • Subjects will be randomly assigned to the Group I arm which will consist of Cromolyn for inhalation, OR the Group I placebo arm, which will consist of inhaled placebo.
  • TABLE 7
    Treatment Groups
    Number of
    Group subjects Treatment
    I 150 ALZT- OP1a (q.d. inhalation of active cromolyn)
    II 150 q.d. inhalation of placebo
  • A minimum of 200 evaluable subjects will be randomized to receive one of two possible treatment assignments containing active study drug or placebo.
  • To account for subject dropouts (estimated rate of 30%), it is anticipated that up to 300 (or 150 subjects per treatment arm) may be recruited and randomized, to achieve a minimum of 100 evaluable subjects per treatment arm.
  • Dosage and Formulation.
  • Study drugs are dispensed as 12 or 24-week supplies at Day 1, Weeks 12, 24, and 48. Cromolyn powder blend or placebo will be filled into capsules for use with an AZHALER dry powder inhaler device. Each capsule will contain 17.1 mg of the active product ingredient (cromolyn) and inhalation grade lactose monohydrate as well as magnesium stearate as excipients, or placebo (lactose alone). The once-daily cromolyn dose to be tested in this study is about 21%, the dose from the four times daily approved dose level (80 mg cromolyn total per day [17.1/80]) for the treatment of asthma.
  • Study Regimen.
  • The total duration of subject participation is expected to be approximately 74 weeks or 7 visits. All subjects are required to have an initial screening visit (Day −21 to Day −1) to determine eligibility and a baseline visit (Day 1) to confirm eligibility prior to treatment start, and then asked to complete 72 weeks of daily dosing that requires them to return to the clinic at weeks 4, 12, 24, 48 and 72.
  • Study drug compliance and concomitant medication use will be evaluated at scheduled visits throughout the study.
  • Study Objective(s).
      • To evaluate the efficacy by CDR-SB assessment of a treatment regimen of oral inhaled cromolyn, compared to placebo.
      • To determine whether this treatment regimen slows down, arrests or reverses cognitive and functional decline in subjects with evidence of early AD.
  • Study Endpoints.
  • The primary endpoint of this study is a significant stabilization/improvement in cognitive and functional performance, as measured by the Clinical Dementia Rating test (CDR-SB), from baseline to Week 72, for subjects in the cromolyn treatment group compared to the placebo group.
  • Assessment of Efficacy
  • Methods for Assessing, Recording, and Analyzing Efficacy Parameters.
  • The primary efficacy variable will be analyzed using a mixed-effect model for repeated measures (MMRM) approach. The model will include the fixed effects treatment, visit, treatment by visit interaction, region (US, Europe, Asia/Pacific) and the covariates APOe4 (one or more alleles, no alleles), anti-dementia drug use (absent, present), baseline CDR-SB and age assuming an unstructured correlation matrix.
  • Clinical Dementia Rating (CDR).
  • The CDR is a clinical staging instrument for dementia. It characterizes six domains of cognitive and functional performance: Memory, Orientation, Judgment & Problem Solving, Community Affairs, Home & Hobbies, and Personal Care. The necessary information to make each rating is obtained through a semi-structured interview of the patient and a reliable informant or collateral source (e.g., a family member). The CDR questionnaire provides descriptive anchors that guide the clinician in making appropriate ratings based on interview data and clinical judgment. The CDR is conducted at Screening and at Weeks 12, 24, 48, and 72.
  • Global Clinical Dementia Rating (CDR):
  • Disease Staging. The Washington University Global CDR score (5 stages: 0, 0.5, 1, 2, 3) will be used as an inclusion criterion to screen and select a subject population consistent with early AD. Subjects are required to have a Global CDR score of 0.5 (mildly impaired), with the Memory Box score being at least 0.5. Each global score will be calculated using the Washington University online algorithm.
  • Clinical Dementia Rating-Sum of Boxes (CDR-SB):
  • Disease Severity. The CDR-SB score (score range 0-18) will serve as the primary endpoint for the study and will be used for study group comparisons, monitoring disease progression, and evaluating treatment effect between active and placebo groups. A CDR-SB score of 0 represents normal functioning and a score of 18 represents severe impairment. Overall, the CDR-SB is expected to provide a more detailed quantitative general index than the Global CDR in patients with mild dementia. The CDR-SB is conducted at Screening and at Weeks 12, 24, 48, and 72.
  • Mini-Mental State Examination (MMSE).
  • The MMSE is a brief examination intended to evaluate an adult participant's level of cognitive functioning. The test is performed in following areas: orientation in time and place, learning and immediate recall, mental control and concentration, short-term recall, naming ability, language expression, verbal comprehension, writing comprehension, writing ability, and visual-spatial coordination. Scores range from 0 (maximum cognitive deficit) to 30 (no cognitive deficit). The MMSE is conducted at Screening and at Weeks 12, 24, 48, and 72.
  • Wechsler Memory Scale-Third Edition (WMS-III):
  • Logical Memory II (LM II). The Wechsler Memory Scale-Third Edition (WMS-III) is a neuropsychological test designed to measure different memory functions. The complete WMS-III is made up of eleven subtests:
      • 1) Brief Cognitive Status Exam (Optional),
      • 2) Visual Reproduction I,
      • 3) Logical Memory I,
      • 4) Spatial Addition,
      • 5) Visual Reproduction II,
      • 6) Logical Memory II,
      • 7) Verbal Paired Associates I,
      • 8) Designs I,
      • 9) Symbol Span,
      • 10) Verbal Paired Associates II,
      • 11) Designs II.
  • In the current study, the Logical Memory II subtest will be used to assess delayed memory function. The WMS-III Logical Memory I (Immediate Recall) and Logical Memory II (Delayed Recall) will both be administered at Screening (only), but only the Logical Memory II score will be used as an inclusion criterion. LM H scores acceptable for inclusion will be categorized by subjects' years of education.
  • Logical Memory II will be administered 30 minutes after the administration of Logical Memory I.
  • CSF Biomarker Analysis.
  • CSF Aβ—42 levels ≥200 pg/mL and ≤600 pg/mL are required for study entry. Following consent for the CSF collection procedure, subjects who meet all other study entry requirements will undergo lumbar puncture for CSF collection at Screening (Day −21 to Day −1) to confirm study eligibility. On a voluntary basis, subjects may also provide CSF samples at End of Study (Week 72 or within 1 week following Week 72) for follow-up and change from baseline analyses. Duplicate CSF samples will be collected at Screening and End of Study. One Screening CSF sample will be analyzed to confirm study eligibility; the back-up sample will be stored frozen at −20° C. or −80° C. until study completion, at which point the sample will be shipped on dry ice to a central testing facility. End of Study samples will be stored frozen at −20° C. or −80° C. for subsequent analysis. Lumbar puncture should be performed according to clinical site protocols and follow current CSF sample standardization procedures.
  • The cromolyn monotherapy administered daily at the dose of 17.1 mg to patients having early stage AD can be as effective in stabilization/improvement of cognitive and functional performance as the combination therapy of cromolyn and ibuprofen.

Claims (21)

1. A solution for oral and/or nasal inhalation, comprising about 5 mg/mL to about 20 mg/mL of a pharmaceutically acceptable salt or ester of cromolyn in the form of solution droplets, the majority of solution droplets having a Mass Median Aerodynamic Diameter (MMAD) of about 2 to about 5 microns, wherein said solution is capable of being delivered using a device that deposits (a) at least 3.0 mg and (b) at least 20% by weight of the administered amount of pharmaceutically acceptable salt or ester of cromolyn to Stage 4 and higher of a Next Generational Pharmaceutical Impactor (NGI) cascade impactor device at a flow rate of 60-80 L/min.
2. The solution of claim 1, wherein the solution comprises about 8.5 mg/mL of the pharmaceutically acceptable salt or ester of cromolyn.
3. The solution of claim 1, wherein the device is a nebulizer selected from the group consisting of a jet nebulizer, an ultrasonic nebulizer, and an electronic nebulizer.
4. The solution of claim 1, wherein the device is an inhalation device that delivers the solution as an aerosol.
5. The solution of claim 1, wherein the solution further comprises one or more pharmaceutically acceptable excipients.
6. The solution of claim 1, wherein the solution is hypotonic or isotonic.
7.-8. (canceled)
9. The solution of claim 1, wherein the solution does not comprise an active ingredient other than the pharmaceutically acceptable salt or ester of cromolyn.
10. The solution of claim 1, wherein the solution does not comprise a non-steroidal anti-inflammatory drug.
11. The solution of claim 1, wherein the solution does not comprise ibuprofen.
12. A method of delivering cromolyn to a patient in need thereof, the method comprising administering the solution of claim 1 to the patient via oral and/or nasal inhalation, wherein the solution droplets have a mass median particle size (D50) of less than about 5 microns; and wherein the solution is administered using a device that deposits about 4 mg to about 5 mg of cromolyn to the secondary bronchi at a flow rate of 60-80 L/min.
13. The method of claim 12, wherein the method comprises administering about 17.1 mg to of the pharmaceutically acceptable salt or ester of cromolyn, and wherein said solution optionally comprises one or more pharmaceutically acceptable excipients.
14. The method of claim 12, wherein the device is a nebulizer selected from the group consisting of a jet nebulizer, an ultrasonic nebulizer, and an electronic nebulizer.
15. The method of claim 12, wherein administering comprises suspending the solution droplets into an inhaled gas stream.
16.-27. (canceled)
28. The method of claim 12, wherein said solution is administered at a frequency of one or two times daily.
29. A method of treating or preventing a disease or disorder in a subject in need thereof, comprising administering the solution of claim 1, wherein the disease or disorder is an amyloid-associated condition, an inflammatory lung disease, or an allergic lung disease.
30. The method of claim 29, wherein the disease or disorder is an amyloid-associated condition selected from dementia, a motor neuron disease, and traumatic brain injury.
31. The method of claim 29, wherein the amyloid-associated condition is selected from Alzheimer's disease, diabetes mellitus type 2, Parkinson's disease, amyloid polyneuropathy, amyloid cardiomyopathy, systemic senile amyloidosis, transmissible spongiform encephalopathy, Down's syndrome, Huntington's Disease, medullary carcinoma of the thyroid, atherosclerosis, rheumatoid arthritis, aortic medial amyloid, prolactinoma, familial amyloid polyneuropathy, frontotemporal lobar degeneration-tau, Familial British and Danish dementia, hereditary non-neuropathic systemic amyloidosis, dialysis-related amyloidosis, Finnish amyloidosis, atrial amyloidosis, transthyretin familial amyloidosis, hereditary fibrinogen alpha-chain amyloidosis, lattice corneal dystrophy, amyotrophic lateral sclerosis, and cerebral amyloid angiopathy.
32. The method of claim 29, wherein the disease or disorder is an inflammatory lung disease or an allergic lung disease.
33. The method of claim 32, wherein the inflammatory lung disease or the allergic lung disease is selected from asthma, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), pulmonary fibrosis, and cystic fibrosis.
US16/733,667 2013-05-23 2020-01-03 Cromolyn compositions and methods thereof Abandoned US20200383908A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US16/733,667 US20200383908A1 (en) 2013-05-23 2020-01-03 Cromolyn compositions and methods thereof
US17/108,895 US11013686B2 (en) 2013-05-23 2020-12-01 Cromolyn compositions and methods thereof
US17/972,772 US20230248646A1 (en) 2013-05-23 2022-10-25 Cromolyn compositions and methods thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201361826798P 2013-05-23 2013-05-23
PCT/US2014/039118 WO2015002703A1 (en) 2013-05-23 2014-05-22 Methods for delivering cromolyn
US201514893487A 2015-11-23 2015-11-23
US15/830,980 US10525005B2 (en) 2013-05-23 2017-12-04 Cromolyn compositions and methods thereof
US16/733,667 US20200383908A1 (en) 2013-05-23 2020-01-03 Cromolyn compositions and methods thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/830,980 Continuation US10525005B2 (en) 2013-05-23 2017-12-04 Cromolyn compositions and methods thereof

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US17/108,895 Continuation US11013686B2 (en) 2013-05-23 2020-12-01 Cromolyn compositions and methods thereof
US17/972,772 Continuation US20230248646A1 (en) 2013-05-23 2022-10-25 Cromolyn compositions and methods thereof

Publications (1)

Publication Number Publication Date
US20200383908A1 true US20200383908A1 (en) 2020-12-10

Family

ID=62239947

Family Applications (4)

Application Number Title Priority Date Filing Date
US15/830,980 Active US10525005B2 (en) 2013-05-23 2017-12-04 Cromolyn compositions and methods thereof
US16/733,667 Abandoned US20200383908A1 (en) 2013-05-23 2020-01-03 Cromolyn compositions and methods thereof
US17/108,895 Active US11013686B2 (en) 2013-05-23 2020-12-01 Cromolyn compositions and methods thereof
US17/972,772 Pending US20230248646A1 (en) 2013-05-23 2022-10-25 Cromolyn compositions and methods thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/830,980 Active US10525005B2 (en) 2013-05-23 2017-12-04 Cromolyn compositions and methods thereof

Family Applications After (2)

Application Number Title Priority Date Filing Date
US17/108,895 Active US11013686B2 (en) 2013-05-23 2020-12-01 Cromolyn compositions and methods thereof
US17/972,772 Pending US20230248646A1 (en) 2013-05-23 2022-10-25 Cromolyn compositions and methods thereof

Country Status (1)

Country Link
US (4) US10525005B2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11013686B2 (en) 2013-05-23 2021-05-25 The General Hospital Corporation Cromolyn compositions and methods thereof
US11110097B2 (en) 2012-10-25 2021-09-07 The General Hospital Corporation Combination therapies for the treatment of alzheimer's disease and related disorders
US11291648B2 (en) 2018-07-02 2022-04-05 The General Hospital Corporation Powdered formulations of cromolyn sodium and alpha-lactose
US11666669B2 (en) 2013-10-22 2023-06-06 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment
US11679095B2 (en) 2016-08-31 2023-06-20 The General Hospital Corporation Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases
US11801316B2 (en) 2009-01-29 2023-10-31 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113038945A (en) * 2018-09-05 2021-06-25 通用医疗公司 Methods of treating cytokine release syndrome
AU2019397436A1 (en) * 2018-12-10 2021-07-22 The General Hospital Corporation Cromolyn esters and uses thereof
WO2021247601A1 (en) * 2020-06-02 2021-12-09 Model Medicines, Inc. Methods and compositions for treating rna viral infections

Family Cites Families (116)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1018875A (en) 1911-02-13 1912-02-27 Francois Edouard Cardon Finger-ring construction.
US1005853A (en) 1911-06-24 1911-10-17 Louis B Lewis Educational toy.
GB1144905A (en) 1965-03-25 1969-03-12 Fisons Pharmaceuticals Ltd Substituted bis-(2-carboxy-chromonyl-oxy) derivatives and preparation and pharmaceutical compositions thereof
US3686412A (en) 1965-03-25 1972-08-22 Colin Fitzmaurice Compositions containing bis-chromonyl compounds for inhibiting antigen-antibody reactions
GB1242211A (en) 1967-08-08 1971-08-11 Fisons Pharmaceuticals Ltd Pharmaceutical composition
US3957965A (en) 1967-08-08 1976-05-18 Fisons Limited Sodium chromoglycate inhalation medicament
GB1257162A (en) 1968-02-16 1971-12-15
US4120285A (en) 1976-11-01 1978-10-17 Owens-Illinois, Inc. Modular tubular solar energy collector apparatus
DE3104294A1 (en) 1981-02-07 1982-08-19 Hoechst Ag, 6000 Frankfurt SUBSTITUTED DIAZASPIRODECANE, THEIR PRODUCTION, THEIR USE AS STABILIZERS FOR ORGANIC POLYMERS, AND THE STABILIZED POLYMERS
US4429545A (en) 1981-08-03 1984-02-07 Ocean & Atmospheric Science, Inc. Solar heating system
BE897058A (en) 1982-06-25 1983-12-16 Sandoz Sa USE OF A SPIRO-SUCCINIMIDE DERIVATIVE IN THE TREATMENT OF ALZHEIMER-TYPE DEMENTIA
US4996296A (en) 1983-07-27 1991-02-26 Yeda Research & Development Co., Ltd. Cromolyn binding protein in highly purifed form, and methods for the isolation thereof
US5231170A (en) 1986-08-27 1993-07-27 Paul Averback Antibodies to dense microspheres
US4919915A (en) 1987-03-03 1990-04-24 Paul Averback Method for detecting the ability to prevent red-to-green congophilic birefringence
ES2087911T3 (en) 1989-04-28 1996-08-01 Riker Laboratories Inc DRY DUST INHALATION DEVICE.
US5376386A (en) 1990-01-24 1994-12-27 British Technology Group Limited Aerosol carriers
DE4405387A1 (en) 1994-02-19 1995-08-24 Hoechst Ag Process for the preparation of polyalkyl-1-oxa-diazaspirodecane compounds
US6168776B1 (en) 1994-07-19 2001-01-02 University Of Pittsburgh Alkyl, alkenyl and alkynyl Chrysamine G derivatives for the antemortem diagnosis of Alzheimer's disease and in vivo imaging and prevention of amyloid deposition
CA2174583A1 (en) 1995-05-05 1996-11-06 Alexander Chucholowski Sulfuric acid esters of sugar alcohols
US6026809A (en) 1996-01-25 2000-02-22 Microdose Technologies, Inc. Inhalation device
US6126919A (en) 1997-02-07 2000-10-03 3M Innovative Properties Company Biocompatible compounds for pharmaceutical drug delivery systems
ATE238035T1 (en) 1997-09-29 2003-05-15 Inhale Therapeutic Syst PERFORATED MICROPARTICLES AND USE THEREOF
US6565885B1 (en) 1997-09-29 2003-05-20 Inhale Therapeutic Systems, Inc. Methods of spray drying pharmaceutical compositions
US6309623B1 (en) * 1997-09-29 2001-10-30 Inhale Therapeutic Systems, Inc. Stabilized preparations for use in metered dose inhalers
US5904937A (en) 1997-10-03 1999-05-18 Fmc Corporation Taste masked pharmaceutical compositions
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
BR9911098A (en) 1998-06-12 2002-01-29 Microdose Technologies Inc Measurement, packaging and administration of pharmaceutical drugs and medicines
US6197963B1 (en) 1998-08-13 2001-03-06 The Trustees Of The University Of Pennsylvania Non-peptide peptidomimetics
PL349501A1 (en) 1998-12-24 2002-07-29 Janssen Pharmaceutica Nv Controlled release galantamine composition
JP2001151673A (en) 1999-09-06 2001-06-05 Nikken Chem Co Ltd Production of powdery preparation for inhalation
US20020107173A1 (en) 1999-11-04 2002-08-08 Lawrence Friedhoff Method of treating amyloid beta precursor disorders
US20020009491A1 (en) 2000-02-14 2002-01-24 Rothbard Jonathan B. Compositions and methods for enhancing drug delivery across biological membranes and tissues
ATE306933T1 (en) 2000-02-21 2005-11-15 Pharmexa As METHOD FOR DOWN-REGULATION OF AMYLOID
JP2003530437A (en) 2000-04-13 2003-10-14 マヨ ファウンデーション フォー メディカル エデュケーション アンド リサーチ Aβ42 lowering substance
US20080021085A1 (en) 2000-04-13 2008-01-24 Mayo Foundation For Medical Education And Research Method of reducing abeta42 and treating diseases
PE20011227A1 (en) 2000-04-17 2002-01-07 Chiesi Farma Spa PHARMACEUTICAL FORMULATIONS FOR DRY POWDER INHALERS IN THE FORM OF HARD AGGLOMERATES
MY120279A (en) 2000-05-26 2005-09-30 Pharmacia Corp Use of a celecoxib composition for fast pain relief
US20020016359A1 (en) 2000-06-29 2002-02-07 Hellberg Mark R. Compositions and methods of treating neurodegenerative diseases
US8519005B2 (en) 2000-07-27 2013-08-27 Thomas N. Thomas Compositions and methods to prevent toxicity of antiinflammatory agents and enhance their efficacy
FR2815030A1 (en) 2000-10-05 2002-04-12 Lipha NITROSO DIPHENYLAMINE DERIVATIVES, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM AND THEIR USE FOR THE PREPARATION OF MEDICAMENTS
US6511960B2 (en) 2001-01-05 2003-01-28 Alphamed Pharmaceuticals Corp Cromolyn for eye and ear infections
TWI245764B (en) 2001-04-23 2005-12-21 Hank F Kung Amyloid plaque aggregation inhibitors and diagnostic imaging agents
AU2002352974A1 (en) 2001-11-29 2003-06-10 Emisphere Technologies, Inc. Formulations for oral administration of cromolyn sodium
PT1521609E (en) 2002-05-10 2010-03-03 Oriel Therapeutics Inc Dry powder inhaler for use with piezoelectric polymer-driven delivery means, and associated blister package comprising a piezoelectric polymer material
EP1601379A1 (en) 2003-02-13 2005-12-07 Licentia OY Use of a mast cell activation or degranulation blocking agent in the manufacture of a medicament for the treatment of a patient subjected to thrombolyses
EP1601380A1 (en) 2003-02-13 2005-12-07 Licentia OY Use of a mast cell activation or degranulation blocking agent in the manufacture of a medicament for the treatment of cerebral ischemia
US20040223918A1 (en) 2003-05-07 2004-11-11 Chrysalis Technologies Incorporated Aerosolization of cromolyn sodium using a capillary aerosol generator
GB0321607D0 (en) 2003-09-15 2003-10-15 Vectura Ltd Manufacture of pharmaceutical compositions
JP4471340B2 (en) 2003-10-07 2010-06-02 芦森工業株式会社 Buckle device
DE10355559A1 (en) 2003-11-21 2005-06-23 Orthogen Ag Transskin
WO2005063732A1 (en) 2003-12-23 2005-07-14 Microbia, Inc. Compounds and methods for the treatment of asthma
US20070093457A1 (en) 2004-02-11 2007-04-26 Nadir Arber Compositions for treatment of cancer and inflammation with curcumin and at least one nsaid
US20070293538A1 (en) 2004-04-13 2007-12-20 Myriad Genetics, Incorporated Pharmaceutical Composition And Methods For Treating Neurodegenerative Disorders
MXPA06012240A (en) 2004-04-23 2007-01-31 Cydex Inc Dpi formulation containing sulfoalkyl ether cyclodextrin.
KR101414810B1 (en) 2004-08-30 2014-07-18 주식회사 유스팜인터내셔널 Neuroprotective effect of solubilized udca in focal ischemic model
AU2005302452B2 (en) 2004-11-01 2010-12-09 Seo Hong Yoo Methods and compositions for reducing neurodegeneration in Amyotrophic Lateral Sclerosis
JP2008538767A (en) 2005-04-22 2008-11-06 ジェネンテック・インコーポレーテッド Method for treating dementia or Alzheimer's disease with CD20 antibody
WO2006125324A1 (en) 2005-05-27 2006-11-30 Queen's University At Kingston Treatment of protein folding disorders
WO2007070851A2 (en) 2005-12-15 2007-06-21 Acusphere, Inc. Processes for making particle-based pharmaceutical formulations for pulmonary or nasal administration
US8263645B2 (en) * 2006-02-03 2012-09-11 Pari Pharma Gmbh Disodium cromoglycate compositions and methods for administering same
AR059356A1 (en) 2006-02-14 2008-03-26 Astrazeneca Ab NEW RADIOLIGANDS
WO2007102059A1 (en) 2006-03-06 2007-09-13 Pfizer Japan Inc. Sulfonyl benzimidazole derivatives
EA200801967A1 (en) 2006-03-09 2009-04-28 Ворэта Фармэсьютиклз Инк. DOSED DOSAGE FORM ON THE BASIS OF CYCLOGEXANE POLYOL (VARIANTS), MAKING THE TREATMENT, MEANS OF ITS TREATMENT, A METHOD OF TREATING THE DISEASE OF THE ALTSHEIMER WITH ITS HELP AND CYCLOGEXANE POLYOUS AS A QUALITY, ITSELF COURSE
WO2009017467A1 (en) 2007-07-27 2009-02-05 Elan Pharma International Limited Treatment of amyloidogenic diseases
US7700616B2 (en) 2006-05-08 2010-04-20 Molecular Neuroimaging, Llc. Compounds and amyloid probes thereof for therapeutic and imaging uses
US8445437B2 (en) 2006-07-27 2013-05-21 The Brigham And Women's Hospital, Inc. Treatment and prevention of cardiovascular disease using mast cell stabilizers
US20100173960A1 (en) 2006-09-21 2010-07-08 Antonio Cruz The Combination of a Cyclohexanehexol and a NSAID for the Treatment of Neurodegenerative Diseases
US20100292157A1 (en) 2006-11-24 2010-11-18 Antonio Cruz Combination Treatments for Alzheimer's Disease and Similar Diseases
WO2008131298A2 (en) 2007-04-18 2008-10-30 Elan Pharma International Limited Prevention and treatment of cerebral amyloid angiopathy
WO2008128981A1 (en) 2007-04-18 2008-10-30 Probiodrug Ag Nitrovinyl-diamine derivatives as glutaminyl cyclase inhibitors
US7858803B2 (en) 2007-04-27 2010-12-28 The General Hospital Corporation Imaging tracers for early detection and treatment of amyloid plaques caused by Alzheimer's disease and related disorders
CN101848733A (en) 2007-07-13 2010-09-29 艾博特生物技术有限公司 The method and composition that is used for pulmonary administration TNF alpha inhibitor
GB0714134D0 (en) 2007-07-19 2007-08-29 Norton Healthcare Ltd Dry-powder medicament
JO3076B1 (en) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap Immunotherapy regimes dependent on apoe status
EP2268647B1 (en) 2008-03-21 2017-01-25 The General Hospital Corporation Compounds and compositions for the detection and treatment of alzheimer's disease and related disorders
CN102065858B (en) 2008-04-29 2014-07-02 法奈科斯公司 Combination compositions for treating alzheimer disease and related disorders with zonisamide and acamprosate
KR20110117146A (en) 2009-01-22 2011-10-26 라퀄리아 파마 인코포레이티드 N-substituted saturated heterocyclic sulfone compounds with cb2 receptor agonistic activity
US8381454B1 (en) 2009-01-23 2013-02-26 Markus R. Robinson Segmented, elongated, expandable, 4-season, double-walled, low-cost, rigid extruded plastic panel structures
AU2010208136B2 (en) * 2009-01-29 2015-03-05 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment
US9925282B2 (en) 2009-01-29 2018-03-27 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment
EP2772269A3 (en) 2009-03-05 2015-01-14 Abbvie Inc. IL-17 binding proteins
WO2010104973A1 (en) 2009-03-11 2010-09-16 Plexxikon, Inc. Pyrrolo [2, 3-b] pyridine derivatives for the inhibition of raf kinases
JP2012520883A (en) 2009-03-16 2012-09-10 アイピントゥル,エルエルシー Treatment of Alzheimer's disease and osteoporosis and reduction of aging
EP2248517B1 (en) 2009-05-08 2014-03-26 PARI Pharma GmbH Concentrated mast cell stabilizing pharmaceutical formulations
WO2011017030A2 (en) 2009-08-06 2011-02-10 Neuraltus Pharmaceuticals, Inc. Treatment of macrophage-related disorders
US20120175082A1 (en) 2009-09-14 2012-07-12 Kmetovicz Ronald E Solar heat pipe heat exchanger
PT3378856T (en) 2009-09-24 2021-02-02 Wista Lab Ltd Crystalline methylthioninium chloride hydrates
EP2322163A1 (en) 2009-11-03 2011-05-18 Pharnext New therapeutics approaches for treating alzheimer disease
WO2011057199A1 (en) 2009-11-06 2011-05-12 Adenios, Inc. Compositions for treating cns disorders
BR112012012999A2 (en) 2009-11-30 2019-04-02 Aptalis Pharmatech Inc PHARMACEUTICAL COMPOSITION, AND COMPOSITION OF ORAL DISINTEGRATION TABLE (ODT) QUICK DISPERSION OUTABLET (RDT)
US8455755B2 (en) 2009-12-07 2013-06-04 Electrotherm Concentrated photovoltaic and thermal solar energy collector
US20110256064A1 (en) 2010-04-16 2011-10-20 Ac Immune, S.A. Novel Compounds for the Treatment of Diseases Associated with Amyloid or Amyloid-like Proteins
WO2011136754A1 (en) 2010-04-26 2011-11-03 Mahmut Bilgic A medicament developed for the treatment of respiratory diseases
WO2012041717A1 (en) 2010-09-30 2012-04-05 Chiesi Farmaceutici S.P.A. Use of magnesium stearate in dry powder formulations for inhalation
US20120121656A1 (en) 2010-11-15 2012-05-17 Revalesio Corporation Methods and compositions for protecting against neurotoxicity of a neurotoxic agent, and improving motor coordination associated with a neurodegenerative condition or disease
SI2673266T1 (en) 2011-02-11 2016-11-30 Wista Laboratories Ltd. Phenothiazine diaminium salts and their use
RU2607892C2 (en) 2012-05-25 2017-01-11 Экслиар, Инк. Composition based on xylitol, preventing separation of slime and related methods and compositions
WO2014065275A1 (en) 2012-10-23 2014-05-01 国立大学法人福井大学 Therapeutic or prophylactic agent for tumor lysis syndrome
US10058530B2 (en) 2012-10-25 2018-08-28 The General Hospital Corporation Combination therapies for the treatment of Alzheimer's disease and related disorders
WO2014066318A1 (en) 2012-10-25 2014-05-01 The General Hospital Corporation Combination therapies for the treatment of alzheimer's disease and related disorders
US10525005B2 (en) 2013-05-23 2020-01-07 The General Hospital Corporation Cromolyn compositions and methods thereof
CA2913235C (en) 2013-05-23 2021-07-13 Aztherapies, Inc. Methods for delivering cromolyn
JP2016534063A (en) 2013-10-22 2016-11-04 ザ ジェネラル ホスピタル コーポレイション Cromolyn derivatives and related imaging and treatment methods
CN106456595A (en) 2014-02-10 2017-02-22 帕塔拉制药有限责任公司 Mast cell stabilizers treatment for systemic disorders
CN106535889A (en) 2014-02-10 2017-03-22 帕塔拉制药有限责任公司 Mast cell stabilizers for lung disease treatment
WO2016196401A1 (en) 2015-05-29 2016-12-08 The Texas A&M University System Antimicrobial and anti-inflammatory compositions
US20180311236A1 (en) 2015-10-28 2018-11-01 Ab Science Use of masitinib and other mast cell inhibitors for treatment of parkinson's disease
KR20180081807A (en) 2015-11-19 2018-07-17 아즈테라피즈 인코포레이티드 Methods of treating Alzheimer's disease and related disorders
EP3380095B1 (en) 2015-11-23 2022-01-05 The General Hospital Corporation Compositions and methods for treating ischemic stroke
EA038531B1 (en) 2016-03-25 2021-09-10 Аб Сьянс Method for treatment of an amyotrophic lateral sclerosis, use of a pharmaceutical composition comprising masitinib
EP3506894B1 (en) 2016-08-31 2023-08-23 The General Hospital Corporation Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases
EP3654946A4 (en) 2017-07-20 2021-04-21 AZTherapies, Inc. Powdered formulations of cromolyn sodium and ibuprofen
CN112912072A (en) 2018-04-09 2021-06-04 通用医疗公司 Combination therapy for treating amyotrophic lateral sclerosis and related diseases
EP3817739A4 (en) 2018-07-02 2022-04-13 The General Hospital Corporation Powdered formulations of cromolyn sodium and ?-lactose
CN113038945A (en) 2018-09-05 2021-06-25 通用医疗公司 Methods of treating cytokine release syndrome
AU2019397436A1 (en) 2018-12-10 2021-07-22 The General Hospital Corporation Cromolyn esters and uses thereof

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11801316B2 (en) 2009-01-29 2023-10-31 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment
US11110097B2 (en) 2012-10-25 2021-09-07 The General Hospital Corporation Combination therapies for the treatment of alzheimer's disease and related disorders
US11013686B2 (en) 2013-05-23 2021-05-25 The General Hospital Corporation Cromolyn compositions and methods thereof
US11666669B2 (en) 2013-10-22 2023-06-06 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment
US11679095B2 (en) 2016-08-31 2023-06-20 The General Hospital Corporation Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases
US11291648B2 (en) 2018-07-02 2022-04-05 The General Hospital Corporation Powdered formulations of cromolyn sodium and alpha-lactose

Also Published As

Publication number Publication date
US20230248646A1 (en) 2023-08-10
US20210085601A1 (en) 2021-03-25
US11013686B2 (en) 2021-05-25
US20180153803A1 (en) 2018-06-07
US10525005B2 (en) 2020-01-07

Similar Documents

Publication Publication Date Title
AU2019271898B2 (en) Methods for delivering cromolyn
US11013686B2 (en) Cromolyn compositions and methods thereof
RU2648849C2 (en) Device for drug delivery for treatment of patients with diseases of breathing organs
CN103501781B (en) Glycopyrrolate is used for treating tachycardic purposes
EP3148521B1 (en) Fluticasone furoate in the treatment of copd
Wallin et al. Pulmonary drug delivery to older people
PT2398473E (en) 5-(2-{[6-(2, 2-difluoro-2-phenylethoxy)hexyl]amino}-1-hydroxyethyl)-8-hydroxyquinolin-2-(1h)-one for the treatment of lung function
Tang et al. Method to introduce mannitol powder to intubated patients to improve sputum clearance
TWI436761B (en) Methods of using a thiazole derivative
AU2017442079A1 (en) Cromolyn compositions and methods thereof
JP2016027032A (en) Therapeutic agent for chronic obstructive pulmonary disease
NZ614603B2 (en) Use of glycopyrrolate for treating tachycardia

Legal Events

Date Code Title Description
AS Assignment

Owner name: ELMALEH, DAVID R., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:AZ THERAPIES, INC.;REEL/FRAME:052039/0661

Effective date: 20181206

Owner name: AZ THERAPIES, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ELMALEH, DAVID R.;REEL/FRAME:052039/0627

Effective date: 20180226

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ELMALEH, DAVID R.;REEL/FRAME:052039/0741

Effective date: 20181206

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

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STCB Information on status: application discontinuation

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