Cromolyn Compositions for Treatment of Chronic Cough Due to Idiopathic Pulmonary Fibrosis

- Respivant Sciences GmbH

The disclosure provides a method of treating chronic cough in a subject having idiopathic pulmonary fibrosis (IPF) comprising administering to the subject a pharmaceutical composition comprising from about 2% by weight to about 6% by weight of cromolyn sodium and an ionic osmotic agent with a nebulizer.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a Continuation Application of U.S. application Ser. No. 15/887,830 filed on Feb. 2, 2018, which is a Continuation Application of PCT/US17/49205, filed on Aug. 29, 2017 which claims the priority benefit of U.S. provisional application No. 62/381,914, filed on Aug. 31, 2016, and 62/417,898, filed on Nov. 4, 2016, each of which is incorporated by reference herein in their entirety.

FIELD OF THE DISCLOSURE

The disclosure is directed to the field of medicine and, in, particular the use of compositions comprising cromolyn for the treatment of chronic cough due to idiopathic pulmonary fibrosis.

BACKGROUND

There has been a long-felt but unmet need in the art for an effective treatment of chronic cough due to idiopathic pulmonary fibrosis (IPF), as the majority of individuals with condition are unresponsive to currently available treatment. The disclosure provides a solution to this long-felt but unmet need.

SUMMARY

The disclosure provides compositions and methods for the treatment of chronic cough due to IPF. Subjects treated with the compositions and formulations provided in the disclosure demonstrate a statistically significant and medically relevant improvement in objective cough rates after 14 days of treatment compared to those subjects who received a placebo. These subjects had refractory chronic cough associated with IPF and are unable to find relief with the currently-available treatments (not including the compositions or methods disclosed herein).

Specifically, the disclosure provides a method of treating chronic cough in a subject having idiopathic pulmonary fibrosis (IPF) comprising administering to the subject a pharmaceutical composition comprising from about 1% by weight to about 10% by weight of cromolyn sodium and an ionic osmotic agent with a nebulizer. In certain embodiments, the pharmaceutical composition comprises about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10% by weight of cromolyn sodium. In certain embodiments, the pharmaceutical composition comprises about 2% by weight of cromolyn sodium. In certain embodiments, the pharmaceutical composition comprises about 4% by weight of cromolyn sodium. In certain embodiments, the pharmaceutical composition comprises about 6% by weight of cromolyn sodium.

The disclosure provies a pharmaceutically acceptable aerosol for the treatment of chronic cough in a subject having idiopathic pulmonary fibrosis (IPF), comprising droplets of an aqueous solution comprising (i) from about 2% to about 6% by weight of cromolyn sodium and (ii) an osmolarity adjusting agent comprising (a) between about 0.1% and about 0.5% by weight of sodium chloride, inclusive of the endpoints, and (b) optionally salts of ethylenediaminetetraacetic acid (EDTA), wherein said aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30%, and wherein said treatment of said chronic cough in said subject is achieved via delivery of a therapeutically effective amount of cromolyn sodium to the lungs of the subject by said subject orally inhaling said pharmaceutically acceptable aerosol.

The disclosure provides a pharmaceutically acceptable aerosol for the treatment of chronic cough in a subject having idiopathic pulmonary fibrosis (IPF), consisting of droplets of an aqueous solution consisting of (i) from about 2% to about 6% by weight of cromolyn sodium and (ii) an osmolarity adjusting agent consisting of (a) between about 0.1% and about 0.5% by weight of sodium chloride, inclusive of the endpoints, and (b) optionally salts of ethylenediaminetetraacetic acid (EDTA), wherein said aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30%, and wherein said treatment of said chronic cough in said subject is achieved via delivery of a therapeutically effective amount of cromolyn sodium to the lungs of the subject by said subject orally inhaling said pharmaceutically acceptable aerosol.

The disclosure provides a pharmaceutically acceptable solution, comprising from about 1% to about 10% by weight of cromolyn sodium and an osmotic agent, wherein an aerosol created from the pharmaceutically acceptable solution is suitable for inhalation by a subject having chronic cough due to idiopathic pulmonary fibrosis. In certain embodiments, the aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30%. In certain embodiments, the aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30% and a respirable fraction (≤5 μm) as measured by USP <1601> of at least about 75%.

The disclosure provides a dosage form for the treatment of chronic cough in a subject having idiopathic pulmonary fibrosis (IPF) comprising:

    • (a) an aqueous pharmaceutical composition comprising (i) from about 2% to about 6% by weight of cromolyn sodium, and (ii) an osmolarity adjusting agent consisting of (A) between about 0.1% to about 0.5% sodium chloride, inclusive of the endpoints; and (B) optionally salts of EDTA; and (b) an inhalation device that forms an aerosol of said pharmaceutical composition, said aerosol exhibiting a respirable fraction of said pharmaceutical composition (≤5 μm) as measured by USP <1601> of at least about 60%. In certain embodiments, the pharmaceutical composition further comprises purified water and sodium EDTA. In certain embodiments, the pharmaceutical composition comprises from about 5 mg to about 80 mg of cromolyn sodium. In certain embodiments, the pharmaceutical composition comprises from about 36 mg to about 44 mg of cromolyn sodium. In certain embodiments, the aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30%. In certain embodiments, the aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30% and a respirable fraction (≤5 μm) as measured by USP <1601> of at least about 75%. In certain embodiments, wherein the osmolarity adjusting agent consists of between 0.1% to 0.2% sodium chloride, inclusive of the endpoints.

The terms pharmaceutical composition, composition, solution, and formulation are used interchangeably throughout the disclosure.

In certain embodiments of the compositions and formulations of the disclosure, the ionic osmotic agent may comprise or consist of an ionic osmotic agent. In certain embodiments of the compositions and formulations of the disclosure, the ionic osmotic agent may comprise or consist of sodium chloride. The ionic osmotic agent of the compositions of the disclosure may comprise between 0.1% and 0.7%, by weight, of the composition, inclusive of the endpoints. In certain embodiments, the compositions of the disclosure may comprise between 0.1% and 0.2%, by weight of the composition, of the ionic osmotic agent inclusive of the endpoints. In certain embodiments, the compositions of the disclosure may comprise 0.2%, by weight, of the composition, of the ionic osmotic agent.

Compositions and formulations of the disclosure may further comprise a chelating agent. In certain embodiments, the chelating agent may comprise about 0.01%, or about 0.02%, or about 0.03%, or about 0.04%, or about 0.05%, or about 0.06%, or about 0.07%, or about 0.08%, or about 0.09%, or about 0.1%, or about 0.2%, or about 0.3%, or about 0.4%, or about 0.5%, or about 0.6%, or about 0.7%, or about 0.8% or about 0.9%, or about 1% by weight, of the composition. In certain embodiments, the chelating agent comprises ethylenediaminetetraacetic acid (EDTA), sodium-EDTA, or sodium citrate. In certain embodiments, the chelating agent comprises EDTA. In certain embodiments, the chelating agent comprises sodium-EDTA. In certain embodiments, the chelating agent comprises sodium citrate.

Compositions and formulations of the disclosure may further comprise a non-ionic osmotic agent, preferably, wherein the non-ionic osmotic agent comprises or consists of mannitol.

Compositions and formulations of the disclosure may exclude, or, may not comprise a non-ionic osmotic agent. In certain embodiments, compositions of the disclosure may not comprise a non-ionic osmotic agent comprising or consisting of mannitol, a sugar alcohol and/or propylene glycol. In certain embodiments, compositions of the disclosure do not comprise propylene glycol, regardless of any potential functional role of the propylene glycol.

Compositions and formulations of the disclosure may exclude, or, may not comprise a non-ionic osmotic agent. In certain embodiments, compositions of the disclosure may not comprise a non-ionic osmotic agent comprising or consisting of mannitol, any other sugar alcohol and/or propylene glycol. In certain embodiments, compositions of the disclosure do not comprise mannitol, any other sugar alcohol and/or propylene glycol, regardless of any potential functional role, chemical property or use of mannitol, any other sugar alcohol and/or propylene glycol known to those of ordinary skill in the art.

Compositions and formulations of the disclosure may have a surface tension effective for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments, the compositions and formulations of the disclosure may have a surface tension in the range similar to that or water or higher. In certain embodiments, the compositions and formulations according to the present disclosure has a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m, such as in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

Compositions and formulations of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, compositions of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkoniumrn chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present compositions and formulations include any compound or agent that lowers the surface tension of a composition.

Compositions and formulations of the disclosure may further comprise purified water for injection. The amount of the water may vary depending upon, for example, a fill volume required for the particular high-efficiency nebulizer used. In certain embodiments, compositions of the disclosure comprise purified water for injection in a quantum sufficiat (q.s.).

Compositions and formulations of the disclosure may have an osmolality of between about 100 mOsm/kg and about 200 mOsm/kg, inclusive of the endpoints. Compositions and formulations of the disclosure may have an osmolality of between about 125 mOsm/kg and about 135 mOsm/kg, inclusive of the endpoints. Compositions and formulations of the disclosure may have an osmolality of between about 100 mOsm/kg and about 125 mOsm/kg, inclusive of the endpoints. Compositions and formulations of the disclosure may have an osmolality of between about 100 mOsm/kg and about 135 mOsm/kg, inclusive of the endpoints. Compositions and formulations of the disclosure may have an osmolality of about 100 mOsm/kg. Compositions and formulations of the disclosure may have an osmolality of about 125 mOsm/kg. Compositions and formulations of the disclosure may have an osmolality of about 135 mOsm/kg. Compositions and formulations of the disclosure may have an osmolality of about 200 mOsm/kg.

Compositions and formulations of the disclosure may be in the form of a solution having a fill volume of about 0.1 mL to about 5 mL. Compositions and formulations of the disclosure may be in the form of an aqeuous solution having a fill volume of about 0.1 mL, about 0.5 mL, about 1 mL, about 1.5 mL, about 2 mL, about 2.5 mL, about 3 mL, about 3.5 mL, about 4 mL, about 4.5 mL, or about 5 mL.

Compositions and formulations of the disclosure may comprise from about 5 mg to about 80 mg of cromolyn sodium, inclusive of the endpoints. Compositions and formulations of the disclosure may comprise from about 36 mg to about 44 mg of cromolyn sodium, inclusive of the endpoints.

Nebulizers administering a composition or formulation of the disclosure may provide an aerosol having a respirable fraction ≤3.3 μm as measured by USP <1601> of at least about 30% and a respirable fraction ≤5 μm as measured by USP <1601> of at least about 65%. Nebulizers administering a composition or formulation of the disclosure may provide an aerosol having a respirable fraction ≤3.3 μm as measured by USP <1601> of at least about 45% and a respirable fraction ≤5 μm as measured by USP <1601> of at least about 75%.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure with a nebulizer may result in primarily lung deposition, and minimal deposition in other respiratory tracts, of the administered aerosol. In certain embodiments, sedimentation is the major mechanism of deposition of the aerosol. In certain embodiments, administration of pharmaceutical compositions of the disclosure with a nebulizer provides a lung deposition (deposited lung dose) of at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, about 20% to about 40%, about 25% to about 35%, about 25% to about 30%, about 25% to about 75%, about 30% to about 50%, about 35% to about 90%, about 40% to about 80%, about 40% to about 60%, about 50% to about 60%, about 50% to about 70%, or about 60% to about 75% based on the nominal dose of the composition.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure with a nebulizer may produce in the subject an AUC(0-∞) of the cromolyn sodium greater than about 150 ng*hr/mL, a Cmax of the cromolyn sodium greater than about 50 ng/mL, and a deposited lung dose of cromolyn sodium greater than about 4 mg. According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure with a nebulizer may produce in the subject an AUC(0-∞) of the cromolyn sodium greater than about 175 ng*hr/mL, a Cmax of the cromolyn sodium greater than about 60 ng/mL, and a deposited lung dose of the cromolyn sodium greater than about 4 mg. According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure with a nebulizer may produce in the subject an AUC(0-∞) of the cromolyn sodium greater than about 100 ng*hr/mL, a Cmax of the cromolyn sodium greater than about 40 ng/mL, and a deposited lung dose of the cromolyn sodium greater than about 4 mg.

Lung Deposited Percentage

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a nebulizer may produce in a subject an AUC(0-∞) of the cromolyn sodium that is between about 120 ng*hr/mL and about 350 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a nebulizer may produce in a subject an AUC(0-∞) of the cromolyn sodium that is within 80% to 125% of about 340 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a nebulizer may produce in a subject an AUC(0-6) of the cromolyn sodium that is between about 120 ng*hr/mL and about 350 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a nebulizer may produce in a subject an AUC(0-6) of the cromolyn sodium that is within 80% to 125% of about 237 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a nebulizer may produce in a subject a Cmax of the cromolyn sodium of between about 40 ng/mL and about 150 ng/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a nebulizer may produce in a subject a Cmax of the cromolyn sodium that is within 80% to 125% of about 85 ng/mL, or about 75 ng/mL, or about 82 ng/mL, or about 93 ng/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a nebulizer may produce in a subject an AUC(0-∞) of the cromolyn sodium that is between about 250 ng*hr/mL and about 1000 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a nebulizer may produce in a subject an AUC(0-∞) of the cromolyn sodium that is within 80% to 125% of about 542 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a nebulizer may produce in a subject an AUC(0-6) of the cromolyn sodium that is between about 200 ng*hr/mL and about 700 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a nebulizer may produce in a subject an AUC(0-6) of the cromolyn sodium that is within 80% to 125% of about 389 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a nebulizer may produce in a subject a Cmax of the cromolyn sodium of between about 50 ng/mL and about 250 ng/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a nebulizer may produce in a subject Cmax of the cromolyn sodium that is within 80% to 125% of about 134 ng/mL, or about 119 ng/mL, or about 148 ng/mL, or about 157 ng/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 80 mg of cromolyn sodium with a nebulizer may produce in a subject an AUC(0-∞) of the cromolyn sodium that is between about 300 ng*hr/mL and about 800 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 80 mg of cromolyn sodium with a nebulizer may produce in a subject an AUC(0-∞) of the cromolyn sodium that is within 80% to 125% of about 526 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 80 mg of cromolyn sodium with a nebulizer may produce in a subject a Cmax of the cromolyn sodium of between about 90 ng/mL and about 450 ng/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure with a dry powder inhaler may produce in the subject an AUC(0-∞) of the cromolyn sodium greater than about 150 ng*hr/mL, a Cmax of the cromolyn sodium greater than about 50 ng/mL, and a deposited lung dose of cromolyn sodium greater than about 4 mg.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure with a nebulizer may produce in the subject an AUC(0-∞) of the cromolyn sodium greater than about 175 ng*hr/mL, a Cmax of the cromolyn sodium greater than about 60 ng/mL, and a deposited lung dose of the cromolyn sodium greater than about 4 mg.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure with a nebulizer may produce in the subject an AUC(0-∞) of the cromolyn sodium greater than about 100 ng*hr/mL, a Cmax of the cromolyn sodium greater than about 40 ng/mL, and a deposited lung dose of the cromolyn sodium greater than about 4 mg.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a dry powder inhaler may produce in a subject an AUC(0-∞) of the cromolyn sodium that is between about 120 ng*hr/mL and about 350 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a dry powder inhaler may produce in a subject an AUC(0-∞) of the cromolyn sodium that is within 80% to 125% of about 340 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a dry powder inhaler may produce in a subject an AUC(0-6) of the cromolyn sodium that is between about 120 ng*hr/mL and about 350 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a dry powder inhaler may produce in a subject an AUC(0-6) of the cromolyn sodium that is within 80% to 125% of about 237 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a dry powder inhaler may produce in a subject a Cma of the cromolyn sodium of between about 40 ng/mL and about 150 ng/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 40 mg of cromolyn sodium with a dry powder inhaler may produce in a subject a Cmax of the cromolyn sodium that is within 80% to 125% of about 85 ng/mL, or about 75 ng/mL, or about 82 ng/mL, or about 93 ng/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a dry powder inhaler may produce in a subject an AUC(0-∞) of the cromolyn sodium that is between about 250 ng*hr/mL and about 1000 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a dry powder inhaler may produce in a subject an AUC(0-∞) of the cromolyn sodium that is within 80% to 125% of about 542 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a dry powder inhaler may produce in a subject an AUC(0-6) of the cromolyn sodium that is between about 200 ng*hr/mL and about 700 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a dry powder inhaler may produce in a subject an AUC(0-6) of the cromolyn sodium that is within 80% to 125% of about 389 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a dry powder inhaler may produce in a subject a Cma of the cromolyn sodium of between about 50 ng/mL and about 250 ng/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 60 mg of cromolyn sodium with a dry powder inhaler may produce in a subject Cmax of the cromolyn sodium that is within 80% to 125% of about 134 ng/mL, or about 119 ng/mL, or about 148 ng/mL, or about 157 ng/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 80 mg of cromolyn sodium with a dry powder inhaler may produce in a subject an AUC(0-∞) of the cromolyn sodium that is between about 300 ng*hr/mL and about 800 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 80 mg of cromolyn sodium with a dry powder inhaler may produce in a subject an AUC(0-∞) of the cromolyn sodium that is within 80% to 125% of about 526 ng*hr/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 80 mg of cromolyn sodium with a dry powder inhaler may produce in a subject a Cmax of the cromolyn sodium of between about 90 ng/mL and about 450 ng/mL.

According to the methods of the disclosure, administration of pharmaceutical compositions of the disclosure comprising about 80 mg of cromolyn sodium with a dry powder inhaler may produce in a subject a Cmax of the cromolyn sodium that is within 80% to 125% of about 236 ng/mL.

According to the methods of the disclosure, in certain embodiments, the pharmaceutical composition is administered daily. In certain embodiments, the pharmaceutical composition is administered once per day. In certain embodiments, the pharmaceutical composition is administered twice per day. In certain embodiments, the pharmaceutical composition is administered three times per day. In certain embodiments, the pharmaceutical composition is administered four times per day. In certain embodiments, the pharmaceutical composition is administered five times per day.

According to the methods of the disclosure, in certain embodiments, the pharmaceutical composition is administered three times per day for at least 7 days. In certain embodiments, the pharmaceutical composition is administered three times per day for at least 14 days. In certain embodiments, the pharmaceutical composition is administered three times per day as a daily maintenance therapy (e.g. at least 7 or at least 14 days without a limit for the total length of treatment).

According to the methods of the disclosure, in certain embodiments, the pharmaceutical composition may be administered as a combination therapy with any other therapeutic composition for the treatment of chronic cough associated or due to a diagnosis of IPF in a subject.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 is a series of graphs showing the average number of daytime coughs for each subject in Example 1 following treatment with either PA101 or with one of two placebo treatments.

FIG. 2 is a series of graphs showing the total number of daytime coughs for each subject in Example 1 following treatment with either PA101 or with one of two placebo treatments.

FIG. 3 is a graph depicting pulmonary function (measured as Forced Expiratory Volume in One Second (FEV1)) as a function of time for each of the treatment groups described in Example 1.

FIG. 4 is a schematic diagram of the design of the Phase II study described in Example 4.

FIG. 5 is a graph showing the average number of daytime coughs (coughs/hour) as a function of time in both placebo-treated (n=21) and PA101-treated subjects (n=23) at baseline, Day 7, and Day 14 of the study described in Example 4. The average number of daytime coughs per hour is a primary efficacy endpoint of this phase II study.

FIG. 6 is a graph showing the mean change from baseline of the average number of daytime coughs per hour as a function of time in both placebo-treated and PA101-treated subjects at baseline, Day 7, and Day 14 of the study described in Example 4. At Day 7 and Day 14, the mean change from baseline in the PA101-treated group (the closed circles) was statistically significant (p=0.03). At Day 14, the mean change from baseline in the PA101-treated group (the closed circles) was statistically significant (p=0.04).

FIG. 7 is a pair of graphs demonstrating a placebo-adjusted reduction in cough (left-hand graph), expressed as a percent change from baseline in the daytime average cough/hour at Day 14 for placebo versus PA101-treated subjects of the study described in Example 4 and a PA101/placebo LS mean ratio (95% CI) for this reduction in cough (right-hand graph), expressed as a change in the log transformed daytime average cough/hour at Day 14 for placebo versus PA101-treated subjects of the study described in Example 4. At Day 14, the change in the log transformed daytime average cough/hour in the PA101-treated group was statistically significant (p=0.02).

FIG. 8 is a graph showing the PA101/placebo LS mean ratio (95% CI) for the reduction in daytime cough in PA101-treated subjects at Days 7 and 14 (see Example 4). At Day 7, the change in the log transformed daytime average cough/hour in the PA101-treated group was statistically significant (p=0.03). At Day 14, the change in the log transformed daytime average cough/hour in the PA101-treated group was statistically significant (p=0.02).

FIG. 9 is a pair of graphs depicting the average daytime coughs per hour for each subject in each of the placebo and PA101 treated groups at either baseline or day 14 of the study described in Example 4.

FIG. 10 is a graph providing the results of a responder analysis for daytime cough, expressed as the percentage (%) of subjects in the study described in Example 4 who experienced a percentage (%) reduction in daytime average coughs/hour compared to baseline. Greater than 30% responders: n=8 with PA101 (3 subjects concomitantly taking one or more other IPF treatments vs. 5 subjects without conconmitant administration of one or more other IPF treatments) and n=4 with placebo (1 subject concomitantly taking one or more other IPF treatments vs. 3 subjects without concomitant administration of one or more other IPF treatments). By “conconmitantly taking” or “conconmitant administration” is meant that a particular subject is being administered one or more other IPF treatments, such as pirfenidone or nintedanib esylate while being administered at least one formulation disclosed herein that comprises cromolyn sodium.

FIG. 11 is a graph providing the results of a responder analysis for 24-hour cough, expressed as the percentage (%) of subjects in the study described in Example 4 who experienced a percentage (%) reduction in 24-hour average coughs/hour compared to baseline. Greater than 30% responders: n=8 with PA101 (3 subjects concomitantly taking one or more other IPF treatments vs. 5 subjects without conconmitant administration of one or more other IPF treatments) and n=4 with placebo (1 subject concomitantly taking one or more other IPF treatments vs. 3 subjects without conconmitant administration of one or more other IPF treatments). By “conconmitantly taking” or “conconmitant administration” is meant that a particular subject is being administered one or more other IPF treatments, such as pirfenidone or nintedanib esylate while being administered at least one formulation disclosed herein that comprises cromolyn sodium.

FIG. 12 is a graph showing the nighttime average coughs per hour in (coughs/hour) as a function of time in both placebo-treated and PA101-treated subjects at baseline, Day 7, and Day 14 of the study described in Example 4. The average number of nighttime coughs per hour is a secondary efficacy endpoint of this phase II study.

FIG. 13 is a graph showing the mean change from baseline of the average number of nighttime coughs per hour as a function of time in both placebo-treated and PA101-treated subjects at Day 14 of the study described in Example 4.

FIG. 14 is a graph showing the average number of coughs (coughs/hour) in a 24-hour period as a function of time in both placebo-treated and PA101-treated subjects at baseline, Day 7, and Day 14 of the study described in Example 4. The average number of coughs per hour in a 24-hour period is a secondary efficacy endpoint of this phase II study.

FIG. 15 is a graph demonstrating placebo-adjusted reduction in coughs during a 24-hour period, expressed as a percent change from baseline in the average coughs/hour during a 24-hour period at Days 7 and 14 for placebo versus PA101-treated subjects of the study described in Example 4.

FIG. 16 is a graph showing the PA101/placebo LS mean ratio (95% CI) for the placebo-adjusted reduction in coughs, expressed as a change in the log transformed average coughs/hour during a 24-hour period at Days 7 and 14 for placebo versus PA101-treated subjects of the study described in Example 4. At Day 7, the change in the log transformed average coughs/hour during a 24-hour period in the PA101-treated group was statistically significant (p=0.016). At Day 14, the change in the log transformed average coughs/hour during a 24-hour period in the PA101-treated group was statistically significant (p=0.025).

FIG. 17 is a pair of graphs demonstrating a placebo-adjusted reduction in cough (left-hand graph), expressed as a percent change from baseline in the 24-hour average cough/hour at Day 14 for placebo versus PA101-treated subjects of the study described in Example 4 and a PA101/placebo LS mean ratio (95% CI) for this reduction in cough (right-hand graph), expressed as a change in the log transformed 24-hour average cough/hour at Day 14 for placebo versus PA101-treated subjects of the study described in Example 4. At Day 14, the change in the log transformed 24-hour average cough/hour in the PA101-treated group was statistically significant (p=0.025).

FIG. 18 is a pair of graphs depicting a 24-hour cough profile for a subject with chronic cough due to IPF receiving either placebo or PAl01 as part of the Phase II study described in Example 4.

FIG. 19 is a pair of graphs showing the results of the Responder Analysis from Example 4. LCQ and VAS scores were compared in Responders from this study at baseline and day 14 of treatment with PA101.

FIG. 20 is a schematic diagram of the design of the study described in Example 5.

FIG. 21 is a pair of graphs depicting the average daytime coughs per hour for each subject in each of the placebo and PA101 treated groups at either baseline or day 14 of the study described in Example 5.

FIG. 22 is a graph showing the average number of 24-hour coughs (coughs/hour) as a function of time in both placebo-treated and PA101-treated subjects at baseline, Day 7, and Day 14 of the study described in Example 5. The average number of 24-hour coughs per hour is a secondary efficacy endpoint of this phase II study.

FIG. 23 is a graph showing the PA101/placebo LS mean ratio (95% CI) for the reduction in daytime cough in PA101-treated subjects at Days 7 and 14 (see Example 5). At Day 7, the change in the log transformed daytime average cough/hour in the PA101-treated group was statistically significant (p=0.038). At Day 14, the change in the log transformed daytime average cough/hour in the PA101-treated group was not statistically significant (p=0.33).

FIG. 24 is a graph providing the results of a responder analysis for daytime cough, expressed as the percentage (%) of subjects in the study described in Example 5 who experienced a percentage (%) reduction in daytime average coughs/hour compared to baseline.

FIG. 25 is a graph showing the PA101/placebo LS mean ratio (95% CI) for the reduction in cough during a 24-hour period in PA101-treated subjects at Days 7 and 14 (see Example 5).

FIG. 26 is a pair of graphs showing a Responder Analysis for daytime average cough/hour for subjects with chronic cough due to IPF (Example 4) versus subjects with chronic idiopathic cough (CIC) (Example 5).

FIG. 27 is a pair of graphs showing a Responder Analysis for 24-hour average cough/hour for subjects with chronic cough due to IPF (Example 4) versus subjects with chronic idiopathic cough (CIC) (Example 5).

FIG. 28 is a graph showing the log transformed daytime average cough/hour subjects with chronic cough due to IPF (Example 4) versus subjects with chronic idiopathic cough (CIC) (Example 5).

FIG. 29 is a graph showing the log transformed 24-hour average cough/hour subjects with chronic cough due to IPF (Example 4) versus subjects with chronic idiopathic cough (CIC) (Example 5).

DETAILED DESCRIPTION

Compositions of the disclosure comprising cromolyn sodium and an ionic osmotic agent are safe and efficacious for the treatment of chronic cough due to IPF. Subjects treated with the compositions and formulations provided in the disclosure demonstrate a statistically significant and medically relevant improvement in objective cough rates after 14 days of treatment compared to those subjects who received a placebo. These subjects had refractory chronic cough associated with IPF, each of them unable to find relief with the currently-available treatments (not including the formulations and methods of the present disclosure).

Chronic Cough

In the United States, cough is the most common complaint for which subjects seek medical attention and is the second most common reason for a general medical examination, accounting for more than 26 million office visits annually (National Center for Health Statistics. National Ambulatory Medical Care Survey: 2008). Upper respiratory tract infection (i.e. the common cold) is by far the most common cause of cough, but post-infectious cough, unexplained chronic cough, and cough due to pulmonary disorders, such as asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and lung cancer are also common.

Cough serves a potentially beneficial purpose by clearing the airways of excessive mucus, irritants, or abnormal substances such as edema fluid or pus. But while productive cough may serve a useful function, coughing can also lead to a variety of problems, including exhaustion (57%), feeling self, conscious (55%), insomnia (45%), changes in lifestyle (45%), musculoskeletal pain (45%), hoarseness (43%), excessive perspiration (42%), and urinary incontinence (39%). These problems are more likely to be prominent in the setting of chronic versus acute cough. As a consequence, chronic cough is responsible for up to 38% of pulmonary outsubject visits. There is a significant economic cost for the individual with cough and for society, for example absence from work and lost productivity.

Cough results from activation of myelinated cough receptors and unmyelinated C fibers whose cell bodies are in the jugular and nodose ganglia. Extensive C fiber endings are found under the airway epithelium while cough receptors endings terminate in the mucosa between the epithelium and smooth muscle. Mast cells play an important role in cough, as degranulated mast cells release mediators which can activate C fibers, causing release of Substance P, histamine, serotonin, and proteases. Release of Substance P results in inflammation, vasodilatation, sensitization of nerves, and a hyperactive cough reflex.

Idiopathic Pulmonary Fibrosis (IPF)

IPF is a progressive life threatening disease that is characterized anatomically by scarring of the lungs and symptomatically by exertional dyspnea. Its etiology is unknown, and the pathogenic mechanisms involved in its initiation and progression are poorly understood. IPF is characterized by the accumulation of lung fibroblasts and extracellular matrix deposition, ultimately leading to compromised tissue architecture and lung function capacity. Clinical features of IPF include dry cough, breathlessness, restrictive respiratory physiology, end inspiratory crackles, reduced oxygenation, and finger clubbing High-resolution computer tomography (HRCT) shows a distinctive pattern of subpleural shadowing and honeycomb fibrosis later in the disease.

Chronic Cough in IPF (Also Referred to as Refractory Chronic Cough in IPF)

IPF is a rare lung disorder of unknown etiology impacting approximately 135,000 people in the United States. One common characteristic of IPF appears to be aberrant wound healing that leads to fibrosis. As of the date of this disclosure, no curative therapy exists. The median survival of a subject with IPF is between three and five years.

As used herein, chronic cough associated with IPF may be defined as a dry non-productive cough (up to 100 coughs per hour) that persists for more than eight weeks despite treatment with standard care. Chronic cough associated with IPF significantly impacts quality of life resulting in, for example, one or more of headache, dizziness, sweating, incontinence, fractured ribs, laryngeal trauma, vomiting, exacerbation of respiratory diseases, loss of sleep, exhaustion, and anxiety. Moreover, chronic cough associated with IPF significantly impacts psychosocial factors, including, but not limited to, job loss, reluctance to attend public events, and depression.

Chronic cough associated with IPF may be an independent predictor of IPF disease progression. Approximately 80% of IPF subjects have a chronic cough. Of those 80%, approximately 40% of those subjects have a form of chronic cough that is refractory to available treatments (available treatments do not include the formulations, compositions or methods of the disclosure).

Available treatments options for cough have significant limitations. Over-the-counter drugs provide negligible efficacy. Prescription drugs are opioid-based and only occasionally effective, but frequently associated with constipation, drowsiness, respiratory depression and risk of addiction. No new non-opioid products have been approved for cough in the last 50 years.

Individuals with chronic cough due to IPF may have frequent coughing during the day with relatively little nocturnal cough. Cough rates may be wide ranging and do not appear to be influenced by the subject's age or gender.

Cromolyn, and Analogs, Derivatives, and Pharmaceutically Acceptable Salts Thereof

As used herein, cromolyn refers to disodium 5,5′-(2-hydroxypropane-1,3-diyl)bis(oxy)bis(4-oxo-4H-chromene-2-carboxylate) and has the following structure:

Cromolyn is also known as sodium cromolyn, cromoglicic acid, disodium cromoglicate (DSCG), sodium cromoglicate, and cromoglicate. Pharmaceutically acceptable salts of cromolyn include but are not limited to cromolyn sodium, cromolyn lysinate, ammonium cromonglycate, and magnesium cromoglycate. Cromolyn sodium is also known as disodium 5,5′-[(2-hydroxytrimethylene)dioxy]bis[4-oxo-4H-1-benzopyran-2-carboxylate].

Cromolyn and the pharmaceutically acceptable salts described herein may be prepared as prodrugs. A “prodrug” refers to an agent that is converted into the parent drug in vivo. The prodrug can be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug, or to alter other characteristics or properties of a drug. In certain embodiments, the prodrug has improved bioavailability relative to the parent drug. In certain embodiments, the prodrug has improved solubility in pharmaceutical compositions over the parent drug. In certain embodiments, prodrugs may be designed as reversible drug derivatives, for use as modifiers to enhance drug transport to site-specific tissues. In certain embodiments, a prodrug of cromolyn is an ester of cromolyn, which is hydrolyzed to the carboxylic acid, the parent compound. In certain embodiments, a prodrug comprises a short peptide (polyaminoacid) bonded to an acid group, wherein the peptide is metabolized in vivo to reveal the parent drug. In certain embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically active form of cromolyn. In certain embodiments, a prodrug is enzymatically metabolized by one or more steps or processes to the parent compound. In certain embodiments, prodrug of cromolyn is used. In a specific embodiment, the prodrug of cromolyn is cromoglicate lisetil.

To produce a prodrug, a pharmaceutically active cromolyn is modified such that the active compound will be regenerated upon in vivo administration. In certain embodiments, prodrugs of cromolyn are designed by virtue of knowledge of pharmacodynamic processes and drug metabolism in vivo. See, e.g., Nogrady (1985) Medicinal Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392; Silverman (1992), The Organic Chemistry of Drug Design and Drug Action, Academic Press, Inc., San Diego, pages 352-401, Saulnier et al., (1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985; Rooseboom et al., Pharmacological Reviews, 56:53-102, 2004; Miller et al., J. Med. Chem. Vol. 46, no. 24, 5097-5116, 2003; Aesop Cho, “Recent Advances in Oral Prodrug Discovery”, Annual Reports in Medicinal Chemistry, Vol. 41, 395-407, 2006.

In certain embodiments, cromolyn and pharmaceutically acceptable salts thereof disclosed herein are isotopically-labeled compounds, which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into the present compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine and chlorine, such as, for example, 2H, 3H, 13C, 14C, 15N, 18O, 17O, 35S, 18F, 36Cl, respectively. Certain isotopically labeled compounds described herein, for example those with isotopes such as deuterium, i.e., 2H, can afford certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. In certain embodiments, isotopically labeled cromolyn is co-administered. In some, the pharmaceutically acceptable salt of cromolyn, such as cromolyn sodium, is isotopically labeled. In certain embodiments, the pharmaceutically acceptable salt of cromolyn is deuterium-labeled cromolyn sodium.

In certain embodiments, cromolyn and the pharmaceutically acceptable salt thereof described herein are pegylated, wherein one or more polyethylene glycol (PEG) polymers are covalently attached to cromolyn or the pharmaceutically acceptable salt thereof. In certain embodiments, pegylation increases the half-life of the pegylated compound in the body. In certain embodiments, pegylation increases the hydrodynamic size of the pegylated compound and reduces renal clearance. In certain embodiments, pegylation increases the solubility of the pegylated compound. In certain embodiments, protects the pegylated compound from proteolytic degradation.

Cromolyn and pharmaceutically acceptable salts, prodrugs, and adducts thereof, may be prepared by methods known in the art.

Cromolyn or a pharmaceutically acceptable salt thereof may be administered in the methods disclosed herein in a suitable dose or nominal dose as determined by one of ordinary skill in the art. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered at a dosage or nominal dosage of less than about 1 mg/dose, about 1 mg/dose to about 100 mg/dose, about 1 mg/dose to about 120 mg/dose, about 5 mg/dose to about 80 mg/dose, about 20 mg/dose to about 60 mg/dose, about 30 mg/dose to about 50 mg/dose, or greater than about 100 mg/dose. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in less than about 1 mg, about 1 mg, about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 105 mg, about 110 mg, about 115 mg, about 120 mg, about 125 mg, about 130 mg doses, about 135 mg, about 140 mg, about 145 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, or about 1000 mg doses. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 30 mg. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 40 mg. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 50 mg. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 60 mg. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 70 mg. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 80 mg.

In certain embodiments of the methods disclosed herein, cromolyn sodium is administered at a dosage or nominal dosage of less than about 1 mg/dose, about 1 mg/dose to about 100 mg/dose, about 1 mg/dose to about 120 mg/dose, about 5 mg/dose to about 80 mg/dose, about 20 mg/dose to about 60 mg/dose, or about 30 mg/dose to about 50 mg/dose, or greater than about 100 mg/dose. In other embodiments, cromolyn sodium is administered in less than about 1 mg, about 1 mg, about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 105 mg, about 110 mg, about 115 mg, about 120 mg, about 125 mg, about 130 mg doses, about 135 mg, about 140 mg, about 145 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, or about 1000 mg doses. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 30 mg. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 40 mg. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 50 mg. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 60 mg. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 70 mg. In certain embodiments, cromolyn or a pharmaceutically acceptable salt thereof is administered in about 80 mg.

Formulations

DSCG has a long history of use for its anti-allergy, anti-inflammatory, and immune-modulating properties as well as its exceptional safety profile. However, the available formulations of DSCG (that do not include the compositions of the disclosure) are not suitable for use in chronic cough associated with IPF, because the available formulations of DSCG are too limited by poor delivery efficiency and very low bioavailability (approximately 1%).

The compositions and formulations of the disclosure enhance bioavailability and provide efficacious treatment of the debilitating symptoms of chronic cough associated with IPF. The compositions and formulations of the disclosure achieve significantly higher lung and peripheral distribution than currently available formulations, parameters that are both required for efficacy in treatment of chronic cough associated with IPF.

Table 1a provides exemplary, nonlimiting, formulations of the compositions of the disclosure, wherein the amounts of each component of the formulations are expressed as weight percent of the total weight of the formulation.

PA101 PA101B PA101B PA101B Component Function (wt %) (wt %) (wt %) (wt %) Cromolyn Active Substance 4 2 4 6 Sodium Chloride Osmotic Agent 0.2 0.2 0.2 0.2 EDTA Chelating Agent 0.02 0.02 0.02 0.02 Mannitol Non-ionic Osmotic Agent 1.25 0 0 0 Water for Injection (WFI) Quantum sufficiat (q.s.) q.s. q.s. q.s. q.s. Osmolality (mOsm/kg) Tonicity 200 106 125 135

Table 1b provides further exemplary, nonlimiting, formulations of the compositions of the disclosure, wherein the amounts of each component of the formulations are expressed as weight percent of the total weight of the formulation.

Cromolyn Sodium sodium Mannitol chloride EDTA Osmolality Formulation (%) (%) (%) (%) (mOsm/kg) 1 2 0 0 0.02 42 2 2 0 0.2 0.02 106 3 2 0 0.4 0.02 170 4 2 0 0.6 0.02 235 5 2 0 0.8 0.02 299 6 4 1.25 0.2 0.02 199 7 4 1 0.2 0.02 183 8 4 0.75 0.2 0.02 169 9 4 0.5 0.2 0.02 154 10 4 0.25 0.2 0.02 139 11 4 0 0.2 0.02 125 12 5 1.25 0.2 0.02 207 13 5 0 0.2 0.02 131 14 5 0 0.25 0.02 147 15 6 1.25 0.2 0.02 214 16 6 0 0.2 0.02 138 17 6 0 0.25 0.02 154

PA101B formulations 4% by weight and 6% by weight, are each highly concentrated, well-tolerated, room-temperature stable formulations of disodium cromoglycate optimized for delivery via an electronic nebulizer.

In certain embodiments, compositions of the disclosure may comprise an ionic osmolarity or osmolality adjusting agent but, do not comprise a non-ionic osmolarity or osmolality adjusting agent. Ionic osmolarity or osmolality adjusting agents can be selected from, for example, alkali metal salts, such as sodium and potassium salts. Examples of such salts include, but are not limited to, sodium chloride, sodium gluconate, sodium pyruvate, and potassium chloride. It is possible to use a single ionic tonicity-adjusting agent, such as sodium chloride, or a mixture of such agents. The salts may be either added or formed in situ due to a salt formation process. In a particular embodiment of the disclosure, however, the non-ionic osmolarity or osmolality adjusting agent is mannitol. The non-ionic osmolarity or osmolality adjusting agent can be selected from, for example, the group of carbohydrates. Examples of carbohydrates that can be used for isotonisation include, but are not limited to, sugars such as glucose, lactose, sucrose and trehalose, and sugar alcohols such as mannitol, xylitol, sorbitol, and isomaltol. In a particular embodiment of the disclosure, however, the non-ionic osmolarity or osmolality adjusting agent is not propylene glycol, a cyclodextrin or mannitol.

The disclosure provides a pharmaceutically acceptable aerosol for the treatment of chronic cough in a subject having idiopathic pulmonary fibrosis (IPF), comprising droplets of an aqueous solution comprising (i) from about 2% to about 6% by weight of cromolyn sodium and (ii) an osmolarity adjusting agent comprising (a) between about 0.1% and about 0.5% by weight of sodium chloride, inclusive of the endpoints, and (b) optionally salts of ethylenediaminetetraacetic acid (EDTA), wherein said aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30%, and wherein said treatment of said chronic cough in said subject is achieved via delivery of a therapeutically effective amount of cromolyn sodium to the lungs of the subject by said subject orally inhaling said pharmaceutically acceptable aerosol. In certain embodiments, the pharmaceutically acceptable aerosol comprises droplets of an aqueous solution comprising salts of EDTA, wherein the salts are selected from sodium EDTA. In certain embodiments are the pharmaceutically aerosols wherein the aqueous solution contains from about 5 mg to about 80 mg of cromolyn sodium. In certain embodiments are the pharmaceutically aerosols wherein the aqueous solution contains from about 36 mg to about 44 mg of cromolyn sodium. In certain embodiments are the pharmaceutically aerosols wherein the aqueous solution contains about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 31 mg, about 32 mg, about 33 mg, about 34 mg, about 35 mg, about 36 mg, about 37 mg, about 38 mg, about 39 mg, about 40 mg, about 41 mg, about 42 mg, about 43 mg, about 44 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, or about 80 mg of cromolyn sodium. In certain embodiments, are the pharmaceutically acceptable aerosols having a respirable fraction (≤5 μm) as measured by USP <1601> of at least about 75%.. In certain embodiments are the pharmaceutically acceptable aerosols wherein said sodium chloride is at a concentration of between 0.1% to 0.2% by weight, inclusive of the endpoints.

The disclosure provides a pharmaceutically acceptable aerosol for the treatment of chronic cough in a subject having idiopathic pulmonary fibrosis (IPF), consisting of droplets of an aqueous solution consisting of (i) from about 2% to about 6% by weight of cromolyn sodium and (ii) an osmolarity adjusting agent consisting of (a) between about 0.1% and about 0.5% by weight of sodium chloride, inclusive of the endpoints, and (b) optionally salts of ethylenediaminetetraacetic acid (EDTA), wherein said aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30%, and wherein said treatment of said chronic cough in said subject is achieved via delivery of a therapeutically effective amount of cromolyn sodium to the lungs of the subject by said subject orally inhaling said pharmaceutically acceptable aerosol. In certain embodiments, the pharmaceutically acceptable aerosol comprises droplets of an aqueous solution comprising salts of EDTA, wherein the salts are selected from sodium EDTA. In certain embodiments are the pharmaceutically aerosols wherein the aqueous solution contains from about 5 mg to about 80 mg of cromolyn sodium. In certain embodiments are the pharmaceutically aerosols wherein the aqueous solution contains from about 36 mg to about 44 mg of cromolyn sodium. In certain embodiments are the pharmaceutically aerosols wherein the aqueous solution contains about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 31 mg, about 32 mg, about 33 mg, about 34 mg, about 35 mg, about 36 mg, about 37 mg, about 38 mg, about 39 mg, about 40 mg, about 41 mg, about 42 mg, about 43 mg, about 44 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, or about 80 mg of cromolyn sodium. In certain embodiments, are the pharmaceutically acceptable aerosols having a respirable fraction (≤5 μm) as measured by USP <1601> of at least about 75%. In certain embodiments are the pharmaceutically acceptable aerosols wherein said sodium chloride is at a concentration of between 0.1% to 0.2% by weight, inclusive of the endpoints.

In certain embodiments are provided methods of treating chronic cough in a subject having idiopathic pulmonary fibrosis (IPF), comprising administering a pharmaceutically acceptable aerosol to said subject by oral inhalation, said pharmaceutically acceptable aerosol acceptable consisting of droplets of an aqueous solution consisting of_(i) from about 2% to about 6% by weight of cromolyn sodium, (ii) between about 0.1% and about 0.5% by weight of sodium chloride, inclusive of the endpoints, and (iii) optionally salts of ethylenediaminetetraacetic acid (EDTA). In certain embodiments are the pharmaceutically acceptable aerosols, wherein said aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30%. In certain embodiments are the pharmaceutically acceptable aerosols, wherein said aerosol has a respirable fraction (≤5 μm) as measured by USP <1601> of at least about 75%. In certain embodiments, the pharmaceutically acceptable aerosol comprises droplets of an aqueous solution comprising salts of EDTA, wherein the salts are selected from sodium EDTA. In certain embodiments are the pharmaceutically aerosols wherein the aqueous solution contains from about 5 mg to about 80 mg of cromolyn sodium. In certain embodiments are the pharmaceutically aerosols wherein the aqueous solution contains from about 36 mg to about 44 mg of cromolyn sodium. In certain embodiments are the pharmaceutically aerosols wherein the aqueous solution contains about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 31 mg, about 32 mg, about 33 mg, about 34 mg, about 35 mg, about 36 mg, about 37 mg, about 38 mg, about 39 mg, about 40 mg, about 41 mg, about 42 mg, about 43 mg, about 44 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, or about 80 mg of cromolyn sodium. In certain embodiments, are the pharmaceutically acceptable aerosols having a respirable fraction (≤5 μm) as measured by USP <1601> of at least about 75%.. In certain embodiments are the pharmaceutically acceptable aerosols wherein said sodium chloride is at a concentration of between 0.1% to 0.2% by weight, inclusive of the endpoints.

In certain embodiments, formulations administered in the methods disclosed herein produce in a subject AUC(0-∞) of cromolyn or a pharmaceutically acceptable salt thereof greater than about 100 ng*hr./mL, greater than about 110 ng*hr./mL, greater than about 120 ng*hr./mL, greater than about 130 ng*hr./mL, greater than about 140 ng*hr./mL, greater than about 150 ng*hr./mL, greater than about 160 ng*hr./mL, greater than about 170 ng*hr./mL, greater than about 180 ng*hr./mL, greater than about 190 ng*hr./mL, greater than about 200 ng*hr./mL, greater than about 225 ng*hr./mL, greater than about 250 ng*hr./mL, greater than about 275 ng*hr./mL, greater than about 300 ng*hr./mL, greater than about 325 ng*hr./mL, greater than about 350 ng*hr./mL, greater than about 375 ng*hr./mL, greater than about 400 ng*hr./mL, greater than about 425 ng*hr./mL, greater than about 450 ng*hr./mL, greater than about 475 ng*hr./mL, greater than about 500 ng*hr./mL, greater than about 525 ng*hr./mL, greater than about 550 ng*hr./mL, greater than about 575 ng*hr./mL, greater than about 600 ng*hr./mL, greater than about 625 ng*hr./mL, greater than about 650 ng*hr./mL, greater than about 675 ng*hr./mL, greater than about 700 ng*hr./mL, greater than about 725 ng*hr./mL, greater than about 750 ng*hr./mL, greater than about 775 ng*hr./mL, greater than about 800 ng*hr./mL, greater than about 825 ng*hr./mL, greater than about 850 ng*hr./mL, greater than about 875 ng*hr./mL, greater than about 900 ng*hr./mL, greater than about 925 ng*hr./mL, greater than about 950 ng*hr./mL, greater than about 975 ng*hr./mL, or greater than about 1000 ng*hr./mL after administration of the formulation to the subject. In certain embodiments, formulations administered in the methods disclosed herein produce in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 100 ng*hr./mL, greater than about 110 ng*hr./mL, greater than about 120 ng*hr./mL, greater than about 130 ng*hr./mL, greater than about 140 ng*hr./mL, greater than about 150 ng*hr./mL, greater than about 160 ng*hr./mL, greater than about 170 ng*hr./mL, greater than about 180 ng*hr./mL, greater than about 190 ng*hr./mL, greater than about 200 ng*hr./mL, greater than about 225 ng*hr./mL, greater than about 250 ng*hr./mL, greater than about 275 ng*hr./mL, greater than about 300 ng*hr./mL, greater than about 325 ng*hr./mL, greater than about 350 ng*hr./mL, greater than about 375 ng*hr./mL, greater than about 400 ng*hr./mL, greater than about 425 ng*hr./mL, greater than about 450 ng*hr./mL, greater than about 475 ng*hr./mL, greater than about 500 ng*hr./mL, greater than about 525 ng*hr./mL, greater than about 550 ng*hr./mL, greater than about 575 ng*hr./mL, greater than about 600 ng*hr./mL, greater than about 625 ng*hr./mL, greater than about 650 ng*hr./mL, greater than about 675 ng*hr./mL, greater than about 700 ng*hr./mL, greater than about 725 ng*hr./mL, greater than about 750 ng*hr./mL, greater than about 775 ng*hr./mL, greater than about 800 ng*hr./mL, greater than about 825 ng*hr./mL, greater than about 850 ng*hr./mL, greater than about 875 ng*hr./mL, greater than about 900 ng*hr./mL, greater than about 925 ng*hr./mL, greater than about 950 ng*hr./mL, greater than about 975 ng*hr./mL, or greater than about 1000 ng*hr./mL after administration of the formulation to the subject or subject.

In certain embodiments, formulations administered in the methods disclosed herein produce in a subject AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof of about 100 ng*hr./mL, about 110 ng*hr./mL, about 120 ng*hr./mL, about 130 ng*hr./mL, about 140 ng*hr./mL, about 150 ng*hr./mL, about 160 ng*hr./mL, about 170 ng*hr./mL, about 180 ng*hr./mL, about 190 ng*hr./mL, about 200 ng*hr./mL, about 225 ng*hr./mL, about 250 ng*hr./mL, about 275 ng*hr./mL, about 300 ng*hr./mL, about 325 ng*hr./mL, about 350 ng*hr./mL, about 375 ng*hr./mL, about 400 ng*hr./mL, about 425 ng*hr./mL, about 450 ng*hr./mL, about 475 ng*hr./mL, about 500 ng*hr./mL, about 525 ng*hr./mL, about 550 ng*hr./mL, about 575 ng*hr./mL, about 600 ng*hr./mL, about 625 ng*hr./mL, about 650 ng*hr./mL, about 675 ng*hr./mL, about 700 ng*hr./mL, about 725 ng*hr./mL, about 750 ng*hr./mL, about 775 ng*hr./mL, about 800 ng*hr./mL, about 825 ng*hr./mL, about 850 ng*hr./mL, about 875 ng*hr./mL, about 900 ng*hr./mL, about 925 ng*hr./mL, about 950 ng*hr./mL, about 975 ng*hr./mL, or about 1000 ng*hr./mL after administration of the formulation to the subject. In certain embodiments, formulations administered in the methods disclosed herein produce in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof of about 100 ng*hr./mL, about 110 ng*hr./mL, about 120 ng*hr./mL, about 130 ng*hr./mL, about 140 ng*hr./mL, about 150 ng*hr./mL, about 160 ng*hr./mL, about 170 ng*hr./mL, about 180 ng*hr./mL, about 190 ng*hr./mL, about 200 ng*hr./mL, about 225 ng*hr./mL, about 250 ng*hr./mL, about 275 ng*hr./mL, about 300 ng*hr./mL, about 325 ng*hr./mL, about 350 ng*hr./mL, about 375 ng*hr./mL, about 400 ng*hr./mL, about 425 ng*hr./mL, about 450 ng*hr./mL, about 475 ng*hr./mL, about 500 ng*hr./mL, about 525 ng*hr./mL, about 550 ng*hr./mL, about 575 ng*hr./mL, about 600 ng*hr./mL, about 625 ng*hr./mL, about 650 ng*hr./mL, about 675 ng*hr./mL, about 700 ng*hr./mL, about 725 ng*hr./mL, about 750 ng*hr./mL, about 775 ng*hr./mL, about 800 ng*hr./mL, about 825 ng*hr./mL, about 850 ng*hr./mL, about 875 ng*hr./mL, about 900 ng*hr./mL, about 925 ng*hr./mL, about 950 ng*hr./mL, about 975 ng*hr./mL, or about 1000 ng*hr./mL after administration of the formulation to the subject or subject.

In certain embodiments, formulations administered in the methods disclosed herein produce in a subject an AUC(0-∞) of cromolyn sodium greater than about 100 ng*hr./mL, greater than about 110 ng*hr./mL, greater than about 120 ng*hr./mL, greater than about 130 ng*hr./mL, greater than about 140 ng*hr./mL, greater than about 150 ng*hr./mL, greater than about 160 ng*hr./mL, greater than about 170 ng*hr./mL, greater than about 180 ng*hr./mL, greater than about 190 ng*hr./mL, greater than about 200 ng*hr./mL, greater than about 225 ng*hr./mL, greater than about 250 ng*hr./mL, greater than about 275 ng*hr./mL, greater than about 300 ng*hr./mL, greater than about 325 ng*hr./mL, greater than about 350 ng*hr./mL, greater than about 375 ng*hr./mL, greater than about 400 ng*hr./mL, greater than about 425 ng*hr./mL, greater than about 450 ng*hr./mL, greater than about 475 ng*hr./mL, greater than about 500 ng*hr./mL, greater than about 525 ng*hr./mL, greater than about 550 ng*hr./mL, greater than about 575 ng*hr./mL, greater than about 600 ng*hr./mL, greater than about 625 ng*hr./mL, greater than about 650 ng*hr./mL, greater than about 675 ng*hr./mL, greater than about 700 ng*hr./mL, greater than about 725 ng*hr./mL, greater than about 750 ng*hr./mL, greater than about 775 ng*hr./mL, greater than about 800 ng*hr./mL, greater than about 825 ng*hr./mL, greater than about 850 ng*hr./mL, greater than about 875 ng*hr./mL, greater than about 900 ng*hr./mL, greater than about 925 ng*hr./mL, greater than about 950 ng*hr./mL, greater than about 975 ng*hr./mL, or greater than about 1000 ng*hr./mL after administration of the formulation to the subject. In certain embodiments, formulations administered in the methods disclosed herein produce in a subject an AUC(0-∞) of cromolyn sodium greater than about 100 ng*hr./mL, greater than about 110 ng*hr./mL, greater than about 120 ng*hr./mL, greater than about 130 ng*hr./mL, greater than about 140 ng*hr./mL, greater than about 150 ng*hr./mL, greater than about 160 ng*hr./mL, greater than about 170 ng*hr./mL, greater than about 180 ng*hr./mL, greater than about 190 ng*hr./mL, greater than about 200 ng*hr./mL, greater than about 225 ng*hr./mL, greater than about 250 ng*hr./mL, greater than about 275 ng*hr./mL, greater than about 300 ng*hr./mL, greater than about 325 ng*hr./mL, greater than about 350 ng*hr./mL, greater than about 375 ng*hr./mL, greater than about 400 ng*hr./mL, greater than about 425 ng*hr./mL, greater than about 450 ng*hr./mL, greater than about 475 ng*hr./mL, greater than about 500 ng*hr./mL, greater than about 525 ng*hr./mL, greater than about 550 ng*hr./mL, greater than about 575 ng*hr./mL, greater than about 600 ng*hr./mL, greater than about 625 ng*hr./mL, greater than about 650 ng*hr./mL, greater than about 675 ng*hr./mL, greater than about 700 ng*hr./mL, greater than about 725 ng*hr./mL, greater than about 750 ng*hr./mL, greater than about 775 ng*hr./mL, greater than about 800 ng*hr./mL, greater than about 825 ng*hr./mL, greater than about 850 ng*hr./mL, greater than about 875 ng*hr./mL, greater than about 900 ng*hr./mL, greater than about 925 ng*hr./mL, greater than about 950 ng*hr./mL, greater than about 975 ng*hr./mL, or greater than about 1000 ng*hr./mL after administration of the formulation to the subject or subject.

In certain embodiments, formulations administered in the methods disclosed herein produce in a subject an AUC(0-∞) of cromolyn sodium of about 100 ng*hr./mL, about 110 ng*hr./mL, about 120 ng*hr./mL, about 130 ng*hr./mL, about 140 ng*hr./mL, about 150 ng*hr./mL, about 160 ng*hr./mL, about 170 ng*hr./mL, about 180 ng*hr./mL, about 190 ng*hr./mL, about 200 ng*hr./mL, about 225 ng*hr./mL, about 250 ng*hr./mL, about 275 ng*hr./mL, about 300 ng*hr./mL, about 325 ng*hr./mL, about 350 ng*hr./mL, about 375 ng*hr./mL, about 400 ng*hr./mL, about 425 ng*hr./mL, about 450 ng*hr./mL, about 475 ng*hr./mL, about 500 ng*hr./mL, about 525 ng*hr./mL, about 550 ng*hr./mL, about 575 ng*hr./mL, about 600 ng*hr./mL, about 625 ng*hr./mL, about 650 ng*hr./mL, about 675 ng*hr./mL, about 700 ng*hr./mL, about 725 ng*hr./mL, about 750 ng*hr./mL, about 775 ng*hr./mL, about 800 ng*hr./mL, about 825 ng*hr./mL, about 850 ng*hr./mL, about 875 ng*hr./mL, about 900 ng*hr./mL, about 925 ng*hr./mL, about 950 ng*hr./mL, about 975 ng*hr./mL, or about 1000 ng*hr./mL after administration of the formulation to the subject. In certain embodiments, formulations administered in the methods disclosed herein produce in a subject an AUC(0-∞) of cromolyn sodium of about 100 ng*hr./mL, about 110 ng*hr./mL, about 120 ng*hr./mL, about 130 ng*hr./mL, about 140 ng*hr./mL, about 150 ng*hr./mL, about 160 ng*hr./mL, about 170 ng*hr./mL, about 180 ng*hr./mL, about 190 ng*hr./mL, about 200 ng*hr./mL, about 225 ng*hr./mL, about 250 ng*hr./mL, about 275 ng*hr./mL, about 300 ng*hr./mL, about 325 ng*hr./mL, about 350 ng*hr./mL, about 375 ng*hr./mL, about 400 ng*hr./mL, about 425 ng*hr./mL, about 450 ng*hr./mL, about 475 ng*hr./mL, about 500 ng*hr./mL, about 525 ng*hr./mL, about 550 ng*hr./mL, about 575 ng*hr./mL, about 600 ng*hr./mL, about 625 ng*hr./mL, about 650 ng*hr./mL, about 675 ng*hr./mL, about 700 ng*hr./mL, about 725 ng*hr./mL, about 750 ng*hr./mL, about 775 ng*hr./mL, about 800 ng*hr./mL, about 825 ng*hr./mL, about 850 ng*hr./mL, about 875 ng*hr./mL, about 900 ng*hr./mL, about 925 ng*hr./mL, about 950 ng*hr./mL, about 975 ng*hr./mL, or about 1000 ng*hr./mL after administration of the formulation to the subject.

In certain embodiments, formulation administered in the methods disclosed herein produce in a subject a Cmax of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 40 ng/mL, greater than about 50 ng/mL, greater than about 60 ng/mL, greater than about 70 ng/mL, greater than about 80 ng/mL, greater than about 90 ng/mL, greater than about 100 ng/mL, greater than about 110 ng/mL, greater than about 120 ng/mL, greater than about 130 ng/mL, greater than about 140 ng/mL, greater than about 150 ng/mL, greater than about 160 ng/mL, greater than about 170 ng/mL, greater than about 180 ng/mL, greater than about 190 ng/mL, greater than about 200 ng/mL, greater than about 210 ng/mL, greater than about 220 ng/mL, greater than about 230 ng/mL, greater than about 240 ng/mL, greater than about 250 ng/mL, greater than about 260 ng/mL, greater than about 270 ng/mL, greater than about 280 ng/mL, greater than about 290 ng/mL, greater than about 300 ng/mL, greater than about 310 ng/mL, greater than about 320 ng/mL, greater than about 330 ng/mL, greater than about 340 ng/mL, greater than about 350 ng/mL, greater than about 360 ng/mL, greater than about 370 ng/mL, greater than about 380 ng/mL, greater than about 390 ng/mL, or greater than about 400 ng/mL after administration of the formulation to the subject. In certain embodiments, formulations administered in the methods disclosed herein produce in a subject a Cm of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 40 ng/mL, greater than about 50 ng/mL, greater than about 60 ng/mL, greater than about 70 ng/mL, greater than about 80 ng/mL, greater than about 90 ng/mL, greater than about 100 ng/mL, greater than about 110 ng/mL, greater than about 120 ng/mL, greater than about 130 ng/mL, greater than about 140 ng/mL, greater than about 150 ng/mL, greater than about 160 ng/mL, greater than about 170 ng/mL, greater than about 180 ng/mL, greater than about 190 ng/mL, greater than about 200 ng/mL, greater than about 210 ng/mL, greater than about 220 ng/mL, greater than about 230 ng/mL, greater than about 240 ng/mL, greater than about 250 ng/mL, greater than about 260 ng/mL, greater than about 270 ng/mL, greater than about 280 ng/mL, greater than about 290 ng/mL, greater than about 300 ng/mL, greater than about 310 ng/mL, greater than about 320 ng/mL, greater than about 330 ng/mL, greater than about 340 ng/mL, greater than about 350 ng/mL, greater than about 360 ng/mL, greater than about 370 ng/mL, greater than about 380 ng/mL, greater than about 390 ng/mL, or greater than about 400 ng/mL after administration of the formulation to the subject or subject.

In certain embodiments, formulations administered in the methods disclosed herein produce in a subject a Cmax of cromolyn or a pharmaceutically-acceptable salt thereof of about 50 mg/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, 90 ng/mL, about 100 ng/mL, about 110 ng/mL, about 120 ng/mL, about 130 ng/mL, about 140 ng/mL, about 150 ng/mL, about 160 ng/mL, about 170 ng/mL, about 180 ng/mL, about 190 ng/mL, about 200 ng/mL, about 210 ng/mL, about 220 ng/mL, about 230 ng/mL, about 240 ng/mL, about 250 ng/mL, 260 ng/mL, about 270 ng/mL, about 280 ng/mL, about 290 ng/mL, about 300 ng/mL, about 310 ng/mL, about 320 ng/mL, about 330 ng/mL, about 340 ng/mL, about 350 ng/mL, about 360 ng/mL, about 370 ng/mL, about 380 ng/mL, about 390 ng/mL, or about 400 ng/mL after administration of the formulation to the subject. In certain embodiments, formulations administered in the methods disclosed herein produce in a subject a Cm of cromolyn or a pharmaceutically-acceptable salt thereof of about 50 mg/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, 90 ng/mL, about 100 ng/mL, about 110 ng/mL, about 120 ng/mL, about 130 ng/mL, about 140 ng/mL, about 150 ng/mL, about 160 ng/mL, about 170 ng/mL, about 180 ng/mL, about 190 ng/mL, about 200 ng/mL, about 210 ng/mL, about 220 ng/mL, about 230 ng/mL, about 240 ng/mL, about 250 ng/mL, 260 ng/mL, about 270 ng/mL, about 280 ng/mL, about 290 ng/mL, about 300 ng/mL, about 310 ng/mL, about 320 ng/mL, about 330 ng/mL, about 340 ng/mL, about 350 ng/mL, about 360 ng/mL, about 370 ng/mL, about 380 ng/mL, about 390 ng/mL, or about 400 ng/mL after administration of the formulation to the subject or subject.

In certain embodiments, formulations administered in the methods disclosed herein produce in a subject a Cmax of cromolyn sodium greater than about 40 ng/mL, greater than about 50 ng/mL, greater than about 60 ng/mL, greater than about 70 ng/mL, greater than about 80 ng/mL, greater than about 90 ng/mL, greater than about 100 ng/mL, greater than about 110 ng/mL, greater than about 120 ng/mL, greater than about 130 ng/mL, greater than about 140 ng/mL, greater than about 150 ng/mL, greater than about 160 ng/mL, greater than about 170 ng/mL, greater than about 180 ng/mL, greater than about 190 ng/mL, greater than about 200 ng/mL, greater than about 210 ng/mL, greater than about 220 ng/mL, greater than about 230 ng/mL, greater than about 240 ng/mL, greater than about 250 ng/mL, greater than about 260 ng/mL, greater than about 270 ng/mL, greater than about 280 ng/mL, greater than about 290 ng/mL, greater than about 300 ng/mL, greater than about 310 ng/mL, greater than about 320 ng/mL, greater than about 330 ng/mL, greater than about 340 ng/mL, greater than about 350 ng/mL, greater than about 360 ng/mL, greater than about 370 ng/mL, greater than about 380 ng/mL, greater than about 390 ng/mL, or greater than about 400 ng/mL after administration of the formulation to the subject. In certain embodiments, formulations administered in the methods disclosed herein produce in a subject a Cmax of cromolyn sodium greater than about 40 ng/mL, greater than about 50 ng/mL, greater than about 60 ng/mL, greater than about 70 ng/mL, greater than about 80 ng/mL, greater than about 90 ng/mL, greater than about 100 ng/mL, greater than about 110 ng/mL, greater than about 120 ng/mL, greater than about 130 ng/mL, greater than about 140 ng/mL, greater than about 150 ng/mL, greater than about 160 ng/mL, greater than about 170 ng/mL, greater than about 180 ng/mL, greater than about 190 ng/mL, greater than about 200 ng/mL, greater than about 210 ng/mL, greater than about 220 ng/mL, greater than about 230 ng/mL, greater than about 240 ng/mL, greater than about 250 ng/mL, greater than about 260 ng/mL, greater than about 270 ng/mL, greater than about 280 ng/mL, greater than about 290 ng/mL, greater than about 300 ng/mL, greater than about 310 ng/mL, greater than about 320 ng/mL, greater than about 330 ng/mL, greater than about 340 ng/mL, greater than about 350 ng/mL, greater than about 360 ng/mL, greater than about 370 ng/mL, greater than about 380 ng/mL, greater than about 390 ng/mL, or greater than about 400 ng/mL after administration of the formulation to the subject or subject.

In certain embodiments, formulations administered in the methods disclosed herein produce in a subject a Cmax of cromolyn sodium of about 50 mg/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, 90 ng/mL, about 100 ng/mL, about 110 ng/mL, about 120 ng/mL, about 130 ng/mL, about 140 ng/mL, about 150 ng/mL, about 160 ng/mL, about 170 ng/mL, about 180 ng/mL, about 190 ng/mL, about 200 ng/mL, about 210 ng/mL, about 220 ng/mL, about 230 ng/mL, about 240 ng/mL, about 250 ng/mL, 260 ng/mL, about 270 ng/mL, about 280 ng/mL, about 290 ng/mL, about 300 ng/mL, about 310 ng/mL, about 320 ng/mL, about 330 ng/mL, about 340 ng/mL, about 350 ng/mL, about 360 ng/mL, about 370 ng/mL, about 380 ng/mL, about 390 ng/mL, or about 400 ng/mL after administration of the formulation to the subject. In certain embodiments, formulations administered in the methods disclosed herein produce in a subject a Cmax of cromolyn sodium of about 50 mg/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, 90 ng/mL, about 100 ng/mL, about 110 ng/mL, about 120 ng/mL, about 130 ng/mL, about 140 ng/mL, about 150 ng/mL, about 160 ng/mL, about 170 ng/mL, about 180 ng/mL, about 190 ng/mL, about 200 ng/mL, about 210 ng/mL, about 220 ng/mL, about 230 ng/mL, about 240 ng/mL, about 250 ng/mL, 260 ng/mL, about 270 ng/mL, about 280 ng/mL, about 290 ng/mL, about 300 ng/mL, about 310 ng/mL, about 320 ng/mL, about 330 ng/mL, about 340 ng/mL, about 350 ng/mL, about 360 ng/mL, about 370 ng/mL, about 380 ng/mL, about 390 ng/mL, or about 400 ng/mL after administration of the formulation to the subject or subject.

Cromolyn or a pharmaceutically acceptable salt thereof may be administered to a subject in the methods disclosed herein with an inhalation device, e.g., a nebulizer.

Cromolyn or a pharmaceutically acceptable salt thereof may be formulated into any suitable dosage form, including but not limited to aerosols, aqueous oral dispersions, self-emulsifying dispersions, liposomal dispersions, pegylated liposomes, liquids, elixirs, suspensions, aerosols, controlled release formulations, lyophilized formulations, powders, delayed release formulations, extended release formulations, multiparticulate formulations or mixed immediate release formulations. Such formulations may be manufactured in a conventional manner, such as, by way of example only, conventional mixing, dissolving, or granulating processes.

In certain embodiments, the formulations disclosed herein may include one or more inactive ingredients or pharmaceutical excipients that provide suitable properties of the formulation. Such inactive ingredients may include one or more of the following classes.

“Albumin” refers to a family of globular proteins, the most common of which is serum albumin. Albumins are commonly found in blood plasma and function to regulate colloidal osmotic pressure of the blood. Albumin proteins found in the plasma bind some pharmaceutical compounds to form complexes. Complexation of albumin with pharmaceutical compounds, e.g., cromolyn or a pharmaceutical salt thereof, can influence the pharmaceutical compounds' plasma half-life and/or biological half-life in the body by preventing metabolism and/or excretion of the complexed compounds. In certain embodiments, compositions disclosed herein include albumin and cromolyn or a pharmaceutical salt thereof (e.g. cromolyn sodium).

“Antifoaming agents” reduce foaming during processing which can result in coagulation of aqueous dispersions, bubbles in the finished film, or generally impair processing. Exemplary anti-foaming agents include silicon emulsions or sorbitan sesquoleate

“Antioxidants” include, for example, butylated hydroxytoluene (BHT), sodium ascorbate, ascorbic acid, sodium metabisulfite and tocopherol. In certain embodiments, antioxidants enhance chemical stability where required.

“Binders” impart cohesive qualities and include, e.g., alginic acid and salts thereof; cellulose derivatives such as carboxymethylcellulose, methylcellulose (e.g., Methocel®), hydroxypropylmethylcellulose, hydroxyethylcellulose, hydroxypropylcellulose (e.g., Klucel®), ethylcellulose (e.g., Ethocel®), and microcrystalline cellulose (e.g., Avicel®); microcrystalline dextrose; amylose; magnesium aluminum silicate; polysaccharide acids; bentonites; gelatin; polyvinylpyrrolidone/vinyl acetate copolymer; crosspovidone; povidone; starch; pregelatinized starch; tragacanth, dextrin, a sugar, such as sucrose (e.g., Dipac®), glucose, dextrose, molasses, mannitol, sorbitol, xylitol (e.g., Xylitab®), and lactose; a natural or synthetic gum such as acacia, tragacanth, ghatti gum, mucilage of isapol husks, polyvinylpyrrolidone (e.g., Polyvidone® CL, Kollidon® CL, Polyplasdone® XL-10), larch arabogalactan, Veegum®, polyethylene glycol, waxes, sodium alginate, and the like.

“Carriers” or “carrier materials” include any commonly used excipients in pharmaceutics and should be selected on the basis of compatibility with cromolyn or a pharmaceutically acceptable salt thereof and the release profile properties of the desired dosage form. Exemplary carrier materials include, e.g., binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, and the like. “Pharmaceutically compatible carrier materials” may include, but are not limited to, acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerine, magnesium silicate, polyvinylpyrrollidone (PVP), cholesterol, cholesterol esters, sodium caseinate, soy lecithin, taurocholic acid, phosphotidylcholine, sodium chloride, tricalcium phosphate, dipotassium phosphate, cellulose and cellulose conjugates, sugars sodium stearoyl lactylate, carrageenan, monoglyceride, diglyceride, pregelatinized starch, and the like. See, e.g., Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa. 1975; Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins 1999).

“Dispersing agents” and/or “viscosity modulating agents” include materials that control the diffusion and homogeneity of a drug through liquid media or a granulation method or blend method. In certain embodiments, these agents also facilitate the effectiveness of a coating or eroding matrix. Exemplary diffusion facilitators/dispersing agents include, e.g., hydrophilic polymers, electrolytes, Tween® 60 or 80, PEG, Tyloxapol, polyvinylpyrrolidone (PVP; commercially known as Plasdone®), and the carbohydrate-based dispersing agents such as, for example, hydroxypropyl celluloses (e.g., HPC, HPC-SL, and HPC-L), hydroxypropyl methylcelluloses (e.g., HPMC K100, HPMC K4M, HPMC K15M, and HPMC K100M), carboxymethylcellulose sodium, methylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose acetate stearate (HPMCAS), noncrystalline cellulose, magnesium aluminum silicate, triethanolamine, polyvinyl alcohol (PVA), vinyl pyrrolidone/vinyl acetate copolymer (S630), 4-(1,1,3,3-tetramethylbutyl)-phenol polymer with ethylene oxide and formaldehyde (also known as tyloxapol), poloxamers (e.g., Pluronics F68®, F88®, and F108®, which are block copolymers of ethylene oxide and propylene oxide); and poloxamines (e.g., Tetronic 908®, also known as Poloxamine 908®, which is a tetrafunctional block copolymer derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine (BASF Corporation, Parsippany, N.J.)), polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30, polyvinylpyrrolidone/vinyl acetate copolymer (S-630), polyethylene glycol, e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 7000 to about 5400, sodium carboxymethylcellulose, methylcellulose, polysorbate-80, sodium alginate, gums, such as, e.g., gum tragacanth and gum acacia, guar gum, xanthans, including xanthan gum, sugars, cellulosics, such as, e.g., sodium carboxymethylcellulose, methylcellulose, sodium carboxymethylcellulose, polysorbate-80, sodium alginate, polyethoxylated sorbitan monolaurate, polyethoxylated sorbitan monolaurate, povidone, carbomers, polyvinyl alcohol (PVA), alginates, chitosans and combinations thereof. Plasticizcers such as cellulose or triethyl cellulose can also be used as dispersing agents. Dispersing agents particularly useful in liposomal dispersions and self-emulsifying dispersions are dimyristoyl phosphatidylcholine, natural phosphatidyl choline from eggs, natural phosphatidyl glycerol from eggs, cholesterol and isopropyl myristate.

“Diluent” refers to chemical compounds that are used to dilute the compound of interest (i.e. cromolyn or a pharmaceutically acceptable salt thereof) prior to delivery. Diluents can also be used to stabilize compounds because they can provide a more stable environment. Salts dissolved in buffered solutions, including, but not limited to, a phosphate buffered saline solution, are utilized as diluents in the art, and can also provide pH control or maintenance. In certain embodiments, diluents increase bulk of the composition to facilitate compression or create sufficient bulk for homogenous blend for capsule filling. Such compounds include e.g., lactose, starch, mannitol, sorbitol, dextrose, microcrystalline cellulose such as Avicel®; dibasic calcium phosphate, dicalcium phosphate dihydrate; tricalcium phosphate, calcium phosphate; anhydrous lactose, spray-dried lactose; pregelatinized starch, compressible sugar, such as Di-Pac® (Amstar); mannitol, hydroxypropylmethylcellulose, hydroxypropylmethylcellulose acetate stearate, sucrose-based diluents, confectioner's sugar; monobasic calcium sulfate monohydrate, calcium sulfate dihydrate; calcium lactate trihydrate, dextrates; hydrolyzed cereal solids, amylose; powdered cellulose, calcium carbonate; glycine, kaolin; mannitol, sodium chloride; inositol, bentonite, and the like.

“Flavoring agents” and/or “sweeteners” useful in the formulations described herein, include, e.g., acacia syrup, acesulfame K, alitame, anise, apple, aspartame, banana, Bavarian cream, berry, black currant, butterscotch, calcium citrate, camphor, caramel, cherry, cherry cream, chocolate, cinnamon, bubble gum, citrus, citrus punch, citrus cream, cotton candy, cocoa, cola, cool cherry, cool citrus, cyclamate, cylamate, dentomint, dextrose, eucalyptus, eugenol, fructose, fruit punch, ginger, glycyrrhetinate, glycyrrhiza (licorice) syrup, grape, grapefruit, honey, isomalt, lemon, lime, lemon cream, monoammonium glyrrhizinate (MagnaSweet®), maltol, mannitol, maple, marshmallow, menthol, mint cream, mixed berry, neohesperidine DC, neotame, orange, pear, peach, peppermint, peppermint cream, Prosweet® Powder, raspberry, root beer, rum, saccharin, safrole, sorbitol, spearmint, spearmint cream, strawberry, strawberry cream, stevia, sucralose, sucrose, sodium saccharin, saccharin, aspartame, acesulfame potassium, mannitol, talin, sylitol, sucralose, sorbitol, Swiss cream, tagatose, tangerine, thaumatin, tutti fruitti, vanilla, walnut, watermelon, wild cherry, wintergreen, xylitol, or any combination of these flavoring ingredients, e.g., anise-menthol, cherry-anise, cinnamon-orange, cherry-cinnamon, chocolate-mint, honey-lemon, lemon-lime, lemon-mint, menthol-eucalyptus, orange-cream, vanilla-mint, and mixtures thereof.

“Lubricants” and “glidants” are compounds that prevent, reduce or inhibit adhesion or friction of materials. Exemplary lubricants include, e.g., stearic acid, calcium hydroxide, talc, sodium stearyl fumerate, a hydrocarbon such as mineral oil, or hydrogenated vegetable oil such as hydrogenated soybean oil (Sterotex®), higher fatty acids and their alkali-metal and alkaline earth metal salts, such as aluminum, calcium, magnesium, zinc, stearic acid, sodium stearates, glycerol, talc, waxes, Stearowet®, boric acid, sodium benzoate, sodium acetate, sodium chloride, leucine, a polyethylene glycol (e.g., PEG-4000) or a methoxypolyethylene glycol such as Carbowax™, sodium oleate, sodium benzoate, glyceryl behenate, polyethylene glycol, magnesium or sodium lauryl sulfate, colloidal silica such as Syloid™, Cab-O-Sil®, a starch such as corn starch, silicone oil, a surfactant, and the like.

“Plasticizers” are compounds used to soften the microencapsulation material or film coatings to make them less brittle. Suitable plasticizers include, e.g., polyethylene glycols such as PEG 300, PEG 400, PEG 600, PEG 1450, PEG 3350, and PEG 800, stearic acid, propylene glycol, oleic acid, triethyl cellulose and triacetin. In certain embodiments, plasticizers can also function as dispersing agents or wetting agents.

In certain embodiments, compositions provided herein may also include one or more preservatives to inhibit microbial activity. Suitable preservatives include mercury-containing substances such as merfen and thiomersal; stabilized chlorine dioxide; octinidine; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.

“Solubilizers” include compounds such as triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, sodium lauryl sulfate, sodium doccusate, vitamin E TPGS, polysorbates (Tweens) dimethylacetamide, N-methylpyrrolidone, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropylmethyl cellulose, hydroxypropyl cyclodextrins, ethanol, n-butanol, isopropyl alcohol, cholesterol, bile salts, polyethylene glycol 200-600, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide and the like.

“Stabilizers” include compounds such as any antioxidation agents, e.g., citric acid, EDTA and pharmaceutically acceptable salts thereof, buffers, acids, preservatives and the like.

“Suspending agents” include compounds such as polyvinylpyrrolidone, e.g., polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25, or polyvinylpyrrolidone K30, vinyl pyrrolidone/vinyl acetate copolymer (S630), polyethylene glycol, e.g., the polyethylene glycol can have a molecular weight of about 300 to about 6000, or about 3350 to about 4000, or about 7000 to about 5400, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, hydroxymethylcellulose acetate stearate, polysorbate-80, hydroxyethylcellulose, sodium alginate, gums, such as, e.g., gum tragacanth and gum acacia, guar gum, xanthans, including xanthan gum, sugars, cellulosics, such as, e.g., sodium carboxymethylcellulose, methylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, hydroxyethylcellulose, polysorbate-80, sodium alginate, polyethoxylated sorbitan monolaurate, polyethoxylated sorbitan monolaurate, povidone and the like.

“Surfactants” include compounds such as sodium lauryl sulfate, sodium docusate, Tween 60 or 80, triacetin, vitamin E TPGS, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF), and the like. Some other surfactants include polyoxyethylene fatty acid glycerides and vegetable oils, e.g., polyoxyethylene (60) hydrogenated castor oil; and polyoxyethylene alkylethers and alkylphenyl ethers, e.g., octoxynol 10, octoxynol 40. In certain embodiments, surfactants may be included to enhance physical stability or for other purposes.

“Viscosity enhancing agents” include, e.g., methyl cellulose, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, hydroxypropylmethyl cellulose acetate stearate, hydroxypropylmethyl cellulose phthalate, carbomer, polyvinyl alcohol, alginates, acacia, chitosans, and combinations thereof.

“Wetting agents” include compounds such as oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium docusate, sodium oleate, sodium lauryl sulfate, sodium doccusate, triacetin, Tween 80, vitamin E TPGS, ammonium salts and the like.

It should be appreciated that there is considerable overlap between classes of inactive ingredients. Thus, the above-listed ingredients should be taken as merely exemplary, and not limiting, of the types of inactive ingredients that can be included in formulations described herein. The amounts of such inactive ingredients can be readily determined by one skilled in the art, according to the particular properties desired.

Liquid Oral Formulations

Liquid formulation dosage forms for oral administration can be aqueous suspensions selected from the group including, but not limited to, pharmaceutically acceptable aqueous oral dispersions, emulsions, solutions, elixirs, gels, and syrups. See, e.g., Singh et al., Encyclopedia of Pharmaceutical Technology, 2nd Ed., pp. 754-757 (2002). In addition to the particles of cromolyn or a pharmaceutically-acceptable salt thereof, the liquid dosage forms may include additives, such as: (a) disintegrating agents; (b) dispersing agents; (c) wetting agents; (d) at least one preservative, (e) viscosity enhancing agents, (f) at least one sweetening agent, and (g) at least one flavoring agent. In certain embodiments, the aqueous dispersions can further include a crystalline inhibitor. In certain embodiments, systemically effective amounts of cromolyn or a pharmaceutically-acceptable salt thereof are achieved with liquid oral formulations by including permeation enhancers in the liquid oral formulations.

Examples of disintegrating agents for use in the aqueous suspensions and dispersions include, but are not limited to, a starch, e.g., a natural starch such as corn starch or potato starch, a pregelatinized starch such as National 1551 or Amijel®, or sodium starch glycolate such as Promogel® or Explotab®; a cellulose such as a wood product, methylcrystalline cellulose, e.g., Avicel®, Avicel® PH101, Avicel® PH102, Avicel® PH105, Elcema® P100, Emcocel®, Vivacel®, Ming Tia®, and Solka-Floc®, methylcellulose, croscarmellose, or a cross-linked cellulose, such as cross-linked sodium carboxymethylcellulose (Ac-Di-Sol®), cross-linked carboxymethylcellulose, or cross-linked croscarmellose; a cross-linked starch such as sodium starch glycolate; a cross-linked polymer such as crospovidone; a cross-linked polyvinylpyrrolidone; alginate such as alginic acid or a salt of alginic acid such as sodium alginate; a clay such as Veegum® HV (magnesium aluminum silicate); a gum such as agar, guar, locust bean, Karaya, pectin, or tragacanth; sodium starch glycolate; bentonite; a natural sponge; a surfactant; a resin such as a cation-exchange resin; citrus pulp; sodium lauryl sulfate; sodium lauryl sulfate in combination starch; and the like.

In certain embodiments, the dispersing agents suitable for the aqueous suspensions and dispersions described herein include, for example, hydrophilic polymers, electrolytes, Tween® 60 or 80, PEG, polyvinylpyrrolidone (PVP; commercially known as Plasdone®), and the carbohydrate-based dispersing agents such as, for example, hydroxypropylcellulose and hydroxypropyl cellulose ethers (e.g., HPC, HPC-SL, and HPC-L), hydroxypropyl methylcellulose and hydroxypropyl methylcellulose ethers (e.g. HPMC K100, HPMC K4M, HPMC K15M, and HPMC K100M), carboxymethylcellulose sodium, methylcellulose, hydroxyethylcellulose, hydroxypropylmethyl-cellulose phthalate, hydroxypropylmethyl-cellulose acetate stearate, noncrystalline cellulose, magnesium aluminum silicate, triethanolamine, polyvinyl alcohol (PVA), polyvinylpyrrolidone/vinyl acetate copolymer (Plasdone®, e.g., S-630), 4-(1,1,3,3-tetramethylbutyl)-phenol polymer with ethylene oxide and formaldehyde (also known as tyloxapol), poloxamers (e.g., Pluronics F68®, F88®, and F108®, which are block copolymers of ethylene oxide and propylene oxide); and poloxamines (e.g., Tetronic 908®, also known as Poloxamine 908®, which is a tetrafunctional block copolymer derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine (BASF Corporation, Parsippany, N.J.)). In other embodiments, the dispersing agent is selected from a group not comprising one of the following agents: hydrophilic polymers; electrolytes; Tween® 60 or 80; PEG; polyvinylpyrrolidone (PVP); hydroxypropylcellulose and hydroxypropyl cellulose ethers (e.g., HPC, HPC-SL, and HPC-L); hydroxypropyl methylcellulose and hydroxypropyl methylcellulose ethers (e.g. HPMC K100, HPMC K4M, HPMC K15M, HPMC K100M, and Pharmacoat® USP 2910 (Shin-Etsu)); carboxymethylcellulose sodium; methylcellulose; hydroxyethylcellulose; hydroxypropylmethyl-cellulose phthalate; hydroxypropylmethyl-cellulose acetate stearate; non-crystalline cellulose; magnesium aluminum silicate; triethanolamine; polyvinyl alcohol (PVA); 4-(1,1,3,3-tetramethylbutyl)-phenol polymer with ethylene oxide and formaldehyde; poloxamers (e.g., Pluronics F68®, F88®, and F108®, which are block copolymers of ethylene oxide and propylene oxide); or poloxamines (e.g., Tetronic 908®, also known as Poloxamine 908®).

Wetting agents suitable for the aqueous suspensions and dispersions described herein include, but are not limited to, cetyl alcohol, glycerol monostearate, polyoxyethylene sorbitan fatty acid esters (e.g., the commercially available Tweens® such as e.g., Tween 20® and Tween 80® (ICI Specialty Chemicals)), and polyethylene glycols (e.g., Carbowaxs 3350® and 1450®, and Carbopol 934® (Union Carbide)), oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium oleate, sodium lauryl sulfate, sodium docusate, triacetin, vitamin E TPGS, sodium taurocholate, simethicone, phosphotidylcholine and the like.

Compositions and formulations of the disclosure may have a surface tension effective for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments, the compositions and formulations of the disclosure may have a surface tension in the range similar to that or water or higher. In certain embodiments, the compositions and formulations according to the present disclosure has a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. In some embodiments, the compositions and formulations has a surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

Compositions and formulations of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, compositions of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkoniumrn chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present compositions and formulations include any compound or agent that lowers the surface tension of a composition.

Inhalation Therapy

An “inhalation device,” as used herein, refers to any device that is capable of administering a drug formulation to the respiratory airways of a subject. Inhalation devices include conventional inhalation devices such as nebulizers, metered dose inhalers (MDIs), dry powder inhalers (DPIs), jet nebulizers, ultrasonic wave nebulizers, heat vaporizers, and soft mist inhalers. Inhalation devices also include nebulizers. Nebulizers, metered dose inhalers, and soft mist inhalers deliver pharmaceuticals by forming an aerosol which includes droplet sizes that can easily be inhaled. The aerosol can be used by a subject within the bounds of an inhalation therapy, whereby the cromolyn or a pharmaceutically-acceptable salt thereof reaches the subject's respiratory tract upon inhalation. In certain embodiments, the methods disclosed herein comprise administering to a subject a nominal dose of cromolyn or a pharmaceutically-acceptable salt thereof by an inhalation device, such as a nebulizer. In certain embodiments of the methods disclosed herein, an inhalation device is not a bronchoscope.

In certain embodiments of the methods disclosed herein, administration of a composition comprising cromolyn or a pharmaceutically acceptable salt thereof, e.g., cromolyn sodium, to a subject with an inhalation device, e.g., a nebulizer, a dry powder inhaler, a metered dose inhaler, a thermal aerosol inhaler, or an electrohydrodynamic-based solution misting inhaler, is effective for the treatment or prophylaxis of chronic cough due to IPF because both a systemically effective amount of the cromolyn or a pharmaceutically acceptable salt thereof and a high deposited lung dose of the cromolyn or a pharmaceutically acceptable salt thereof is achieved in the subject. Thus, in certain embodiments of the methods disclosed herein, administration of a composition comprising cromolyn or a pharmaceutically acceptable salt thereof, e.g., cromolyn sodium, to a subject with an inhalation device, e.g., a nebulizer, a dry powder inhaler, a metered dose inhaler, a thermal aerosol inhaler, or an electrohydrodynamic-based solution misting inhaler, is effective for the treatment or prophylaxis of chronic cough due to IPF that may not be believed to be susceptible to treatment or prophylaxis with cromolyn or a pharmaceutically acceptable salt thereof because both a systemically effective amount of the cromolyn or a pharmaceutically acceptable salt thereof and a high deposited lung dose of the cromolyn or a pharmaceutically acceptable salt thereof are achieved in the subject. Furthermore, in certain embodiments where cromolyn or a pharmaceutically acceptable salt thereof is administered with an inhalation device, e.g., a nebulizer, a dry powder inhaler, a metered dose inhaler, a thermal aerosol inhaler, or an electrohydrodynamic-based solution misting inhaler, the methods disclosed herein provide improved efficacy for the treatment or prophylaxis of chronic cough due to IPF relative to administration of a systemically effective amount of the cromolyn or a pharmaceutically acceptable salt thereof by a different route of administration, e.g., parenterally or orally, because administration of the cromolyn or a pharmaceutically acceptable salt thereof with an inhalation device, e.g., a nebulizer, a dry powder inhaler, a metered dose inhaler, a thermal aerosol inhaler, or an electrohydrodynamic-based solution misting inhaler, provides both a systemically effective amount of the cromolyn or a pharmaceutically acceptable salt thereof and a high deposited lung dose of the cromolyn or a pharmaceutically acceptable salt thereof in the subject. In certain embodiments, a systemically effective amount of cromolyn or a pharmaceutically acceptable salt thereof is achieved by delivering the cromolyn or a pharmaceutically acceptable salt thereof in an aerosol generated by a vibrating mesh nebulizer that produces droplets with a MMD of 3.0-4.0 μm and a GSD of 1.5-1.8. In certain embodiments of the methods disclosed herein, an aerosol is administered through a mouthpiece of a nebulizer using normal tidal breathing.

Characterization of Inhalation Devices

The efficiency of a particular inhalation device can be characterized in many different ways, including by pharmacokinetic properties, lung deposition (deposited lung dose), respirable dose (RD), delivered dose (DD), respirable fraction (RF), respirable drug delivery rate (RDDR), volumetric or mass median diameter (VMD or MMD), mass median aerodynamic diameter (MMAD) in combination with the geometric standard deviation (GSD), and total output rate (TOR), among others. The MMAD and GSD can be measured using a cascade impactor as described in United States Phamacopeia (USP<1601>). The DD can be measured by using breath simulation apparatus as described in USP<1601>.

The RF is derived from measuring the amount of drug deposited on the cascade impactor plates with a particular cut-off particle size, and expressing that as a fraction of the total amount deposited on the cascade impactor plates, the induction port and the filter. Thus, RF refers to a distribution of particles of an aerosol indicative of the percentages of the total mass or volume of the aerosol that are contained in particles of certain sizes. Such a mass/volume distribution expressed as the resipirable fraction (RF) is different from a distribution based on the percentages of the total number of particles in the aerosol that have certain particle sizes.

Since it takes fewer large particles to equal the mass or volume of a large number of small particles, a number distribution of particles contained in an aerosol can be markedly different from a mass distribution of the same particles in the same aerosol. As a simplified numerical example, consider an aerosol containing in total 9 particles: three 1 μm particles, three 2 μm particles, and three 3 μm particles, in size (diameter). Building a number distribution for these particles will generate a distribution where each particle size accounts for one third of the total. Yet, building a mass distribution for the same particles will generate a distribution, where 75% of the total mass/volume of the aerosol comes from the 3 μm particles, and less than 3% comes from the 1 μm particles. This is determined as follows: the volume of a spherical particle of diameter d is 4/3*π*(d/2)3. The volumes of the 1 μm, 2 μm and 3 μm particles would therefore be 0.52μ3, 4.2μ3, and 14.13μ3. Assuming unit density for all particles, these numbers would also represent the mass of the particles. Thus, the 3 μm particles would constitute 100*14.13/(0.52+4.2+14.13)=74.96% of the total mass. Likewise, the 2 μm particles will constitute 22.3% of the total mass and the 1 μm particles will be 2.8% of the total mass.

The RD is calculated by multiplying the DD by the RF. The TOR is measured by the difference in weight of a nebulizer before and after completion of nebulization divided by the duration of nebulization. VMD or MMD can be measured with a standard laser light scattering apparatus such as the Malvern Spraytec.

Pharmacokinetics is concerned with the uptake, distribution, metabolism and excretion of a drug substance in a subject. A pharmacokinetic profile comprises one or more biological measurements designed to measure the absorption, distribution, metabolism and excretion of a drug substance in a subject. One way of visualizing a pharmacokinetic profile is by means of a blood plasma concentration curve, which is a graph depicting mean active ingredient blood plasma concentration in a subject on the Y-axis and time (usually in hours) on the X-axis. Some pharmacokinetic parameters that may be visualized by means of a blood plasma concentration curve include AUClast, AUC(0-∞), Cmax, T1/2, and Tmax. An enhanced pharmacokinetic profile in a subject can be indicated by increased AUClast, AUC(0-∞), Cmax, or T1/2, a decreased Tmax, or an increased Tmax. Enhanced levels of cromolyn or a pharmaceutically-acceptable salt thereof in the blood plasma of a subject may result in better control of or improved symptoms of chronic cough due to IPF.

The deposited lung dose may be expressed as a percentage of the nominal dose that is deposited in the lung of a subject. For example, a lung deposition of 30% means 30% of the nominal dose is deposited in the lung of a subject. Likewise, a lung deposition of 60% means 60% of the nominal dose is deposited in the lung of a subject, and so forth. Lung deposition (deposited lung dose) can be determined using methods of scintigraphy or deconvolution.

Since cromolyn sodium is not metabolized in the body and approximately 50% of absorbed cromolyn is excreted in urine (Auty et al, Br. J Dis. Chest Vol. 81, No. 4, 1987, 371-380), and since its oral bioavailability is very low (˜1%), the deposited lung dose for cromolyn during inhalation can also be determined by measuring the cromolyn sodium content in the urine and multiplying that number by two.

RF, DD, RD, and RDDR are calculated parameters based on in vitro data that provide technical dimensions for the efficiency of an inhalation device. RF represents the percentage of the delivered aerosol, or inhaled mass, that penetrates into the gas-exchange region of the lungs. RF may be measured with a cascade impactor or laser diffraction apparatus. RF is expressed herein as the percentage of an aerosol delivered with an inhalation device that has a particular particle diameter or range of particle diameters. For example, the term “RF (≤3.3 μm)” as used herein refers to the percentage of an aerosol delivered with an inhalation device that has a particle diameter less than or equal to 3.3 μm. Similarly, the terms “RF (1-5 μm)” and “RF (≤5 μm)” as used herein refer to the percentage of an aerosol delivered with an inhalation device that has a particle diameter in the range of 1 μm to 5 μm, or less than 5 μm, respectively. DD is the portion or percentage of the nominal dose that is actually emitted from the mouthpiece of the device. The difference between the nominal dose and the DD is the amount of drug lost primarily as residues, i.e., the amount of drug remaining in the inhalation device after administration or lost in aerosol form during exhalation. RD is an expression of the delivered mass of drug contained within droplets or particles having a certain diameter emitted from an inhalation device, such as a DPI, MDI, or nebulizer, that are small enough to penetrate into the lung of a subject. The RD is determined by multiplying the DD by the RF. RDDR is the speed at which a respirable dose of the drug is delivered to a subject's lungs. RDDR, measured as a function of g/min or mg/min, is determined by dividing the RD by the amount of time necessary for inhalation. The amount of time necessary for inhalation is measured as the amount of time from the first moment of administration of the emitted droplet or powder from the nebulizer, DPI, or MDI until the emitted or delivered droplet or powder of a respirable diameter is delivered to the lung.

Aerosol particle/droplet size is one factor determining the deposition of aerosol drugs in the airways. The distribution of aerosol particle/droplet size can be expressed in terms of one or more of VMD/MMAD and GSD. GSD is a dimensionless measure of a droplet size distribution curve relevant for characterizing terms such as VMD, MMD, and MMAD. In general, the smaller the GSD for a particular particle size distribution, the narrower the distribution curve.

Inhalation Devices

Inhalation devices may be mechanical or electrical, and include, for example, jet nebulizers, and ultrasonic nebulizers. Jet nebulizers generally utilize compressors to generate compressed air, which breaks the liquid medication into small breathable droplets, which form an aerosolized (atomized) mist. In certain embodiments, when the subject breathes in, a valve at the top opens, which then allows air into the apparatus, thereby speeding up the mist generation; when the subject breathes out, the top valve closes, thereby slowing down the mist generation while simultaneously permitting the subject to breathe out through the opening of a mouthpiece flap. Some nebulizers may provide the aerosol in a continuous mode (e.g., the eFlow from PARI Pharma Stamberg), by a breath enhanced mode (e.g., the PARI LC Plus or Sprint from PARI Stamberg), by breath actuated mode dependent on the breathing pattern of the subject (e.g., the AeroEclipse from Trudell, Canada or the I-Neb from Philips Respironics), or according to given inhalation profile (e.g., the Akita from Activaero, Gmuenden, Germany).

Some conventional inhalation devices are disclosed in U.S. Pat. Nos. 6,513,727, 6,513,519, 6,176,237, 6,085,741, 6,000,394, 5,957,389, 5,740,966, 5,549,102, 5,461,695, 5,458,136, 5,312,046, 5,309,900, 5,280,784, and 4,496,086, each of which is hereby incorporated by reference in its entirety. Commercial conventional inhalation devices are available from: PARI (Germany) under the trade names PARI LC Plus®, LC Star®, and PARI-Jet®; A & H Products, Inc. (Tulsa, Okla.) under the trade name AquaTower®; Hudson RCI (Temecula, Calif.) under the trade name AVA-NEB®; Intersurgical, Inc. (Liverpool, N.Y.) under the trade name Cirrus®; Salter Labs (Arvin, Calif.) under the trade name Salter 8900®; Respironics (Murrysville, Pa.) under the trade name Sidestream®; Bunnell (Salt Lake City, Utah) under the trade name Whisper Jet®; Smiths-Medical (Hyth Kent, UK) under the trade name Downdraft®, and DeVilbiss (Somerset, Pa.) under the trade name DeVilbiss®; or Trudell, Canada under the trade name AeroEclipse®.

In certain embodiments of the methods disclosed herein, compositions comprising cromolyn or a pharmaceutically-acceptable salt thereof are administered with a dry powder inhaler. In certain embodiments of the methods disclosed herein, compositions administered with dry powder inhalers comprise one or more of nanoparticles, spray dried materials, engineered porous particles with low mass median diameter but a high geometric diameter, liposomes, and stealth (or PEGylated) liposomes. In certain embodiments, compositions administered by dry powder inhalers administered in the methods disclosed herein comprise nanoparticle clusters that aggregate into micrometer sized particles at neutral or basic pH but dissociate into nanoparticles at the pH encountered in the lung. In certain embodiments the nanoparticle clusters comprise fumaryl diketopiperazine. In certain embodiments, compositions administered with dry powder inhalers comprise lactose. In certain embodiments, compositions administered with dry powder inhalers do not comprise lactose. In certain embodiments, compositions administered with a dry powder inhaler have a MMAD between 2 and 4 μm, a GSD between 1.5 and 2.5 μm, and an RF (≤5 μm) between 30% and 80%. In certain embodiments, a dry powder inhaler used to administer an inhalation formulation in the methods disclosed herein comprises a pre-metered dose, such as Plastiape Monodose inhaler, which comprises a capsule pre-filled with a powder. In certain embodiments, a dry powder inhaler used to administer an inhalation formulation in the methods disclosed herein has a device-metered system such as Twisthaler, sold by Schering Plough, which comprises a reservoir to store a powder and a twisting top to dispense each dose. Inhalation formulations for administration with a dry powder inhaler may be prepared by blending cromolyn or a pharmaceutically acceptable salt thereof, e.g., cromolyn sodium, with lactose, or spray drying cromolyn or a pharmaceutically acceptable salt thereof, e.g., cromolyn sodium, or by pelletizing cromolyn or a pharmaceutically acceptable salt thereof, e.g., cromolyn sodium, to form free-flowing spherical agglomerates.

In certain embodiments of the methods disclosed herein, compositions comprising cromolyn or a pharmaceutically acceptable salt thereof are administered with a metered dose inhaler. In certain embodiments, a composition administered with a metered dose inhaler in the methods disclosed herein comprises one or more of nanoparticles, spray dried materials, engineered porous particles with low mass median diameter but a high geometric diameter, liposomes, and stealth (or PEGylated) liposomes.

In certain embodiments of the methods disclosed herein, compositions comprising cromolyn or a pharmaceutically acceptable salt thereof are administered with a thermal aerosol inhaler. In certain embodiments, the aerosol in a thermal aerosol inhaler is generated by directly heating and vaporizing a thin solid film of the cromolyn or a pharmaceutically acceptable salt thereof, e.g., cromolyn sodium, or by heating and vaporizing a solution of cromolyn or a pharmaceutically acceptable salt thereof, e.g., cromolyn sodium in solvents such as propylene glycol and/or glycerol and water.

In certain embodiments of the methods disclosed herein, compositions comprising cromolyn or a pharmaceutically acceptable salt thereof are administered with an electrohydrodynamic-based solution misting inhaler. In certain embodiments, the aerosol in the electrohydrodynamic-based solution-misting inhaler is generated by subjecting a solution of cromolyn or a pharmaceutically acceptable salt thereof, e.g., cromolyn sodium, or a liposome or pegylated liposome comprising cromolyn or a pharmaceutically acceptable salt thereof, e.g., cromolyn sodium, to electrohydrodynamic forces through electrostatic energy.

Nebulizers

Nebulizers are inhalation devices that comprise a micro-perforated membrane through which a liquid solution is converted through electrical or mechanical means into aerosol droplets suitable for inhalation. Nebulizers can deliver a large fraction of a loaded dose to a subject. In certain embodiments, the nebulizer also utilizes one or more actively or passively vibrating microperforated membranes. In certain embodiments, the nebulizer contains one or more oscillating membranes. In certain embodiments, the nebulizer contains a vibrating mesh or plate with multiple apertures and optionally a vibration generator with an aerosol mixing chamber. In some such embodiments, the mixing chamber functions to collect (or stage) the aerosol from the aerosol generator. In certain embodiments, an inhalation valve is also used to allow an inflow of ambient air into the mixing chamber during an inhalation phase and is closed to prevent escape of the aerosol from the mixing chamber during an exhalation phase. In some such embodiments, the exhalation valve is arranged at a mouthpiece which is removably mounted at the mixing chamber and through which the subject inhales the aerosol from the mixing chamber. Still yet, in certain embodiments, the nebulizer contains a pulsating membrane. In certain embodiments, the nebulizer is continuously operating.

In certain embodiments, the nebulizer contains a vibrating micro-perforated membrane of tapered nozzles that generates a plume of droplets without the need for compressed gas. In these embodiments, a solution in the micro-perforated membrane nebulizer is in contact with a membrane, the opposite side of which is open to the air. The membrane is perforated by a large number of nozzle orifices of an atomizing head. An aerosol is created when alternating acoustic pressure in the solution is built up in the vicinity of the membrane causing the fluid on the liquid side of the membrane to be emitted through the nozzles as uniformly sized droplets.

Certain embodiments of nebulizers use passive nozzle membranes and a separate piezoelectric transducer that stimulates the membrane. In contrast, some nebulizers employ an active nozzle membrane, which use the acoustic pressure in the nebulizer to generate very fine droplets of solution via the high frequency vibration of the nozzle membrane.

Some nebulizers contain a resonant system. In some such nebulizers, the membrane is driven by a frequency for which the amplitude of the vibrational movement at the center of the membrane is particularly large, resulting in a focused acoustic pressure in the vicinity of the nozzle; the resonant frequency may be about 100 kHz. A flexible mounting is used to keep unwanted loss of vibrational energy to the mechanical surroundings of the atomizing head to a minimum. In certain embodiments, the vibrating membrane of the nebulizer may be made stainless steel, or of a nickel-palladium alloy by electroforming.

In certain embodiments, a nebulizer may be adapted or adaptable to operate in conjunction with a unit dosage form, such as an ampule or vial, which contains a single dose of composition comprising cromolyn or a pharmaceutically-acceptable salt thereof for the treatment of chronic cough due to IPF. The unit dosage form comprises a container that contains an inhalation formulation comprising the cromolyn or a pharmaceutically-acceptable salt thereof, such as cromolyn sodium. The container is adapted to cooperate with the nebulizer device in such a way as to permit administration of the nominal dose of the inhalation formulation to a subject. In certain embodiments, the nebulizer and the unit dosage form are configured so that they are useable together, but not with other devices or dosage forms. In some particular embodiments, the unit dosage form is configured such that it fits into a keyhole-like structure in the nebulizer, but will not operate with other nebulizer devices. In such embodiments, the nebulizer is configured such that it will accept and properly operate with the unit dosage form containing the cromolyn or a pharmaceutically-acceptable salt thereof, but not with other dosage forms.

Commercial high efficiency nebulizers are available from: PARI (Germany) under the trade name eFlow®; Aerogen, Ltd. (Ireland) under the trade names AeroNeb® Go and AeroNeb® Pro, AeroNeb® Solo, and other nebulizers utilizing the OnQ® nebulizer technology; Respironics (Murrysville, Calif.) under the trade names I-Neb®; Omron (Bannockbum, Ill.) under the trade name Micro-Air®; Activaero (Germany) under the trade name Akita®, and AerovectRx (Atlanta, Ga.) under the trade name AerovectRx®.

In certain embodiments, the methods disclosed herein comprise administration to a subject a nominal dose of cromolyn or a pharmaceutically-acceptable salt thereof with a nebulizer, wherein administration of the nominal dose of the cromolyn or a pharmaceutically-acceptable salt thereof to the subject provides one or more of the following advantages: (1) an enhanced pharmacokinetic profile as compared to administration of an oral solution; (2) an enhanced therapeutic effect as compared to administration of an oral solution; (3) an enhanced lung deposition (deposited lung dose) as compared with some inhalation devices used with other cromolyn sodium compositions evidenced by scintigraphy or deconvolution, or derived from suitable in vitro indicators such as enhanced RD, RDDR, RF, and lower GSDs, as compared to administration with some inhalation devices used with other cromolyn sodium compositions; (4) reduced administration times, periods, and/or volumes as compared to administration with some other formulations and inhalation devices; (5) a reduction in adverse side effects associated with oral formulations of cromolyn or a pharmaceutically-acceptable salt thereof, such as gastrointestinal irritation; and (6) a longer duration of therapeutic effect as compared to administration of an oral solution or an inhaled formulations using other formulations of cromolyn sodium with other inhalation devices.

In certain embodiments, the DD expressed as the percentage of the nominal dose of cromolyn or a pharmaceutically-acceptable salt thereof administered with a nebulizer in the methods disclosed herein is at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, about 65%, about 70%, about 30% to about 90%, about 40% to about 80%, about 45% to about 75%, about 50% to about 70%, about 30% to about 75%, about 40% to about 70%, about 45% to about 60%, or about 60% to about 70%.

TOR is the speed at which the liquid containing cromolyn or a pharmaceutically-acceptable salt thereof is administered from the inhalation device. In certain embodiments, administration of the cromolyn or a pharmaceutically-acceptable salt thereof with the nebulizer provides a TOR of at least about 2 times, 3 times or 4 times the TOR achievable with a conventional inhalation device, such as a nebulizer. For example, in certain embodiments the TOR is at least about at least about 150 mg/min, at least about 200 mg/min, at least about 250 mg/min, at least 300 mg/min, at least 350 mg/min, at least 400 mg/min, at least 500 mg/min, or from 200 to about 700 mg/min.

In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution comprising cromolyn or a pharmaceutically-acceptable salt thereof, wherein the aerosol exhibits an RF (≤3.3 μm) of at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, about 20% to about 95%, about 35% to about 90%, about 40% to about 80%, about 40% to about 90%, about 40% to about 95%, about 45% to about 90%, about 45% to about 95%, about 50% to about 90%, about 65% to about 90%, about 60% to about 95%, about 65% to about 95%, about 70% to about 90%, or about 55% to about 90%. In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution comprising cromolyn or a pharmaceutically-acceptable salt thereof, wherein the aerosol exhibits an RF (≤3.3 μm) of cromolyn sodium of at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, about 20% to about 95%, about 35% to about 90%, or about 40% to about 80%, about 40% to about 90%, about 40% to about 95%, about 45% to about 90%, about 45% to about 95%, about 50% to about 90%, about 65% to about 90%, about 60% to about 95%, about 65% to about 95%, about 70% to about 90%, about 55% to about 90%, about 40% to about 50%, about 35% to about 45%, about 35% to about 50%, about 30% to about 50%, about 44%, or about 36%. In certain embodiments, the solution comprises an osmotic agent comprising sodium chloride. In certain embodiments, the solution comprises an osmotic agent consisting of sodium chloride. In certain embodiments, the osmolality of the solution is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the solution is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments, the solution of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, the solution of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkoniumrn chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present solution include any compound or agent that lowers the surface tension of a composition.

In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol that comprises particles having a surface tension suitable for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution that comprises particles having a surface tension in the range similar to that or water or higher. In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution that comprises particles having a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. In some embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution that comprises particles having a surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution comprising cromolyn or a pharmaceutically-acceptable salt thereof, wherein the aerosol exhibits an RF (1-5 μm) of cromolyn or a pharmaceutically-acceptable salt thereof of at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, about 20% to about 95%, about 35% to about 90%, or about 40% to about 80%, about 40% to about 90%, about 40% to about 95%, about 45% to about 90%, about 45% to about 95%, about 50% to about 90%, about 65% to about 90%, about 60% to about 95%, about 65% to about 95%, about 70% to about 90%, or about 55% to about 90%. In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution comprising cromolyn or a pharmaceutically-acceptable salt thereof, wherein the aerosol exhibits an RF (1-5 μm) of cromolyn sodium of at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, about 20% to about 95%, about 35% to about 90%, or about 40% to about 80%, about 40% to about 90%, about 40% to about 95%, about 45% to about 90%, about 45% to about 95%, about 50% to about 90%, about 65% to about 90%, about 60% to about 95%, about 65% to about 95%, about 70% to about 90%, or about 55% to about 90%. In certain embodiments, the solution comprises an osmotic agent comprising sodium chloride. In certain embodiments, the solution comprises an osmotic agent consisting of sodium chloride. In certain embodiments, the solution comprises an osmotic agent comprising sodium chloride. In certain embodiments, the solution comprises an osmotic agent consisting of sodium chloride. In certain embodiments, the osmolality of the solution is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the solution is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments, the solution of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, the solution of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkoniumrn chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present solution include any compound or agent that lowers the surface tension of a composition.

In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol that comprises particles having a surface tension suitable for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution that comprises particles having a surface tension in the range similar to that or water or higher. In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution that comprises particles having a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. In some embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution that comprises particles having a surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution comprising cromolyn or a pharmaceutically-acceptable salt thereof, wherein the aerosol exhibits an RF (≤5 μm) of an cromolyn or a pharmaceutically-acceptable salt thereof of at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, about 20% to about 95%, about 35% to about 90%, about 40% to about 80%, about 40% to about 90%, about 40% to about 95%, about 45% to about 90%, about 45% to about 95%, about 50% to about 90%, about 65% to about 90%, about 60% to about 95%, about 65% to about 95%, about 70% to about 90%, about 55% to about 90%, about 70% to about 80%, or about 75%. In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution comprising cromolyn or a pharmaceutically-acceptable salt thereof, wherein the aerosol exhibits an RF (≤5 μm) of cromolyn sodium of at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, about 20% to about 95%, about 35% to about 90%, about 40% to about 80%, about 40% to about 90%, about 40% to about 95%, about 45% to about 90%, about 45% to about 95%, about 50% to about 90%, about 65% to about 90%, about 60% to about 95%, about 65% to about 95%, about 70% to about 90%, about 55% to about 90%, about 70% to about 80%, about 65% to about 75%, about 65% to about 80%, about 60% to about 80%, about 66%, or about 75%. In certain embodiments, the solution comprises an osmotic agent comprising sodium chloride. In certain embodiments, the solution comprises an osmotic agent consisting of sodium chloride. In certain embodiments, the osmolality of the solution is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the solution is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments, the solution of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, the solution of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Spank (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkoniumrn chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present solution include any compound or agent that lowers the surface tension of a composition.

In certain embodiments, the aerosols provided by the present methods or nebulizer comprise particles having a surface tension suitable for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments, the aerosols provided by the present methods or nebulizer comprise particles having surface tension in the range similar to that or water or higher. In certain embodiments, the aerosols provided by the present methods or nebulizer comprise particles having a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. In some embodiments, the aerosols provided by the present methods or nebulizer comprise particles having surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

The disclosure provides a pharmaceutically acceptable solution, comprising from about 1% to about 10% by weight of cromolyn sodium an osmotic agent, wherein the osmotic agent consists of sodium chloride, wherein an aerosol created from the pharmaceutically acceptable solution is suitable for inhalation by a subject having chronic cough due to idiopathic pulmonary fibrosis. In certain embodiments, the aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30%. In certain embodiments, the aerosol has a respirable fraction (≤3.3 μm) as measured by USP <1601> of at least about 30% and a respirable fraction (≤5 μm) as measured by USP <1601> of at least about 75%. In certain embodiments, the pharmaceutically acceptable solution comprises about 1%, or about 2%, or about 3%, or about 4%, or about 5%, or about 6%, or about 7%, or about 8%, or about 9%, or about 10% by weight of cromolyn sodium. In certain embodiments, the osmostic agent comprises an ionic osmotic agent and excludes any non-ionic osmotic agent. In certain embodiments, the osmotic agent consists of sodium chloride. In certain embodiments, the osmolality of the solution is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the solution is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments, the pharmaceutically acceptable solution of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, the pharmaceutically acceptable solution of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkonium chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present pharmaceutically acceptable solution include any compound or agent that lowers the surface tension of a composition.

In certain embodiments, the pharmaceutically acceptable solution has a surface tension suitable for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments the pharmaceutically acceptable solution has a surface tension in the range similar to that or water or higher. the pharmaceutically acceptable solution has a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. the pharmaceutically acceptable solution has a surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

The disclosure provides any of the methods or pharmaceutically acceptable solutions disclosed herein wherein the pharmaceutically acceptable solution has an osmolality of less than about 250 mOsm/kg, or less than about 225 mOsm/kg, or less than about 200 mOsm/kg, or less than about 190 mOsm/kg, or less than about 180 mOsm/kg, or less than about 175 mOsm/kg, or less than about 170 mOsm/kg, or less than about 165 mOsm/kg, or less than about 160 mOsm/kg, or less than about 155 mOsm/kg, or less than about 150 mOsm/kg, or less than about 145 mOsm/kg, or less than about 140 mOsm/kg, or less than about 135 mOsm/kg, or less than about 130 mOsm/kg, or less than about 125 mOsm/kg, or less than about 120 mOsm/kg, or less than about 115 mOsm/kg, or less than about 110 mOsm/kg, or less than about 105 mOsm/kg, or less than about 100 mOsm/kg. In certain embodiments, the pharmaceutically acceptable solution has an osmolality of between about 70 mOsm/kg and about 200 mOsm/kg, or between about 70 mOsm/kg and about 190 mOsm/kg, or between about 70 and about 180 mOsm/kg, or between about 70 mOsm/kg and about 170 mOsm/kg, or between about 70 mOsm/kg and about 160 mOsm/kg, or between about 70 mOsm/kg and about 150 mOsm/kg, or between about 70 mOsm/kg and about 140 mOsm/kg, or between about 80 mOsm/kg and about 200 mOsm/kg, or between about 80 mOsm/kg and about 190 mOsm/kg, or between about 90 mOsm/kg and about 180 mOsm/kg, or between about 90 mOsm/kg and about 175 mOsm/kg, or between about 90 mOsm/kg and about 170 mOsm/kg, or between about 90 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 110 mOsm/kg and about 145 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 110 mOsm/kg and about 135 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg. In certain embodiments, the osmolality of the pharmaceutically acceptable solution is about 100 mOsm/kg, or about 110 mOsm/kg, or about 115 mOsm/kg, or about 120 mOsm/kg, or about 125 mOsm/kg, or about 130 mOsm/kg, or about 135 mOsm/kg, or about 140 mOsm/kg, or about 145 mOsm/kg, or about 150 mOsm/kg, or about 155 mOsm/kg, or about 160 mOsm/kg, or about 165 mOsm/kg, or about 170 mOsm/kg, or about 175 mOsm/kg, or about 180 mOsm/kg, or about 185 mOsm/kg, or about 190 mOsm/kg, or about 195 mOsm/kg, or about 200 mOsm/kg.

The disclosure provides any of the methods or pharmaceutically acceptable solutions disclosed herein wherein the pharmaceutically acceptable solution may exclude, or, may not comprise a surfactant. In certain embodiments, the pharmaceutically acceptable solution of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkoniumrn chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present pharmaceutically acceptable solution include any compound or agent that lowers the surface tension of a composition.

The disclosure provides any of the methods or pharmaceutically acceptable solutions disclosed herein wherein the pharmaceutically acceptable solution has a surface tension suitable for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments, the pharmaceutically acceptable solution has a surface tension in the range similar to that or water or higher. the pharmaceutically acceptable solution has a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. In some embodiments, the pharmaceutically acceptable solution has a surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

The disclosure provides a pharmaceutically acceptable solution, comprising from about 2% to about 6% by weight of cromolyn sodium and an osmolarity adjusting agent consisting of sodium chloride, wherein an aerosol created from the pharmaceutically acceptable solution is suitable for inhalation by a subject in need thereof. In certain embodiments, the aerosol exhibits an RF (≤3.3 μm) as measured by USP <1601> of at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%. In certain embodiments, the aerosol exhibits and RF (≤5 μm) as measured by USP <1601> of at least about 30%, or at least about 35%, or at least about 40%, or at least about 45%, or at least about 50%, or at least about 55%, or at least about 60%, or at least about 65%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%.

In certain embodiments, the pharmaceutically acceptable solution of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, the pharmaceutically acceptable solution of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkonium chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present pharmaceutically acceptable solution include any compound or agent that lowers the surface tension of a composition.

In certain embodiments, the pharmaceutically acceptable solution has a surface tension suitable for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments the pharmaceutically acceptable solution has a surface tension in the range similar to that or water or higher. the pharmaceutically acceptable solution has a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. the pharmaceutically acceptable solution has a surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

The disclosure provides a pharmaceutically acceptable aerosol, comprising droplets of a solution comprising from about 1% to about 10% by weight of cromolyn sodium and an osmolarity adjusting agent consisting of sodium chloride. In certain embodiments, the aerosol exhibits an RF (≤3.3 μm) as measured by USP <1601> of at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%. In certain embodiments, the aerosol exhibits and RF (≤5 μm) as measured by USP <1601> of at least about 30%, or at least about 35%, or at least about 40%, or at least about 45%, or at least about 50%, or at least about 55%, or at least about 60%, or at least about 65%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%. In certain embodiments, the osmolality of the solution is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the solution is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments, the pharmaceutically acceptable solution of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, the pharmaceutically acceptable solution of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkoniumrn chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present pharmaceutically acceptable solution include any compound or agent that lowers the surface tension of a composition.

In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension suitable for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension in the range similar to that or water or higher. In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. In some embodiments, In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

The disclosure provides a method of administering a therapeutically effective amount of cromolyn sodium to a subject in need thereof, comprising administering to the subject a pharmaceutical composition comprising from about 2% to about 6% by weight of cromolyn sodium and an osmotic agent consisting of sodium chloride, wherein the pharmaceutical composition is administered to the subject by inhalation in the form of an aerosol exhibiting an RF (≤5 μm) as measured by USP <1601> of at least about 60%. In certain embodiments, the aerosol exhibits an RF (≤3.3 μm) as measured by USP <1601> of at least about 30%. In certain embodiments, the aerosol exhibits an RF (≤3.3 rpm) as measured by USP <1601> of at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%. In certain embodiments, the aerosol exhibits and RF (≤5 μm) as measured by USP <1601> of at least about 30%, or at least about 35%, or at least about 40%, or at least about 45%, or at least about 50%, or at least about 55%, or at least about 60%, or at least about 65%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%. In certain embodiments, the osmolality of the pharmaceutical composition is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the pharmaceutical composition is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments, the pharmaceutically acceptable solution or composition of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, the pharmaceutically acceptable solution of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkoniumrn chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present pharmaceutically acceptable solution include any compound or agent that lowers the surface tension of a composition.

In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension suitable for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension in the range similar to that or water or higher. In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. In some embodiments, In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

The disclosure provides a dosage form comprising: (a) a pharmaceutical composition comprising from about 2% to about 6% by weight of cromolyn sodium, and an osmolarity adjusting agent consisting of sodium chloride; and (b) an inhalation device that forms an aerosol of the pharmaceutical composition, the aerosol exhibiting a respirable fraction of the pharmaceutical composition (≤5 μm) as measured by USP <1601> of at least about 60%. In certain embodiments, the aerosol exhibits an RF (≤3.3 μm) as measured by USP <1601> of at least about 30%. In certain embodiments, the aerosol exhibits an RF (≤3.3 μm) as measured by USP <1601> of at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%. In certain embodiments, the aerosol exhibits and RF (≤5 μm) as measured by USP <1601> of at least about 30%, or at least about 35%, or at least about 40%, or at least about 45%, or at least about 50%, or at least about 55%, or at least about 60%, or at least about 65%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%. In certain embodiments, the osmolality of the pharmaceutical composition is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the pharmaceutical composition is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments, the pharmaceutically acceptable dosage form of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, the pharmaceutically acceptable solution of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkoniumrn chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present pharmaceutically acceptable solution include any compound or agent that lowers the surface tension of a composition.

In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension suitable for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension in the range similar to that or water or higher. In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. In some embodiments, In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m.

The disclosure provides a dosage form comprising: (a) a pharmaceutical composition comprising from about 2% to about 6% by weight of cromolyn sodium, and an osmolarity adjusting agent consisting of sodium chloride; and (b) a means for producing an aerosol of the pharmaceutical composition, the aerosol exhibiting a respirable fraction of the pharmaceutical composition (≤5 μm) as measured by USP <1601> of at least about 60%. In certain embodiments, the aerosol exhibits an RF (≤3.3 μm) as measured by USP <1601> of at least about 30%. In certain embodiments, the aerosol exhibits an RF (≤3.3 μm) as measured by USP <1601> of at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%. In certain embodiments, the aerosol exhibits and RF (≤5 μm) as measured by USP <1601> of at least about 30%, or at least about 35%, or at least about 40%, or at least about 45%, or at least about 50%, or at least about 55%, or at least about 60%, or at least about 65%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%. In certain embodiments, the osmolality of the pharmaceutical composition is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the pharmaceutical composition is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments, the pharmaceutically acceptable dosage form of the disclosure may exclude, or, may not comprise a surfactant. In certain embodiments, the pharmaceutically acceptable solution of the disclosure may not comprise any dispersing agent, solubilizing agent, or spreading agent. Some examples of surfactants that are excluded from the present compositions and formulations include: PEG (polyethylene glycol) 400; Sodium lauryl sulfate sorbitan laurate, sorbitan palmitate, sorbitan stearate available under the tradename Span® (20-40-60 etc.); polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (20) sorbitan monopalmitate, polyoxyethylene (20) sorbitan monostearate available under the tradename Tween (polysorbates, 20-40-60 etc.); tyloxapol; propylene glycol; and Benzalkonium chloride, vitamin-TPGS and lecithins, (Exosurf®, GlaxoSmithKline), surfactant proteins. In certain embodiments, surfactants that are excluded from the present pharmaceutically acceptable solution include any compound or agent that lowers the surface tension of a composition.

In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension suitable for deposition, penetration or retention of the composition primarily in the peripheral lung regions, including the bronchioles and alveoli. In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension in the range similar to that or water or higher. In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension of at least about 30 mN/m, or at least about 40 mN/m, or at least about 50 mN/m, or at least about 60 mN/m, or at least about 70 mN/m. In some embodiments, In certain embodiments, droplets of the pharmaceutically acceptable aerosols have a surface tension in the range of about 30 mN/m to about 75 mN/m, or about 50 mN/m to about 75 mN/m, or about 70 mN/m to about 75 mN/m. In certain embodiments, use of a nebulizer in the methods disclosed herein provides a RDDR of at least about 2 times, at least about 3 times or at least about 4 times the RDDR achievable with a conventional inhalation device. For example, where the cromolyn or a pharmaceutically-acceptable salt thereof is cromolyn sodium, in certain embodiments the RDDR is at least about 5 mg/min, at least about 10 mg/min, at least about 15 mg/min, at least about 20 mg/min, at least about 25 mg/min, at least about 30 mg/min, at least about 35 mg/min, at least about 40 mg/min, at least about 45 mg/min, at least about 50 mg/min, at least about 55 mg/min, or at least about 60 mg/min.

In certain embodiments, administration of cromolyn or a pharmaceutically-acceptable salt thereof with a nebulizer in the methods disclosed herein provides a GSD of emitted droplet size distribution of about 1.1 to about 2.1, about 1.2 to about 2.0, about 1.3 to about 1.9, less than about 2, at least about 1.4 to about 1.8, at least about 1.5 to about 1.7, about 1.4, about 1.5, about 1.6, or about 1.7. In certain embodiments, administration of cromolyn or a pharmaceutically-acceptable salt thereof with a nebulizer in the methods disclosed herein provides a MMAD of droplet size of about 1 μm to about 5 m, about 2 to about 4 m, about 3 to about 4 m, about 3.5 to about 4.5 m, or about 3.5 μm. In some particular embodiments, administration of cromolyn or a pharmaceutically-acceptable salt thereof in the methods disclosed herein provides droplets having a particular combination of MMAD and GSD, for example: an MMAD of less than about 5 μm and a GSD of about 1.1 to about 2.1; an MMAD of less than about 4.5 μm and a GSD of about 1.1 to about 2.1; an MMAD of about 1 μm to about 5 μm and a GSD of about 1.1 to about 2.1; an MMAD of about 1.5 to about 4.5 μm and a GSD of about 1.1 to about 2.1; an MMAD of less than about 5 μm and a GSD of about 1.1 to about 2.0; an MMAD of less than about 4.5 μm and a GSD of about 1.1 to about 2.0; an MMAD of about 1 μm to about 5 μm and a GSD of about 1.1 to about 2.0; an MMAD of about 1.5 to about 4.5 μm and a GSD of about 1.1 to about 2.0; an MMAD of less than about 5 μm and a GSD of about 1.1 to about 1.9; an MMAD of less than about 4.5 μm and a GSD of about 1.1 to about 1.9; an MMAD of about 1 μm to about 5 μm and a GSD of about 1.1 to about 1.9; an MMAD of about 1.5 to about 4.5 μm and a GSD of about 1.1 to about 1.9; an MMAD of less than about 5 μm and a GSD of about 1.1 to about 1.8; an MMAD of less than about 4.5 μm and a GSD of about 1.1 to about 1.8; an MMAD of about 1 μm to about 5 μm and a GSD of about 1.1 to about 1.8; an MMAD of about 1.5 to about 4.5 μm and a GSD of about 1.1 to about 1.8; an MMAD of about 3.5 μm or less and a GSD of about 1.7; an MMAD of about 4.1 μm or less and a GSD of about 1.7; an MMAD of about 3.5 μm and a GSD of about 1.7; or an MMAD of about 4.1 μm and a GSD of about 1.7.

In certain embodiments, the median particle size of cromolyn or a pharmaceutically-acceptable salt thereof aerosol administered with a nebulizer is between about 1 μm and about 6 μm, between about 2 μm and about 5 μm, between about 3 μm and about 5 m, between about 3 μm and about 4 m, about 1 m, about 2 μm, about 3 m, about 4 μm, about 5 μm, or about 6 μm. In certain embodiments, the median particle size of cromolyn sodium aerosol administered with a nebulizer is between about 1 μm and about 6 m, between about 2 μm and about 5 m, between about 3 μm and about 5 m, between about 3 μm and about 4 m, about 1 m, about 2 μm, about 3 m, about 4 m, about 5 m, or about 6 μm.

Inhalation Formulations

In certain embodiments disclosed herein are provided formulations comprising from about 2% to about 10% by weight cromolyn sodium and an osmotic agent consisting of sodium chloride, wherein the formulation is stable when stored at 25° C. for at least 4 weeks, or at least 6 weeks, or at least 8 weeks, or at least 10 weeks, or at least 12 weeks, or at least 6 months, or at least 8 months, or at least 10 months, or at least 12 months, or at least 14 months, or at least 16 months, or at least 18 months, or at least 20 months, or at least 24 months. In certain embodiments, the formulations remain clear solutions when stored at 25° C. for these same time periods. In certain embodiments, the formulations exhibit less than 1% by weight total impurities when stored at 25° C. for these same time periods. In certain embodiments, the formulation comprises about 4% by weight of cromolyn sodium. In certain embodiments, the formulation comprises about 6% by weight of cromolyn sodium. In certain embodiments, the osmolality of the formulation is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the formulation is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodimentsdisclosed herein are provided formulations comprising from about 2% to about 10% by weight cromolyn sodium and an osmotic agent consisting of sodium chloride, wherein the formulation is stable when stored at 40° C. for at least 4 weeks, or at least 6 weeks, or at least 8 weeks, or at least 10 weeks, or at least 12 weeks, or at least 6 months, or at least 8 months, or at least 10 months, or at least 12 months, or at least 14 months, or at least 16 months, or at least 18 months, or at least 20 months, or at least 24 months. In certain embodiments, the formulations remain clear solutions when stored at 40° C. for these same time periods. In certain embodiments, the formulations exhibit less than 1% by weight total impurities when stored at 40° C. for these same time periods. In certain embodiments, the formulation comprises about 4% by weight of cromolyn sodium. In certain embodiments, the formulation comprises about 6% by weight of cromolyn sodium In certain embodiments, the osmolality of the formulation is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the formulation is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments of the methods disclosed herein, inhalation formulations are administered by an inhalation device, e.g., a nebulizer, to provide a systemically effective amount of cromolyn or a pharmaceutically-acceptable salt thereof for the treatment of chronic cough due to IPF. In certain embodiments, the methods disclosed herein comprise administering a nominal dose of cromolyn or a pharmaceutically-acceptable salt thereof in an aqueous inhalation solution to the subject with an inhalation device, e.g., a nebulizer.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 100 ng*hr/mL, greater than about 110 ng*hr/mL, greater than about 120 ng*hr/mL, greater than about 130 ng*hr/mL, greater than about 140 ng*hr/mL, greater than about 150 ng*hr/mL, greater than about 160 ng*hr/mL, greater than about 170 ng*hr/mL, greater than about 180 ng*hr/mL, greater than about 190 ng*hr/mL, greater than about 200 ng*hr/mL, greater than about 225 ng*hr/mL, greater than about 250 ng*hr/mL, greater than about 275 ng*hr/mL, greater than about 300 ng*hr/mL, greater than about 325 ng*hr/mL, greater than about 350 ng*hr/mL, greater than about 375 ng*hr/mL, greater than about 400 ng*hr/mL, greater than about 425 ng*hr/mL, greater than about 450 ng*hr/mL, greater than about 475 ng*hr/mL, greater than about 500 ng*hr/mL, greater than about 525 ng*hr/mL, greater than about 550 ng*hr/mL, greater than about 575 ng*hr/mL, greater than about 600 ng*hr/mL, greater than about 625 ng*hr/mL, greater than about 650 ng*hr/mL, greater than about 675 ng*hr/mL, greater than about 700 ng*hr/mL, greater than about 725 ng*hr/mL, greater than about 750 ng*hr/mL, greater than about 775 ng*hr/mL, greater than about 800 ng*hr/mL, greater than about 825 ng*hr/mL, greater than about 850 ng*hr/mL, greater than about 875 ng*hr/mL, greater than about 900 ng*hr/mL, greater than about 925 ng*hr/mL, greater than about 950 ng*hr/mL, greater than about 975 ng*hr/mL, or greater than about 1000 ng*hr/mL after administration of the formulation to the subject. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 100 ng*hr/mL, greater than about 110 ng*hr/mL, greater than about 120 ng*hr/mL, greater than about 130 ng*hr/mL, greater than about 140 ng*hr/mL, greater than about 150 ng*hr/mL, greater than about 160 ng*hr/mL, greater than about 170 ng*hr/mL, greater than about 180 ng*hr/mL, greater than about 190 ng*hr/mL, greater than about 200 ng*hr/mL, greater than about 225 ng*hr/mL, greater than about 250 ng*hr/mL, greater than about 275 ng*hr/mL, greater than about 300 ng*hr/mL, greater than about 325 ng*hr/mL, greater than about 350 ng*hr/mL, greater than about 375 ng*hr/mL, greater than about 400 ng*hr/mL, greater than about 425 ng*hr/mL, greater than about 450 ng*hr/mL, greater than about 475 ng*hr/mL, greater than about 500 ng*hr/mL, greater than about 525 ng*hr/mL, greater than about 550 ng*hr/mL, greater than about 575 ng*hr/mL, greater than about 600 ng*hr/mL, greater than about 625 ng*hr/mL, greater than about 650 ng*hr/mL, greater than about 675 ng*hr/mL, greater than about 700 ng*hr/mL, greater than about 725 ng*hr/mL, greater than about 750 ng*hr/mL, greater than about 775 ng*hr/mL, greater than about 800 ng*hr/mL, greater than about 825 ng*hr/mL, greater than about 850 ng*hr/mL, greater than about 875 ng*hr/mL, greater than about 900 ng*hr/mL, greater than about 925 ng*hr/mL, greater than about 950 ng*hr/mL, greater than about 975 ng*hr/mL, or greater than about 1000 ng*hr/mL after administration of the formulation to the subject.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof of about 100 ng*hr/mL, about 110 ng*hr/mL, about 120 ng*hr/mL, about 130 ng*hr/mL, about 140 ng*hr/mL, about 150 ng*hr/mL, about 160 ng*hr/mL, about 170 ng*hr/mL, about 180 ng*hr/mL, about 190 ng*hr/mL, about 200 ng*hr/mL, about 225 ng*hr/mL, about 250 ng*hr/mL, ng*hr/mL, about 275 ng*hr/mL, about 300 ng*hr/mL, about 325 ng*hr/mL, about 350 ng*hr/mL, about 375 ng*hr/mL, about 400 ng*hr/mL, about 425 ng*hr/mL, about 450 ng*hr/mL, about 475 ng*hr/mL, about 500 ng*hr/mL, about 525 ng*hr/mL, about 550 ng*hr/mL, about 575 ng*hr/mL, about 600 ng*hr/mL, about 625 ng*hr/mL, about 650 ng*hr/mL, about 675 ng*hr/mL, about 700 ng*hr/mL, about 725 ng*hr/mL, about 750 ng*hr/mL, about 775 ng*hr/mL, about 800 ng*hr/mL, about 825 ng*hr/mL, about 850 ng*hr/mL, about 875 ng*hr/mL, about 900 ng*hr/mL, about 925 ng*hr/mL, about 950 ng*hr/mL, about 975 ng*hr/mL, or about 1000 ng*hr/mL after administration of the formulation to the subject. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof of about 100 ng*hr/mL, about 110 ng*hr/mL, about 120 ng*hr/mL, about 130 ng*hr/mL, about 140 ng*hr/mL, about 150 ng*hr/mL, about 160 ng*hr/mL, about 170 ng*hr/mL, about 180 ng*hr/mL, about 190 ng*hr/mL, about 200 ng*hr/mL, about 225 ng*hr/mL, about 250 ng*hr/mL, ng*hr/mL, about 275 ng*hr/mL, about 300 ng*hr/mL, about 325 ng*hr/mL, about 350 ng*hr/mL, about 375 ng*hr/mL, about 400 ng*hr/mL, about 425 ng*hr/mL, about 450 ng*hr/mL, about 475 ng*hr/mL, about 500 ng*hr/mL, about 525 ng*hr/mL, about 550 ng*hr/mL, about 575 ng*hr/mL, about 600 ng*hr/mL, about 625 ng*hr/mL, about 650 ng*hr/mL, about 675 ng*hr/mL, about 700 ng*hr/mL, about 725 ng*hr/mL, about 750 ng*hr/mL, about 775 ng*hr/mL, about 800 ng*hr/mL, about 825 ng*hr/mL, about 850 ng*hr/mL, about 875 ng*hr/mL, about 900 ng*hr/mL, about 925 ng*hr/mL, about 950 ng*hr/mL, about 975 ng*hr/mL, or about 1000 ng*hr/mL after administration of the formulation to the subject or subject.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 100 ng*hr/mL, greater than about 110 ng*hr/mL, greater than about 120 ng*hr/mL, greater than about 130 ng*hr/mL, greater than about 140 ng*hr/mL, greater than about 150 ng*hr/mL, greater than about 160 ng*hr/mL, greater than about 170 ng*hr/mL, greater than about 180 ng*hr/mL, greater than about 190 ng*hr/mL, greater than about 200 ng*hr/mL, greater than about 225 ng*hr/mL, greater than about 250 ng*hr/mL, greater than about 275 ng*hr/mL, greater than about 300 ng*hr/mL, greater than about 325 ng*hr/mL, greater than about 350 ng*hr/mL, greater than about 375 ng*hr/mL, greater than about 400 ng*hr/mL, greater than about 425 ng*hr/mL, greater than about 450 ng*hr/mL, greater than about 475 ng*hr/mL, greater than about 500 ng*hr/mL, greater than about 525 ng*hr/mL, greater than about 550 ng*hr/mL, greater than about 575 ng*hr/mL, greater than about 600 ng*hr/mL, greater than about 625 ng*hr/mL, greater than about 650 ng*hr/mL, greater than about 675 ng*hr/mL, greater than about 700 ng*hr/mL, greater than about 725 ng*hr/mL, greater than about 750 ng*hr/mL, greater than about 775 ng*hr/mL, greater than about 800 ng*hr/mL, greater than about 825 ng*hr/mL, greater than about 850 ng*hr/mL, greater than about 875 ng*hr/mL, greater than about 900 ng*hr/mL, greater than about 925 ng*hr/mL, greater than about 950 ng*hr/mL, greater than about 975 ng*hr/mL, or greater than about 1000 ng*hr/mL after administration of the formulation to the subject. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 100 ng*hr/mL, greater than about 110 ng*hr/mL, greater than about 120 ng*hr/mL, greater than about 130 ng*hr/mL, greater than about 140 ng*hr/mL, greater than about 150 ng*hr/mL, greater than about 160 ng*hr/mL, greater than about 170 ng*hr/mL, greater than about 180 ng*hr/mL, greater than about 190 ng*hr/mL, greater than about 200 ng*hr/mL, greater than about 225 ng*hr/mL, greater than about 250 ng*hr/mL, greater than about 275 ng*hr/mL, greater than about 300 ng*hr/mL, greater than about 325 ng*hr/mL, greater than about 350 ng*hr/mL, greater than about 375 ng*hr/mL, greater than about 400 ng*hr/mL, greater than about 425 ng*hr/mL, greater than about 450 ng*hr/mL, greater than about 475 ng*hr/mL, greater than about 500 ng*hr/mL, greater than about 525 ng*hr/mL, greater than about 550 ng*hr/mL, greater than about 575 ng*hr/mL, greater than about 600 ng*hr/mL, greater than about 625 ng*hr/mL, greater than about 650 ng*hr/mL, greater than about 675 ng*hr/mL, greater than about 700 ng*hr/mL, greater than about 725 ng*hr/mL, greater than about 750 ng*hr/mL, greater than about 775 ng*hr/mL, greater than about 800 ng*hr/mL, greater than about 825 ng*hr/mL, greater than about 850 ng*hr/mL, greater than about 875 ng*hr/mL, greater than about 900 ng*hr/mL, greater than about 925 ng*hr/mL, greater than about 950 ng*hr/mL, greater than about 975 ng*hr/mL, or greater than about 1000 ng*hr/mL after administration of the formulation to the subject or subject.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 100 ng*hr/mL, about 110 ng*hr/mL, about 120 ng*hr/mL, about 130 ng*hr/mL, about 140 ng*hr/mL, about 150 ng*hr/mL, about 160 ng*hr/mL, about 170 ng*hr/mL, about 180 ng*hr/mL, about 190 ng*hr/mL, about 200 ng*hr/mL, about 225 ng*hr/mL, about 250 ng*hr/mL, ng*hr/mL, about 275 ng*hr/mL, about 300 ng*hr/mL, about 325 ng*hr/mL, about 350 ng*hr/mL, about 375 ng*hr/mL, about 400 ng*hr/mL, about 425 ng*hr/mL, about 450 ng*hr/mL, about 475 ng*hr/mL, about 500 ng*hr/mL, about 525 ng*hr/mL, about 550 ng*hr/mL, about 575 ng*hr/mL, about 600 ng*hr/mL, about 625 ng*hr/mL, about 650 ng*hr/mL, about 675 ng*hr/mL, about 700 ng*hr/mL, about 725 ng*hr/mL, about 750 ng*hr/mL, about 775 ng*hr/mL, about 800 ng*hr/mL, about 825 ng*hr/mL, about 850 ng*hr/mL, about 875 ng*hr/mL, about 900 ng*hr/mL, about 925 ng*hr/mL, about 950 ng*hr/mL, about 975 ng*hr/mL, or about 1000 ng*hr/mL after administration of the formulation to the subject. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 100 ng*hr/mL, about 110 ng*hr/mL, about 120 ng*hr/mL, about 130 ng*hr/mL, about 140 ng*hr/mL, about 150 ng*hr/mL, about 160 ng*hr/mL, about 170 ng*hr/mL, about 180 ng*hr/mL, about 190 ng*hr/mL, about 200 ng*hr/mL, about 225 ng*hr/mL, about 250 ng*hr/mL, ng*hr/mL, about 275 ng*hr/mL, about 300 ng*hr/mL, about 325 ng*hr/mL, about 350 ng*hr/mL, about 375 ng*hr/mL, about 400 ng*hr/mL, about 425 ng*hr/mL, about 450 ng*hr/mL, about 475 ng*hr/mL, about 500 ng*hr/mL, about 525 ng*hr/mL, about 550 ng*hr/mL, about 575 ng*hr/mL, about 600 ng*hr/mL, about 625 ng*hr/mL, about 650 ng*hr/mL, about 675 ng*hr/mL, about 700 ng*hr/mL, about 725 ng*hr/mL, about 750 ng*hr/mL, about 775 ng*hr/mL, about 800 ng*hr/mL, about 825 ng*hr/mL, about 850 ng*hr/mL, about 875 ng*hr/mL, about 900 ng*hr/mL, about 925 ng*hr/mL, about 950 ng*hr/mL, about 975 ng*hr/mL, or about 1000 ng*hr/mL after administration of the formulation to the subject or subject.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject a Cmax of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 40 ng/mL, greater than about 50 ng/mL, greater than about 60 ng/mL, greater than about 70 ng/mL, greater than about 80 ng/mL, greater than about 90 ng/mL, greater than about 100 ng/mL, greater than about 110 ng/mL, greater than about 120 ng/mL, greater than about 130 ng/mL, greater than about 140 ng/mL, greater than about 150 ng/mL, greater than about 160 ng/mL, greater than about 170 ng/mL, greater than about 180 ng/mL, greater than about 190 ng/mL, greater than about 200 ng/mL, greater than about 210 ng/mL, greater than about 220 ng/mL, greater than about 230 ng/mL, greater than about 240 ng/mL, greater than about 250 ng/mL, greater than about 260 ng/mL, greater than about 270 ng/mL, greater than about 280 ng/mL, greater than about 290 ng/mL, greater than about 300 ng/mL, greater than about 310 ng/mL, greater than about 320 ng/mL, greater than about 330 ng/mL, greater than about 340 ng/mL, greater than about 350 ng/mL, greater than about 360 ng/mL, greater than about 370 ng/mL, greater than about 380 ng/mL, greater than about 390 ng/mL, or greater than about 400 ng/mL after administration of the formulation to the subject. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject a Cmax of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 40 ng/mL, greater than about 50 ng/mL, greater than about 60 ng/mL, greater than about 70 ng/mL, greater than about 80 ng/mL, greater than about 90 ng/mL, greater than about 100 ng/mL, greater than about 110 ng/mL, greater than about 120 ng/mL, greater than about 130 ng/mL, greater than about 140 ng/mL, greater than about 150 ng/mL, greater than about 160 ng/mL, greater than about 170 ng/mL, greater than about 180 ng/mL, greater than about 190 ng/mL, greater than about 200 ng/mL, greater than about 210 ng/mL, greater than about 220 ng/mL, greater than about 230 ng/mL, greater than about 240 ng/mL, greater than about 250 ng/mL, greater than about 260 ng/mL, greater than about 270 ng/mL, greater than about 280 ng/mL, greater than about 290 ng/mL, greater than about 300 ng/mL, greater than about 310 ng/mL, greater than about 320 ng/mL, greater than about 330 ng/mL, greater than about 340 ng/mL, greater than about 350 ng/mL, greater than about 360 ng/mL, greater than about 370 ng/mL, greater than about 380 ng/mL, greater than about 390 ng/mL, or greater than about 400 ng/mL after administration of the formulation to the subject or subject.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject a Cmax of cromolyn or a pharmaceutically-acceptable salt thereof of about 50 mg/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, about 90 ng/mL, about 100 ng/mL, about 110 ng/mL, about 120 ng/mL, about 130 ng/mL, about 140 ng/mL, about 150 ng/mL, about 160 ng/mL, about 170 ng/mL, about 180 ng/mL, about 190 ng/mL, about 200 ng/mL, about 210 ng/mL, about 220 ng/mL, about 230 ng/mL, about 240 ng/mL, about 250 ng/mL, about 260 ng/mL, about 270 ng/mL, about 280 ng/mL, about 290 ng/mL, about 300 ng/mL, about 310 ng/mL, about 320 ng/mL, about 330 ng/mL, about 340 ng/mL, about 350 ng/mL, about 360 ng/mL, about 370 ng/mL, about 380 ng/mL, about 390 ng/mL, or about 400 ng/mL after administration of the formulation to the subject. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject a Cmax of cromolyn or a pharmaceutically-acceptable salt thereof of about 50 mg/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, about 90 ng/mL, about 100 ng/mL, about 110 ng/mL, about 120 ng/mL, about 130 ng/mL, about 140 ng/mL, about 150 ng/mL, about 160 ng/mL, about 170 ng/mL, about 180 ng/mL, about 190 ng/mL, about 200 ng/mL, about 210 ng/mL, about 220 ng/mL, about 230 ng/mL, about 240 ng/mL, about 250 ng/mL, about 260 ng/mL, about 270 ng/mL, about 280 ng/mL, about 290 ng/mL, about 300 ng/mL, about 310 ng/mL, about 320 ng/mL, about 330 ng/mL, about 340 ng/mL, about 350 ng/mL, about 360 ng/mL, about 370 ng/mL, about 380 ng/mL, about 390 ng/mL, or about 400 ng/mL after administration of the formulation to the subject or subject.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject a Cmax of cromolyn sodium greater than about 40 ng/mL, greater than about 50 ng/mL, greater than about 60 ng/mL, greater than about 70 ng/mL, greater than about 80 ng/mL, greater than about 90 ng/mL, greater than about 100 ng/mL, greater than about 110 ng/mL, greater than about 120 ng/mL, greater than about 130 ng/mL, greater than about 140 ng/mL, greater than about 150 ng/mL, greater than about 160 ng/mL, greater than about 170 ng/mL, greater than about 180 ng/mL, greater than about 190 ng/mL, greater than about 200 ng/mL, greater than about 210 ng/mL, greater than about 220 ng/mL, greater than about 230 ng/mL, greater than about 240 ng/mL, greater than about 250 ng/mL, greater than about 260 ng/mL, greater than about 270 ng/mL, greater than about 280 ng/mL, greater than about 290 ng/mL, greater than about 300 ng/mL, greater than about 310 ng/mL, greater than about 320 ng/mL, greater than about 330 ng/mL, greater than about 340 ng/mL, greater than about 350 ng/mL, greater than about 360 ng/mL, greater than about 370 ng/mL, greater than about 380 ng/mL, greater than about 390 ng/mL, or greater than about 400 ng/mL after administration of the formulation to the subject. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject a Cmax of cromolyn sodium greater than about 40 ng/mL, greater than about 50 ng/mL, greater than about 60 ng/mL, greater than about 70 ng/mL, greater than about 80 ng/mL, greater than about 90 ng/mL, greater than about 100 ng/mL, greater than about 110 ng/mL, greater than about 120 ng/mL, greater than about 130 ng/mL, greater than about 140 ng/mL, greater than about 150 ng/mL, greater than about 160 ng/mL, greater than about 170 ng/mL, greater than about 180 ng/mL, greater than about 190 ng/mL, greater than about 200 ng/mL, greater than about 210 ng/mL, greater than about 220 ng/mL, greater than about 230 ng/mL, greater than about 240 ng/mL, greater than about 250 ng/mL, greater than about 260 ng/mL, greater than about 270 ng/mL, greater than about 280 ng/mL, greater than about 290 ng/mL, greater than about 300 ng/mL, greater than about 310 ng/mL, greater than about 320 ng/mL, greater than about 330 ng/mL, greater than about 340 ng/mL, greater than about 350 ng/mL, greater than about 360 ng/mL, greater than about 370 ng/mL, greater than about 380 ng/mL, greater than about 390 ng/mL, or greater than about 400 ng/mL after administration of the formulation to the subject or subject.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject a Cmax of cromolyn sodium of about 50 mg/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, about 90 ng/mL, about 100 ng/mL, about 110 ng/mL, about 120 ng/mL, about 130 ng/mL, about 140 ng/mL, about 150 ng/mL, about 160 ng/mL, about 170 ng/mL, about 180 ng/mL, about 190 ng/mL, about 200 ng/mL, about 210 ng/mL, about 220 ng/mL, about 230 ng/mL, about 240 ng/mL, about 250 ng/mL, about 260 ng/mL, about 270 ng/mL, about 280 ng/mL, about 290 ng/mL, about 300 ng/mL, about 310 ng/mL, about 320 ng/mL, about 330 ng/mL, about 340 ng/mL, about 350 ng/mL, about 360 ng/mL, about 370 ng/mL, about 380 ng/mL, about 390 ng/mL, or about 400 ng/mL after administration of the formulation to the subject. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject a Cmax of cromolyn sodium of about 50 mg/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, about 90 ng/mL, about 100 ng/mL, about 110 ng/mL, about 120 ng/mL, about 130 ng/mL, about 140 ng/mL, about 150 ng/mL, about 160 ng/mL, about 170 ng/mL, about 180 ng/mL, about 190 ng/mL, about 200 ng/mL, about 210 ng/mL, about 220 ng/mL, about 230 ng/mL, about 240 ng/mL, about 250 ng/mL, about 260 ng/mL, about 270 ng/mL, about 280 ng/mL, about 290 ng/mL, about 300 ng/mL, about 310 ng/mL, about 320 ng/mL, about 330 ng/mL, about 340 ng/mL, about 350 ng/mL, about 360 ng/mL, about 370 ng/mL, about 380 ng/mL, about 390 ng/mL, or about 400 ng/mL after administration of the formulation to the subject.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 120 ng*hr/mL and/or an average Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 120 ng*hr/mL and/or a Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 55 ng/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 200 ng*hr/mL and an average Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 80 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 200 ng*hr/mL and a Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 80 ng/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 330 ng*hr/mL and an average Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 150 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 330 ng*hr/mL and a Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 150 ng/mL.

In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 525 ng*hr/mL and an average Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 230 ng/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 525 ng*hr/mL and a Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 230 ng/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL and/or an average Cmax of cromolyn sodium greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL and/or a Cmax of cromolyn sodium greater than about 55 ng/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL and an average Cmax of cromolyn sodium greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL and a Cmax of cromolyn sodium greater than about 55 ng/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL and an average Cmax of cromolyn sodium greater than about 80 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL and a Cmax of cromolyn sodium greater than about 80 ng/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL and an average Cmax of cromolyn sodium greater than about 150 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL and a Cmax of cromolyn sodium greater than about 150 ng/mL.

In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL and an average Cmax of cromolyn sodium greater than about 230 ng/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL and a Cmax of cromolyn sodium greater than about 230 ng/mL.

In certain embodiments of the methods disclosed herein, the administration of a formulation comprising about 40 mg of cromolyn sodium, or a pharmaceutically acceptable salt thereof, to a subject by means of an inhalation device affords a Cmax or average Cmax of cromolyn sodium in the subject of from about from 30 ng/mL to about 120 ng/mL, or from about 40 ng/mL to about 120 ng/mL, or from about 40 ng/mL to about 110 ng/mL. In certain embodiments, the administration of a formulation comprising about 40 mg of cromolyn sodium, or a pharmaceutically acceptable salt thereof, to a subject by means of an inhalation device affords an AUC(0-∞) of cromolyn sodium in the subject of between about 100 ng*hr/mL and about 350 ng*hr/mL, or between about 100 ng*hr/mL and about 325 ng*hr/mL, or between about 115 ng*hr/mL and about 325 ng*hr/mL, or between about 120 ng*hr/mL and about 320 ng*hr/mL, or between about 125 ng*hr/mL and about 300 ng*hr/mL. In certain embodiments, the formulation comprises between 1% and 10% by weight cromolyn sodium, or between about 4% by weight and 6% by weight cromolyn sodium. In certain embodiments, the formulation comprises 4% by weight cromolyn sodium. In certain embodiments, the formulation comprises 6% by weight cromolyn sodium. In certain embodiments, use of a nebulizer to administer the formulation provides an aerosol of the formulation comprising cromolyn or a pharmaceutically-acceptable salt thereof, wherein the aerosol exhibits an RF (≤5 μm) of an cromolyn or a pharmaceutically-acceptable salt thereof of at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, about 20% to about 95%, about 35% to about 90%, or about 40% to about 80%, about 40% to about 90%, about 40% to about 95%, about 45% to about 90%, about 45% to about 95%, about 50% to about 90%, about 65% to about 90%, about 60% to about 95%, about 65% to about 95%, about 70% to about 90%, about 55% to about 90%, about 70% to about 80%, or about 75%. In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution comprising cromolyn or a pharmaceutically-acceptable salt thereof, wherein the aerosol exhibits an RF (≤5 μm) of cromolyn sodium of at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, about 20% to about 95%, about 35% to about 90%, or about 40% to about 80%, about 40% to about 90%, about 40% to about 95%, about 45% to about 90%, about 45% to about 95%, about 50% to about 90%, about 65% to about 90%, about 60% to about 95%, about 65% to about 95%, about 70% to about 90%, about 55% to about 90%, about 70% to about 80%, about 65% to about 75%, about 65% to about 80%, about 60% to about 80%, about 66%, or about 75%. In certain embodiments, the formulation comprises an osmotic agent comprising sodium chloride. In certain embodiments, the formulation comprises an osmotic agent consisting of sodium chloride. In certain embodiments, the osmolality of the formulation is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the formulation is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments of the methods disclosed herein, the administration of a formulation comprising about 60 mg of cromolyn sodium, or a pharmaceutically acceptable salt thereof, to a subject by means of an inhalation device affords a Cmax or average Cmax of cromolyn sodium in the subject of from about from 50 ng/mL to about 175 ng/mL, or from about 60 ng/mL to about 175 ng/mL, or from about 60 ng/mL to about 170 ng/mL, or from about 60 ng/mL to about 165 ng/mL, or from about 70 ng/mL to about 165 ng/mL. In certain embodiments, the administration of a formulation comprising about 60 mg of cromolyn sodium, or a pharmaceutically acceptable salt thereof, to a subject by means of an inhalation device affords an AUC(0-∞) of cromolyn sodium in the subject of between about 200 ng*hr/mL and about 600 ng*hr/mL, or between about 200 ng*hr/mL and about 575 ng*hr/mL, or between about 200 ng*hr/mL and about 550 ng*hr/mL, or between about 200 ng*hr/mL and about 525 ng*hr/mL, or between about 210 ng*hr/mL and about 525 ng*hr/mL, or between about 215 ng*hr/mL and about 515 ng*hr/mL, or between about 175 ng*hr/mL and about 500 ng*hr/mL, or between about 195 ng*hr/mL and about 515 ng*hr/mL, or between about 200 ng*hr/mL and about 500 ng*hr/mL. In certain embodiments, the formulation comprises between 1% and 10% by weight cromolyn sodium, or between about 4% by weight and 6% by weight cromolyn sodium. In certain embodiments, the formulation comprises 4% by weight cromolyn sodium. In certain embodiments, the formulation comprises 6% by weight cromolyn sodium. In certain embodiments, use of a nebulizer to administer the formulation provides an aerosol of the formulation comprising cromolyn or a pharmaceutically-acceptable salt thereof, wherein the aerosol exhibits an RF (≤5 μm) of an cromolyn or a pharmaceutically-acceptable salt thereof of at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, about 20% to about 95%, about 35% to about 90%, or about 40% to about 80%, about 40% to about 90%, about 40% to about 95%, about 45% to about 90%, about 45% to about 95%, about 50% to about 90%, about 65% to about 90%, about 60% to about 95%, about 65% to about 95%, about 70% to about 90%, about 55% to about 90%, about 70% to about 80%, or about 75%. In certain embodiments, use of a nebulizer in the methods disclosed herein provides an aerosol of a solution comprising cromolyn or a pharmaceutically-acceptable salt thereof, wherein the aerosol exhibits an RF (≤5 μm) of cromolyn sodium of at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, about 20% to about 95%, about 35% to about 90%, or about 40% to about 80%, about 40% to about 90%, about 40% to about 95%, about 45% to about 90%, about 45% to about 95%, about 50% to about 90%, about 65% to about 90%, about 60% to about 95%, about 65% to about 95%, about 70% to about 90%, about 55% to about 90%, about 70% to about 80%, about 65% to about 75%, about 65% to about 80%, about 60% to about 80%, about 66%, or about 75%. In certain embodiments, the formulation comprises an osmotic agent comprising sodium chloride. In certain embodiments, the formulation comprises an osmotic agent consisting of sodium chloride. In certain embodiments, the osmolality of the formulation is between about 100 mOsm/kg and about 175 mOsm/kg, or between about 100 mOsm/kg and about 170 mOsm/kg, or between about 100 mOsm/kg and about 165 mOsm/kg, or between about 100 mOsm/kg and about 160 mOsm/kg, or between about 100 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 150 mOsm/kg, or between about 110 mOsm/kg and about 140 mOsm/kg, or between about 115 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 140 mOsm/kg, or between about 120 mOsm/kg and about 130 mOsm/kg. In certain embodiments, the osmolality of the formulation is about 120 mOsm/kg, about 125 mOsm/kg, about 130 mOsm/kg, about 135 mOsm/kg, about 140 mOsm/kg, about 145 mOsm/kg, or about 150 mOsm/kg.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 330 ng*hr/mL and an average Cmax of cromolyn sodium of about 150 ng/mL when a nominal dose of 40 mg of cromolyn sodium is administered with the inhalation device. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 330 ng*hr/mL and a Cmax of cromolyn sodium of about 150 ng/mL when a nominal dose of 40 mg of cromolyn sodium is administered with the inhalation device.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 525 ng*hr/mL and an average Cmax of cromolyn sodium of about 230 ng/mL when a nominal dose of 80 mg of cromolyn sodium is administered with the inhalation device. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 525 ng*hr/mL and a Cmax of cromolyn sodium of about 230 ng/mL when a nominal dose of 80 mg of cromolyn sodium is administered with the inhalation device.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 180 ng*hr/mL to about 220 ng*hr/mL and an average Cmax of cromolyn sodium of about 70 ng/mL to about 90 ng/mL when a nominal dose of 40 mg of cromolyn sodium is administered with the inhalation device. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 180 ng*hr/mL to about 220 ng*hr/mL and a Cmax of cromolyn sodium of about 70 ng/mL to about 90 ng/mL when a nominal dose of 40 mg of cromolyn sodium is administered with the inhalation device.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 300 ng*hr/mL to about 360 ng*hr/mL and an average Cmax of cromolyn sodium of about 135 ng/mL to about 165 ng/mL when a nominal dose of 40 mg of cromolyn sodium is administered with the inhalation device. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 300 ng*hr/mL to about 360 ng*hr/mL and a Cmax of cromolyn sodium of about 135 ng/mL to about 165 ng/mL when a nominal dose of 40 mg of cromolyn sodium is administered with the inhalation device.

In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 475 ng*hr/mL to about 575 ng*hr/mL and an average Cmax of cromolyn sodium of about 200 ng/mL to about 260 ng/mL when a nominal dose of 80 mg of cromolyn sodium is administered with the inhalation device. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 475 ng*hr/mL to about 575 ng*hr/mL and a Cmax of cromolyn sodium of about 200 ng/mL to about 260 ng/mL when a nominal dose of 80 mg of cromolyn sodium is administered with the inhalation device.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a lung deposition (deposited lung dose) comprising cromolyn or pharmaceutically acceptable salt thereof of at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, about 20% to about 40%, about 25% to about 35%, about 25% to about 30%, about 25% to about 75%, about 30% to about 50%, about 35% to about 90%, about 40% to about 80%, about 40% to about 60%, about 50% to about 60%, about 50% to about 70%, or about 60% to about 75% based on the nominal dose of the cromolyn or a pharmaceutically-acceptable salt thereof. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides cromolyn sodium deposition (deposited lung dose) of at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, about 20% to about 40%, about 25% to about 35%, about 25% to about 30%, about 25% to about 75%, about 30% to about 50%, about 35% to about 90%, about 40% to about 80%, about 40% to about 60%, about 50% to about 60%, about 50% to about 70%, or about 60% to about 75% based on the nominal dose of the cromolyn sodium.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a lung deposition (deposited lung dose) comprising cromolyn or a pharmaceutically-acceptable salt thereof of about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75% about 80%, about 85%, about 90%, about 95%, or about 100% based on the nominal dose of the cromolyn or a pharmaceutically-acceptable salt thereof. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides cromolyn sodium lung deposition (deposited lung dose) of about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75% about 80%, about 85%, about 90%, about 95%, or about 100% based on the nominal dose of the cromolyn sodium.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a lung deposition (deposited lung dose) comprising cromolyn or a pharmaceutically-acceptable salt thereof of greater than about 0.5 mg, greater than about 1 mg, greater than about 1.5 mg, greater than about 2 mg, greater than about 2.5 mg, greater than about 3 mg, greater than about 3.5 mg, greater than about 4 mg, greater than about 5 mg, greater than about 6 mg, greater than about 7 mg, greater than about 8 mg, greater than about 9 mg, greater than about 10 mg, greater than about 11 mg, greater than about 12 mg, greater than about 13 mg, greater than about 14 mg, or greater than about 15 mg. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a lung deposition (deposited lung dose) comprising cromolyn or a pharmaceutically-acceptable salt thereof of about 0.5 mg, about 1.0 mg, about 1.5 mg, about 2.0 mg, about 2.5 mg, about 3.0 mg, about 3.5 mg, about 4.0 mg, about 5.0 mg, about 6.0 mg, about 7.0 mg, about 8.0 mg, about 9.0 mg, about 10 mg, about 11 mg, about 12 mg, about 13 mg, about 14 mg, or about 15 mg.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides cromolyn sodium lung deposition (deposited lung dose) of greater than about 0.5 mg, greater than about 1 mg, greater than about 1.5 mg, greater than about 2 mg, greater than about 2.5 mg, greater than about 3 mg, greater than about 3.5 mg, greater than about 4 mg, greater than about 5 mg, greater than about 6 mg, greater than about 7 mg, greater than about 8 mg, greater than about 9 mg, greater than about 10 mg, greater than about 11 mg, greater than about 12 mg, greater than about 13 mg, greater than about 14 mg, or greater than about 15 mg. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides cromolyn sodium lung deposition (deposited lung dose) of about 0.5 mg, about 1.0 mg, about 1.5 mg, about 2.0 mg, about 2.5 mg, about 3.0 mg, about 3.5 mg, about 4.0 mg, about 5.0 mg, about 6.0 mg, about 7.0 mg, about 8.0 mg, about 9.0 mg, about 10 mg, about 11 mg, about 12 mg, about 13 mg, about 14 mg, or about 15 mg.

In certain embodiments of the methods disclosed herein, an inhalation formulation containing cromolyn or a pharmaceutically-acceptable salt thereof is administered with an inhalation device, e.g., a nebulizer, at an administration of less than about 1 mg/dose, about 1 mg/dose to about 100 mg/dose, about 5 mg/dose to about 80 mg/dose, about 20 mg/dose to about 60 mg/dose, about 30 mg/dose to about 50 mg/dose, or greater than 100 mg/dose. In certain embodiments of the methods disclosed herein, an inhalation formulation containing cromolyn sodium is administered with an inhalation device, e.g., a nebulizer, at an administration of less than about 1 mg/dose, about 1 mg/dose to about 100 mg/dose, about 5 mg/dose to about 80 mg/dose, about 20 mg/dose to about 60 mg/dose, about 30 mg/dose to about 50 mg/dose, or greater than 100 mg/dose. In certain embodiments of the methods disclosed herein, cromolyn or a pharmaceutically-acceptable salt thereof is administered in an inhalation formulation with an inhalation device, e.g., a nebulizer, in about 1 mg, about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 105 mg, about 110 mg, about 115 mg, about 120 mg, about 125 mg, about 130 mg doses, about 135 mg, about 140 mg, about 145 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, or about 1000 mg doses. In certain embodiments of the methods disclosed herein, cromolyn sodium is administered in an inhalation formulation with an inhalation device, e.g., a nebulizer, in about 1 mg, about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 105 mg, about 110 mg, about 115 mg, about 120 mg, about 125 mg, about 130 mg doses, about 135 mg, about 140 mg, about 145 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, or about 1000 mg doses.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof of greater than about 5%, greater than about 6%, greater than about 7%, greater than about 8%, greater than about 9%, greater than about 10%, greater than about 11%, greater than about 12%, greater than about 13%, greater than about 14%, greater than about 15%, greater than about 16%, greater than about 17%, greater than about 18%, greater than about 19%, greater than about 20%, greater than about 25%, greater than about 30%, greater than about 35%, greater than about 40%, greater than about 45%, greater than about 50%, greater than about 55%, or greater than about 60% of the nominal dose. In certain embodiments, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, in the methods disclosed herein provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof of about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, or about 60% of the nominal dose.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer provides a bioavailability of cromolyn sodium of greater than about 5%, greater than about 6%, greater than about 7%, greater than about 8%, greater than about 9%, greater than about 10%, greater than about 11%, greater than about 12%, greater than about 13%, greater than about 14%, greater than about 15%, greater than about 16%, greater than about 17%, greater than about 18%, greater than about 19%, greater than about 20%, greater than about 25%, greater than about 30%, greater than about 35%, greater than about 40%, greater than about 45% or greater than about 50% of the nominal dose. In certain embodiments, an aqueous inhalation formulation administered with an inhalation device, e.g., a nebulizer, in the methods disclosed herein provides a bioavailability of cromolyn sodium of about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, or about 50% of the nominal dose.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 120 ng*hr/mL and/or an average Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 120 ng*hr/mL and/or a Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 120 ng*hr/mL and an average Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 120 ng*hr/mL and a Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 200 ng*hr/mL and an average Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 80 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 200 ng*hr/mL and a Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 80 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 330 ng*hr/mL and an average Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 150 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 330 ng*hr/mL and a Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 150 ng/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 525 ng*hr/mL and an average Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 230 ng/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 525 ng*hr/mL and a Cmax of the cromolyn or a pharmaceutically-acceptable salt thereof greater than about 230 ng/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL and/or an average Cmax of cromolyn sodium greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL and/or a Cmax of cromolyn sodium greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL and an average Cmax of cromolyn sodium greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL and a Cmax of cromolyn sodium greater than about 55 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL and an average Cmax of cromolyn sodium greater than about 80 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL and a Cmax of cromolyn sodium greater than about 80 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL and an average Cmax of cromolyn sodium greater than about 150 ng/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL and a Cmax of cromolyn sodium greater than about 150 ng/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL and an average Cmax of cromolyn sodium greater than about 230 ng/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL and a Cmax of cromolyn sodium greater than about 230 ng/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 120 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 120 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 330 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 330 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 525 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 525 ng*hr/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation comprising 40 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation comprising 40 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation comprising 40 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation comprising 40 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation comprising 80 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation comprising 80 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, provides a bioavailability of cromolyn sodium greater than about 5% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 am) of at least about 30% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 120 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 am) of at least about 30% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 120 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 30% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 30% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 330 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 330 ng*hr/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 525 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn or a pharmaceutically-acceptable salt thereof greater than about 525 ng*hr/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 30% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 30% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 120 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 30% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 30% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL. In certain embodiments, of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation comprising 40 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 30% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation comprising 40 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 30% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 200 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation comprising 40 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation comprising 40 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 330 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation comprising 80 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL. In certain embodiments of the methods disclosed herein, an inhalation formulation comprising 80 mg cromolyn sodium administered with an inhalation device, e.g., a nebulizer, has an RF (≤3.3 μm) of at least about 40% and produces in a subject an AUC(0-∞) of cromolyn sodium greater than about 525 ng*hr/mL.

In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 8.5 ng*hr/mL per mg of cromolyn sodium, and an average Cmax of cromolyn sodium of about 3.9 ng/mL per mg of cromolyn sodium when a nominal dose of 40 mg cromolyn sodium is administered to the subject with an inhalation device. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 8.5 ng*hr/mL per mg of cromolyn sodium, and an average Cmax of cromolyn sodium of about 1.9 ng/mL per mg of cromolyn sodium when a nominal dose of 40 mg cromolyn sodium is administered to the subject with an inhalation device. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 9 ng*hr/mL and a Cmax of cromolyn sodium of about 2.6 ng/mL per mg of cromolyn sodium when a nominal dose of 60 mg of cromolyn sodium administered to the subject with the inhalation device. In certain embodiments of the methods disclosed herein, an inhalation formulation administered with an inhalation device, e.g., a nebulizer, produces in a subject an AUC(0-∞) of cromolyn sodium of about 6.6 ng*hr/mL and an average Cmax of cromolyn sodium of about 2.95 ng/mL per mg of cromolyn sodium when a nominal dose of 80 mg of cromolyn sodium is administered to the subject with the inhalation device.

In certain embodiments of the methods disclosed herein, an inhalation formulation containing cromolyn or a pharmaceutically-acceptable salt thereof such as cromolyn sodium is administered with an inhalation device, e.g., a nebulizer, at a fill volume of less than about 0.25 mL, less than about 0.5 mL, at least about 0.5 mL to about 1.5 mL, at least about 0.5 mL to about 1.8 mL, at least about 1.5 mL, or at least about 2.0 mL. In certain embodiments, an inhalation formulation is administered with an inhalation device, e.g., a nebulizer, at a fill volume about 0.1 mL to about 5.0 mL, about 0.25 mL to about 2.0 mL, about 0.5 mL to about 1.8 mL, about 0.5 mL to about 2 mL, about 0.5 mL to about 1.5 mL, about 0.5 mL to about 1.0 mL, about 0.5 mL or less, about 1 mL or less, about 1.5 mL or less, about 2.0 mL or less, about 2.5 mL or less, about 3.0 mL or less, about 3.5 mL or less, about 4.0 mL or less, about 4.5 mL or less, or about 5.0 mL or less. In certain embodiments, an inhalation formulation is administered with an inhalation device, e.g., a nebulizer, at a fill volume of about 0.5 mL, about 1.0 mL, about 1.5 mL, about 1.8 mL, about 2.0 mL, about 2.5 mL, about 3.0 mL, about 3.5 mL, about 4.0 mL, about 4.5 mL, or about 5.0 mL. In certain embodiments, an inhalation formulation is administered with an inhalation device, e.g., a nebulizer, which provides for a residual volume of cromolyn or a pharmaceutically-acceptable salt thereof after administration of the cromolyn or a pharmaceutically-acceptable salt thereof of less than about 10%, less than about 5%, or less than about 3% of the nominal dose. In certain embodiments of the methods disclosed herein, an inhalation formulation containing cromolyn or a pharmaceutically-acceptable salt thereof is administered with an inhalation device, e.g., a nebulizer, wherein the concentration of the cromolyn or a pharmaceutically-acceptable salt thereof is greater than about 1% by weight, greater than about 2% by weight, greater than about 3% by weight, greater than about 4% by weight, greater than about 5% by weight, greater than about 6% by weight, greater than about 7% by weight, greater than about 8% by weight, greater than about 9% by weight, or greater than about 10% by weight. In certain embodiments of the methods disclosed herein, an inhalation formulation containing cromolyn or a pharmaceutically-acceptable salt thereof is administered with an inhalation device, e.g., a nebulizer, wherein the concentration of the cromolyn or a pharmaceutically-acceptable salt thereof is from about 1% by weight to about 10% by weight, from about 2% by weight to about 8% by weight, from about 2% by weight to about 6% by weight, or from about 3% by weight to about 5% by weight. In certain embodiments of the methods disclosed herein, an inhalation formulation containing cromolyn or a pharmaceutically-acceptable salt thereof is administered with an inhalation device, e.g., a nebulizer, wherein the concentration of the cromolyn or a pharmaceutically-acceptable salt thereof is about 1% by weight, about 2% by weight, about 3% by weight, about 4% by weight, about 5% by weight, about 6% by weight, about 7% by weight, about 8% by weight, about 9% by weight, or about 10% by weight.

In certain embodiments of the methods disclosed herein, an inhalation formulation containing cromolyn sodium is administered with an inhalation device, e.g., a nebulizer, wherein the concentration of the cromolyn sodium is greater than about 1% by weight, greater than about 2% by weight, greater than about 3% by weight, greater than about 4% by weight, greater than about 5% by weight, greater than about 6% by weight, greater than about 7% by weight, greater than about 8% by weight, greater than about 9% by weight, or greater than about 10% by weight. In certain embodiments of the methods disclosed herein, an inhalation formulation containing cromolyn sodium is administered with an inhalation device, e.g., a nebulizer, wherein the concentration of the cromolyn sodium is from about 1% by weight to about 10% by weight, from about 2% by weight to about 8% by weight, from about 2% by weight to about 6% by weight, or from about 3% by weight to about 5% by weight. In certain embodiments of the methods disclosed herein, an inhalation formulation containing cromolyn sodium is administered with an inhalation device, e.g., a nebulizer, wherein the concentration of the cromolyn sodium is about 1% by weight, about 2% by weight, about 3% by weight, about 4% by weight, about 5% by weight, about 6% by weight, about 7% by weight, about 8% by weight, about 9% by weight, or about 10% by weight.

In certain embodiments, an inhalation formulation containing cromolyn or a pharmaceutically-acceptable salt thereof is administered with an inhalation device, e.g., a nebulizer, in about 0.25 to about 10 minutes, about 0.50 to about 8 minutes, less than about 8 minutes, less than about 7 minutes, less than about 6 minutes, less than about 5 minutes, less than about 4 minutes, less than about 3 minutes, less than about 2 minutes, less than about 1.8 minutes, less than about 1.5 minutes, or less than 1 minute. In certain embodiments, the inhalation formulation is administered in about 3 minutes or less. In certain embodiments, the inhalation formulation is administered in about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, or about 10 minutes.

In certain embodiments of the methods disclosed herein, administration of cromolyn or a pharmaceutically-acceptable salt thereof with a nebulizer provides at least about a 1.5-fold, at least about a 1.8-fold, at least about a two-fold, at least about a three-fold, at least about a four-fold, or at least about a five-fold increase in one or more of AUClast, AUC(0-∞), or Cmx as compared to the same or lower nominal dose of the cromolyn or a pharmaceutically-acceptable salt thereof administered with a conventional inhalation device or an oral formulation, e.g., a liquid oral formulation, tablet, or capsule.

In certain embodiments of the methods disclosed herein, inhalation formulations administered with a nebulizer are substantially free of a preservative, such as benzyl alcohol. In certain embodiments of the methods disclosed herein, inhalation formulations administered with a nebulizer further comprise at least one excipient. In certain embodiments, the excipient is selected from the group consisting of stabilizers and antioxidants (such as citric acid, ascorbic acid, ethylenediamine tetra acetic acid (EDTA), sodium metabisulfite, or a salt of any thereof), an osmolarity adjusting agent (such as sodium chloride, mannitol, or sorbitol), a surfactant (such as polysorbate 80, vitamin E, tocopherol polyethylene glycol, and Tyloxapol), or a pH buffer.

In certain embodiments of the methods disclosed herein, inhalation formulations administered with an inhalation device, e.g., a nebulizer, are hypotonic. In certain embodiments of the methods disclosed herein, inhalation formulations administered with an inhalation device, e.g., a nebulizer, are sub-isotonic. In certain embodiments of the methods disclosed herein, inhalation formulations administered with an inhalation device, e.g., a nebulizer, have an osmolality greater than about 70 mOsm/kg. In certain embodiments of the methods disclosed herein, inhalation formulations administered with an inhalation device, e.g., nebulizer, have an osmolality of at least about 100 mOsm/kg. In certain embodiments of the methods disclosed herein, inhalation formulations administered with an inhalation device, e.g., nebulizer, have an osmolality of at least about 150 mOsm/kg.

Combination Therapies

In certain embodiments of the methods disclosed herein, the formulations comprising from about 2% to about 10% by weight of cromolyn sodium and an osmotic agent consisting of sodium chloride, are administered to a subject in need thereof in combination with an additional agent used to treat IPF. In certain embodiments, the additional agent is selected from pirfenidone, an inhibitor of platelet-derived growth factor receptor (PDGFR) a, platelet-derived growth factor receptor (PDGFR) J3, an inhibitor of fibroblast growth factor receptor (FGFR) 1-3, an inhibitor of vascular endothelial growth factor receptor (VEGFR) 1-3, and an inhibitor of Fms-like tyrosine kinase-3 (FLT3). In certain embodiments, the additional agent is pirfenadone or nintedanib esylate. In certain embodiments, the additional agent is pifenadone. In certain embodiments, the additional agent is nintedanib esylate.

In certain embodiments are disclosed methods of treating chronic cough in a subject having IPF, comprising administering to the subject a combination of (a) a formulation comprising from about 2% to about 10% by weight of cromolyn sodium and an osmotic agent consisting of sodium chloride, and (b) an additional agent. In certain embodiments, the additional agent is selected from pirfenidone, an inhibitor of platelet-derived growth factor receptor (PDGFR) u, platelet-derived growth factor receptor (PDGFR) J3, an inhibitor of fibroblast growth factor receptor (FGFR) 1-3, an inhibitor of vascular endothelial growth factor receptor (VEGFR) 1-3, and an inhibitor of Fms-like tyrosine kinase-3 (FLT3). In certain embodiments, the additional agent is pirfenadone or nintedanib esylate. In certain embodiments, the additional agent is pifenadone. In certain embodiments, the additional agent is nintedanib esylate.

In certain embodiments of the methods disclosed herein, one or more different formulations of cromolyn or a pharmaceutically-acceptable salt thereof are co-administered by different routes of administration to provide systemically effective amounts of the cromolyn or a pharmaceutically-acceptable salt thereof. For example, in certain embodiments, a composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is administered with a dry powder inhaler and a different composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is co-administered in a liquid oral formulation to treat chronic cough due to IPF. In certain embodiments, a composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is administered with a metered dose inhaler and a different composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is co-administered in a liquid oral formulation to treat chronic cough due to IPF. In certain embodiments, a composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is administered with a dry powder inhaler and a different composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is co-administered with a metered dose inhaler to treat chronic cough due to IPF. In certain embodiments, a composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is administered with a dry powder inhaler and a different composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is co-administered with a metered dose inhaler to treat chronic cough due to IPF. In certain embodiments, a composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is administered with a nebulizer and a different composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is co-administered in a liquid oral formulation to treat chronic cough due to IPF. In certain embodiments, a composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is administered with a jet nebulizer and a different composition comprising cromolyn or a pharmaceutically-acceptable salt thereof, e.g., cromolyn sodium, is co-administered in a liquid oral formulation to treat chronic cough due to IPF.

Definition of Terms

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of skill in the art to which the inventions described herein belong. All publications, patents, and patent applications mentioned in this specification are hereby incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.

As used herein, the term “about” is used synonymously with the term “approximately.” Illustratively, the use of the term “about” with regard to a certain therapeutically effective pharmaceutical dose indicates that values in a range spanning a cited value, e.g., plus or minus up to 10% of a cited value, are also effective and safe.

As used herein, the terms “comprising,” “including,” “such as,” and “for example” (or “e.g.”) are used in their open, non-limiting sense.

As used herein, the phrase “consisting essentially of” is a transitional phrase used in a claim to indicate that the following list of ingredients, parts or process steps must be present in the claimed composition, machine or process, but that the claim is open to unlisted ingredients, parts or process steps that do not materially affect the basic and novel properties of the invention.

“Nominal dose,” as used herein, refers to the loaded dose, which is the amount of active pharmaceutical ingredient (API) in an inhalation device prior to administration to the subject. The volume of solution containing the nominal dose is referred to as the “fill volume.”

“AUClast” as used herein refers to the area under the curve from time zero to time of last measurable concentration of active pharmaceutical ingredient (API).

“AUClastHEN” as used herein refers to the area under a blood plasma concentration curve up to the last time point for the nominal dose of active pharmaceutical ingredient (API) administered with a nebulizer.

“AUClastConv” as used herein refers to the area under a blood plasma concentration curve up to the last time point for a nominal dose of active pharmaceutical ingredient (API) administered with a conventional inhalation device.

“AUC(0-∞)” as used herein refers to the total area under a blood plasma concentration curve for an active pharmaceutical ingredient (API).

“AUC(0-∞)HEN” as used herein refers to the total area under a blood plasma concentration curve for a nominal dose of active pharmaceutical ingredient (API) administered with a nebulizer.

“AUC(0-∞)CONV” as used herein refers to the total area under a blood plasma concentration curve for a nominal dose of active pharmaceutical ingredient (API) administered with a conventional inhalation device.

AUC(0-∞) can be determined by methods known to those of skill in the art. For example, the AUC(0-∞) of an API can be determined by collecting blood samples from a subject at various time points after administration of an API to the subject, separating plasma from the blood samples, extracting the API from the separated plasma samples, e.g., by solid-phase extraction, quantifying the amount of the API extracted from each sample of separated plasma, e.g., by liquid chromatography-tandem mass spectrometry (LC-MS/MS), plotting the concentration of API in each sample versus the time of collection after administration, and calculating the area under the curve.

“Substantially the same nominal dose” as used herein means that a first nominal dose of an active pharmaceutical ingredient (API) contains approximately the same number of millimoles of the cromolyn or a pharmaceutically-acceptable salt thereof as a second nominal dose of the cromolyn or a pharmaceutically-acceptable salt thereof.

“Bioavailability” as used herein refers to the amount of unchanged API that reaches the systemic circulation, expressed as a percentage of the dosage of the API that is administered to a subject. By definition, the bioavailability of an intravenous solution containing the active pharmaceutical ingredient (API) is 100%. The bioavailability of an API can be determined by methods known to those of skill in the art. For example, the bioavailability of an API can be determined by collecting urine samples from a subject at various time points following administration of the API to the subject, extracting the API from the urine samples, e.g., by solid-phase extraction, quantifying the amount of the API in each urine sample, adjusting the amount of API collected from the urine by a factor based on the amount of API reaching systemic circulation that is excreted in the urine, and calculating the percentage of the API administered to the subject that reaches the systemic circulation of the subject. In a specific embodiment, the bioavailability of cromolyn sodium can be determined as described in Walker et al., 24 J. Pharm. Pharmacol. 525-31 (1972). In the case of cromolyn sodium, the amount of the compound isolated from the urine is multiplied by two to calculate the total amount reaching systemic circulation after administration because the compound is known to be excreted unmetabolized in equal parts in the urine and feces, i.e., approximately 50% of the amount of cromolyn sodium that reaches systemic circulation is excreted in the urine and approximately 50% of the amount of cromolyn sodium that reaches systemic circulation is excreted in the feces.

“Enhanced lung deposition” as used herein refers to an increase in drug deposition (deposited lung dose) arising out of, for example, improved efficiency of drug delivery.

“Deposited dose” or “deposited lung dose” is the amount of cromolyn or a pharmaceutically-acceptable salt thereof deposited in the lung. The deposited dose or deposited lung dose may be expressed in absolute terms, for example in mg or μg of API deposited in the lungs. The deposited lung dose may also be expressed in relative terms, for example calculating the amount of API deposited as a percentage of the nominal dose.

“Cmax” as used herein refers to the maximum plasma concentration for an active pharmaceutical ingredient (API).

“CmaxHEN” as used herein refers to the maximum blood plasma concentration for a nominal dose of the active pharmaceutical ingredient (API) administered with a nebulizer.

“CmaxCONV” as used herein refers to the maximum blood plasma concentration for a nominal dose of the active pharmaceutical ingredient (API) administered with a conventional inhalation device.

Cmax can be determined by methods known to those of skill in the art. For example, the Cmax of an API can be determined by collecting blood samples from a subject at various time points after administration of an API to the subject, separating plasma from the blood samples, extracting the API from the separated plasma samples, e.g., by solid-phase extraction, quantifying the amount of the API extracted from each sample of separated plasma, e.g., by LC-MS/MS, plotting the concentration of API in each sample versus the time of collection after administration, and identifying the peak concentration of the API on the curve.

“Enhanced pharmacokinetic profile” means an improvement in some pharmacokinetic parameter. Pharmacokinetic parameters that may be improved include AUC (0-4 or 0-6 or 0-8 h), AUClast, AUC(0-∞), Tmax, T1/2, and Cmax. In certain embodiments, the enhanced pharmacokinetic profile may be measured quantitatively by comparing a pharmacokinetic parameter obtained for a nominal dose of an active pharmaceutical ingredient (API) administered by one route of administration, such as an inhalation device (e.g., a nebulizer) with the same pharmacokinetic parameter obtained with the same nominal dose of active pharmaceutical ingredient (API) administered by a different route of administration, such as a different type of inhalation device or an oral formulation (e.g., oral tablet, oral capsule, or oral solution).

“Blood plasma concentration” refers to the concentration of an active pharmaceutical ingredient (API) in the plasma component of blood of a subject or subject population.

“Subject” or “subject” refers to the animal (especially mammal) or human being treated.

A “subject group” or “subject group” has a sufficient number of subjects or subjects to provide a statistically significant average measurement of the relevant pharmacokinetic parameter. All members of the “subject group” or “subject group” have pharmacokinetic parameters for the cromolyn or a pharmaceutically-acceptable salts thereof that fall within statistically normal ranges (i.e., there are no outliers), and no member is included on the basis of non-standard or unusual measurements.

“Nebulizer,” as used herein, refers to a device that turns medications, compositions, formulations, suspensions, and mixtures, etc. into a fine aerosol mist for delivery to the lungs.

“Drug absorption” or simply “absorption” typically refers to the process of movement of drug from site of delivery of a drug across a barrier into a blood vessel or the site of action, e.g., a drug being absorbed via the pulmonary capillary beds of the alveoli into the systemic circulation.

“Tmax” as used herein refers to the amount of time necessary for an active pharmaceutical ingredient (API) to attain maximum blood plasma concentration.

“TmaxHEN” as used herein refers to the amount of time necessary for a nominal dose of an active pharmaceutical ingredient (API) to attain maximum blood plasma concentration after administration with a nebulizer.

“TmaxCONV” as used herein refers to the amount of time necessary for a nominal dose of an active pharmaceutical ingredient (API) to attain maximum blood plasma concentration after administration with a conventional inhalation device.

The term “treat” and its grammatical variants (e.g., “to treat,” “treating,” and “treatment”) refer to administration of an active pharmaceutical ingredient to a subject with the purpose of ameliorating or reducing the incidence of one or more symptoms of a condition or disease state in the subject. Such symptoms may be chronic or acute; and such amelioration may be partial or complete. In the present context, treatment entails administering cromolyn or a pharmaceutically-acceptable salt thereof to a subject via any route of administration disclosed herein.

As used herein, the term “high concentration” refers to a concentration greater than 1% by weight. For example, in a specific embodiment, a “high concentration” formulation of cromolyn sodium comprises cromolyn sodium at a concentration of greater than 1% by weight.

As used herein, the term “hypotonic” refers to a formulation that has a tonicity less than 295 mOsm/kg.

The term “prophylaxis” refers to administration of an active pharmaceutical ingredient to a subject with the purpose of reducing the occurrence or recurrence of one or more acute symptoms associated with a disease state or a condition in the subject. In the present context, prophylaxis entails administering cromolyn or a pharmaceutically-acceptable salt thereof to a subject via any route of administration disclosed herein. Thus, prophylaxis includes reduction in the occurrence or recurrence rate of chronic cough due to IPF. However, prophylaxis is not intended to include complete prevention of onset of a disease state or a condition in a subject who has not previously been identified as suffering from chronic cough due to IPF.

As used herein, a “systemically effective amount” is an amount of cromolyn or a pharmaceutically-acceptable salt thereof in the body of a subject as a whole that is effective for the treatment or prophylaxis of chronic cough due to IPF. A “systemically effective amount” may be expressed, for example, as the mass of cromolyn or a pharmaceutically-acceptable salt thereof, or concentration of cromolyn or a pharmaceutically-acceptable salt thereof, in a subject's plasma. A “systemically effective amount” may differ depending on the formulation of cromolyn or a pharmaceutically-acceptable salt thereof.

As used herein, a “locally effective amount” is an amount of cromolyn or a pharmaceutically-acceptable salt thereof in a particular region of the body of a subject that is effective for the treatment or prophylaxis of chronic cough due to IPF. A “locally effective amount” may be expressed, for example, as the mass of cromolyn or a pharmaceutically-acceptable salt thereof, or concentration of cromolyn or a pharmaceutically-acceptable salt thereof, in a subject's tissue. A “locally effective amount” may differ depending on the formulation of cromolyn or a pharmaceutically-acceptable salt thereof.

As used herein, a difference is “significant” if a person skilled in the art would recognize that the difference is probably real. In certain embodiments, significance may be determined statistically, in which case two measured parameters may be referred to as statistically significant. In certain embodiments, statistical significance may be quantified in terms of a stated confidence interval (CI), e.g., greater than 90%, greater than 95%, greater than 98%, etc. In certain embodiments, statistical significance may be quantified in terms of a p value, e.g., less than 0.5, less than 0.1, less than 0.05, etc. The person skilled in the art will recognize these expressions of significance and will know how to apply them appropriately to the specific parameters that are being compared.

EXAMPLES Example 1: Safety and Tolerability of Inhaled PA101 in IPF Subjects with Chronic Cough

As shown in Table 1, PA101 contains 4% (by weight) cromolyn as the active substance, 0.2% sodium chloride as an osmotic agent, 0.02% EDTA as a chelating agent, 1.25% mannitol as a non-ionic osmotic agent, and purified water q.s. PA101 has an osmolality of 200 mOsm/kg. Placebo A contained 0.4% sodium chloride as an osmotic agent, 0.02% EDTA as a chelating agent, 1.25% mannitol as a non-ionic osmotic agent, and purified water q.s., but no cromolyn sodium. The osmolality of Placebo A was adjusted to about 200 mOsm/kg. Placebo B contained 0.6% sodium chloride as an osmotic agent, 0.02% EDTA as a chelating agent, and purified water q.s., but no cromolyn sodium or mannitol. The osmolality of Placebo B was adjusted to about 200 mOsm/kg.

A primary objective of the study was to assess the safety and tolerability of inhaled PA101 (including the excipient mannitol in the formulation) in IPF subjects with refractory chronic cough. A secondary objective of the study was to assess the efficacy potential of inhaled PA101 after 3 days dosing.

The study design was as follows: Phase 1b, randomized, double-blind, single-center, 3-period crossover study in 6 IPF subjects (40-79 years of age) with refractory chronic cough. Each study treatment administered three times daily (TID) for 3 days and one dose the next day (total of 10 doses). 72-hours continuous monitoring for cough count.

The treatments given were one of the following: 1) 40 mg PA101, 2) Placebo-A (A=without cromolyn sodium, but included mannitol), and 3) Placebo-B (B=without mannitol and without cromolyn sodium). All treatments administered as oral inhalation using eFlow nebulizer.

Following administration of the treatment and two placebos to the subjects, any adverse events were recorded. Table 2 provides a summary of adverse events, divided by severity, type, and treatment.

TABLE 2 Adverse Events PA101 Placebo A Placebo B 40 mg (n = 6) (n = 6) (n = 6) Subjects with at 2 (33.3%) 3 (50%) 5 (83.3%) least one AE Related AEs 1 (16.7%) 2 (33.30%) 5 (83.3%) Not related AEs 1 (16.7%) 2 (33.30%) 3 (50%) Mild AEs 2 (33.30%) 3 (50%) 5 (83.3%) Moderate AEs 0 0 1 (16.7%) Severe AEs 0 0 0 Cough 1 (16.7%) 0 4 (66.7%) Throat Irritation 1 (16.7%) 0 3 (50%) Oropharyngeal pain 0 0 1 (16.7%) Rhinorrhoea 0 0 1 (16.7%) Dizziness 1 (16.7%) 2 (33.30%) 2 (33.30%) Headache 0 1 (16.7%) 2 (33.30%) Chills 0 0 1 (16.7%) Malaise 0 1 (16.7%) 0 Flushing 1 (16.7%) 0 1 (16.7%) Defectation urgency 0 0 1 (16.7%) Nausea 1 (16.7%) 1 (16.7%) 0 Nasopharyngitis 1 (16.7%) 0 0

Following administration of the treatment and two placebos to the subjects, the number of daytime coughs was recorded for each subject. FIG. 1 provides a summary of the average number of coughs at three daytime time points for each subject. FIG. 2 provides a summary of the total number of coughs at three daytime time points for each subject.

Whereas the number of coughs provided in FIGS. 1 and 2 are based upon subjective subject reports, the following cough counts are based upon an objective measure. To obtain an objective count of the subjects' coughs, the study used the Leicester Cough Monitor (LCM), a validated 24-h automated cough frequency monitor. The LCM requires the subject to wear a microphone adhered to the subject's chest and attached to a monitor (carried with shoulder strap) that is present on the subject 24 hours each day to record all coughs.

Table 3 provides the LCM count of the average cough per hour for each subject in each treatment condition as well as a breakout of the data across 24 hours, daytime hours, and nighttime hours. SD=Standard Deviation.

TABLE 3 LCM Cough Count Placebo A Placebo B PA101 Mean SD Mean SD Mean SD 24 hr Day 1 25 13 30 18 35 23 Cough/hr Day 2 33 31 32 15 34 21 Day 3 34 28 32 18 32 22 ΔD 3 vs. D 1 8.3 16.7 2.0 5.4 −3.3 11.6 Daytime Day 1 38 20 45 26 53 36 Cough/hr Day 2 48 25 48 22 51 30 Day 3 47 26 46 27 45 30 ΔD 3 vs. D 1 8.6 22.8 1.0 7.3 −8.5 16.2 Nighttime Day 1 4 1 7 6 4 2 Cough/hr Day 2 4 2 5 4 5 2 Day 3 4 4 3 2 6 5 ΔD 3 vs. D 1 0.3 4.5 −4.2 6.6 2.0 3.8

The study includes two additional subjective measures: Cough Severity and Urge-to-Cough, both provided quantitatively as a measure on a visual analogue scale (VAS). When using the visual analogue scale (VAS), for example, to measure cough severity, the subject is asked to mark on a 100 mm scale between ‘no cough’ and ‘the worst cough severity’. When using the visual analogue scale (VAS), for example, to measure urge-to-cough, the subject is asked to mark on a 100 mm scale between ‘no urge’ and ‘the worst urge-to-cough’.

Table 4 provides the mean, standard deviation (SD) and median scores on the VAS for each parameter by treatment at either day 1 or day 4 of the study.

TABLE 4 Parameter (unit) Treatment Visit Mean SD Median Cough Severity Placebo A Day 1 61.7 18.4 62.0 (mm) Day 4 64.0 13.5 58.0 Placebo B Day 1 68.2 11.5 66.5 Day 4 67.3 15.0 72.0 40 mg PA101 Day 1 68.5 10.3 70.5 Day 4 67.0 20.6 72.0 Urge-to-Cough Placebo A Day 1 62.5 16.5 62.0 (mm) Day 4 58.0 19.2 52.0 Placebo B Day 1 69.2 12.1 72.5 Day 4 70.0 14.3 72.5 40 mg PA101 Day 1 70.7 11.4 72.5 Day 4 67.5 20.1 70.0

To assess pulmonary function of each of the subjects following treatment with PA101 or with one of the two placebo formulations, the subjects were evaluated using a forced vital capacity (FVC) test. The Forced Expiratory Volume in One Second (FEV1), the amount of air that is forcefully exhaled in the first second of the FVC test, was measured for each subject either on Day 1 or Day 3 of treatment. FIG. 3 summarizes the results for each treatment group.

The data from this study indicated that treatment with 40 mg PA101 including mannitol as the excipient in the formulation was overall safe and well tolerated following administration three times daily for 3 days in IPF subjects with refractory chronic cough. No difference in tolerability was observed between PA101 formulated with mannitol, placebo with mannitol, and placebo without mannitol. The majority of the adverse events were of mild intensity and did not require treatment. Most commonly reported adverse events were cough, throat irritation, dizziness, and headache. There were no clinically significant changes in cough count, severity of cough and urge to cough between the treatment groups following 3 days of treatment.

Example 2: Phase I Pharmacokinetics, Relative Bioavailability, and Safety Study of PA101 in Healthy Subjects (PK-01)

The primary objective of the study is to determine the systemic availability and pharmacokinetic (PK) profile of single doses of a representative inhaled cromolyn sodium formulation (PA-101) delivered via a nebulizer (eFlow®, PARI) using two different aerosol membranes (30L and 40L) in comparison with marketed formulations of cromolyn sodium (oral solution and an inhalation aerosol) in healthy subjects.

The secondary objective of the study is to assess the safety and tolerability of PA-101 in comparison with marketed formulations of cromolyn sodium (oral solution and an inhalation aerosol).

This was a Phase 1, randomized, open-label, single-centre, dose-ranging, cross-over study conducted in a total of 12 healthy adult subjects of 18-45 years of age.

Study Treatments, Dose and Mode of Administration:

1. 40 mg PA-101 (4% DSCG, 40 mg/1 mL), oral inhalation via eFlow 30L.
2. 80 mg PA-101 (4% DSCG, 80 mg/2 mL), oral inhalation via eFlow 30L.
3. 40 mg PA-101 (4% DSCG, 40 mg/1 mL), oral inhalation via eFlow 40L.
4. 20 mg cromolyn sodium inhalation aerosol (1% DSCG, 20 mg/2 mL) (commercially available product), oral inhalation via LC Plus.
5. 200 mg oral sodium cromoglycate solution (commercially available product), oral administration.

All study subjects received each study treatment in the morning (at 8:00 am, +/−30 minutes) as a single dose treatment. Prior to each dosing day, subjects were admitted to the clinic in the morning for baseline (pre-dose) assessments. Subjects were required to remain in the clinic for 12 h after study drug administration on each dosing day. Treatment Visits were separated by a washout period of 2 to 5 days.

The main delivery device for administering PA-101 was the open system eFlow nebulizer using the 30L aerosol head, which generates aerosol particles with a median size of about 3.0 μm. The duration of the study was one day.

Criteria for Evaluation:

Pharmacokinetic Measurements:

The PK parameters evaluated for plasma cromolyn sodium (DSCG) were maximum concentration (Cmax), time to maximum concentration (Tmax), terminal elimination half-life (T1/2), area under the plasma concentration-time curve from time=0 to time of last measurable drug concentration (AUC0-t), and area under the plasma concentration-time curve from time=0 to infinity (AUC0-∞). Urine DSCG levels were measured for total DSCG excretion in the urine, and the bioavailability of the DSCG was calculated from the measured levels.

Safety measurements: Adverse events including gastrointestinal disturbance (e.g., abdominal pain, nausea, vomiting), changes in vital signs, 12-lead ECG and clinical laboratory tests (hematology, chemistry and urinalysis).

Statistical Measurements:

Pharmacokinetic parameters and plasma concentrations are listed and summarized. The summary statistics are presented as the geometric mean, arithmetic mean, arithmetic standard deviation (SD), min, median, max and n. The geometric statistics are not presented for Tmax. Analysis of variance (ANOVA) including terms for subject and treatment are used to calculate point estimates, and confidence intervals (CI) for treatment differences with respect to PK parameters (90% CI) are calculated.

The incidence of AEs was compared between treatment groups: Summary tables and individual subject listings are provided for all safety measurements and the results are presented by treatment group. Descriptive statistics are used to summarize data where appropriate.

Results:

The pharmacokinetic parameters measured in the single dose study are shown in the following table:

TABLE 5 Inhalation Ratio (PA-101 Ratio (PA-101 Oral aerosol, PA-101 PA-101 PA-101 (30L; 40 mg))/ (30L; 40 mg))/ PK solution, 20 mg (40L), (30L), (30L), (oral solution, (inhalation parameter 200 mg (Intal) 40 mg 40 mg 80 mg 200 mg)) aerosol, 20 mg)) Cmax (ng/mL) 5.2 (±3.1) 17.8 (±10.4) 88.6 (±45.5) 156 (±104) 236 (±124) x30 x8.8 Tmax (h) 3.2 (±2.1) 0.6 (±0.1) 0.6 (±0.1) 0.7 (±0.1) 0.7 (±0.1) AUC0-t 29.4 (±10.4) 39.1 (±15.1) 206 (±94.3) 329 (±144) 514 (±186) x11 x8.4 (h*ng/mL) AUC(0-∞) 33.3 (±11.7) 40.6 (±15.6) 212 (±96.0) 338 (±146) 526 (±198) (h*ng/mL) T1/2 (h) 4.3 (±1.3) 2.5 (±0.8) 2.5 (±0.7) 2.2 (±0.6) 2.1 (±0.5) Bioavailability 0.6 6.5 16.3 25.0 22.7 x42 x3.8 (%)

Values shown in parentheses are (+SD).

Modeling of lung deposition with an aerosol from the 30L and 40L devices using the Finlay model (Finlay, W H, and A R Martin, “Recent advances in predictive understanding respiratory tract deposition”, Journal of Aerosol Medicine, Vol 21:189-205 (2008)) indicated that the lung deposition with the two devices should be very similar. However, the AUC value obtained with 40 mg dose using the 30L device (338 ng*hr/mL) was surprisingly high compared to the value (212 ng*hr/mL) from the 40L device. Cromolyn sodium is not metabolized in the body and is excreted intact via bile and urine. Cromolyn sodium deposited in the lung during inhalation will appear in the plasma, and the AUC would therefore be a surrogate for cromolyn sodium deposited in the lung. Any cromolyn sodium swallowed during inhalation will contribute negligibly to the AUC since the oral bioavailability of cromolyn is only about 1% (Richards et al, J Pharmacol Exp Ther, Vol. 241, No. 3: 1028-1032 (1987)). The AUC data therefore indicate that at the same dose (40 mg), the lung deposition with the 30L device was surprisingly higher than that with the 40L device.

The numbers of adverse events observed in the single dose study are shown in the following table:

TABLE 6 PA-101 PA-101 PA-101 Inhalation Oral (40L), (30 L), (30 L), aerosol, solution, Adverse Event Placebo 40 mg 40 mg 80 mg 20 mg 200 mg Cough 1 1 0 1 1 0 Oropharyngeal pain 0 0 0 0 1 1 Rhinorrhoea 1 0 0 0 0 0 Dizziness 0 0 2 0 0 0 Headache 0 0 0 1 0 1 Dysgeusia 0 0 0 0 0 1 Somnolence 0 0 0 1 0 0 Catheter-site Reaction 0 0 1 0 0 1 Nasopharygitis 0 0 0 0 1 0 Sinusitis 0 0 0 1 0 0 Abdominal Discomfort 0 0 0 0 0 1 Increased Appetite 0 1 0 0 0 0

Table 7 provides a summary of adverse events observed following treatment with PA101 formulations versus placebo or other available cromolyn formulations.

TABLE 7 Part 1 20 mg 200 mg Placebo 40 mg PA 80 mg PA 40 mg PA Intal Nalcrom (30 L) (30 L) (30 L) (40L) (LC Plus) (oral) (N = 12) (N = 12) (N = 12) (N = 12) (N = 12) (N = 12) n (%) n (%) n (%) n (%) n (%) n (%) Any AE 2 (16.7) 3 (25.0) 4 (33.3) 2 (16.7) 3 (25.0) 2 (16.7) Any SAE 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) Probably related AE 0 (0.0) 0 (0.0) 0 (0.0) 1 (8.3) 0 (0.0) 1 (8.3) Possibly related AE 0 (0.0) 2 (16.7) 2 (16.7) 0 (0.0) 0 (0.0) 0 (0.0) Unlikely related AE 1 (8.3) 0 (0.0) 2 (16.7) 1 (8.3) 2 (16.7) 1 (8.3) Not related AE 1 (8.3) 1 (8.3) 0 (0.0) 0 (0.0) 1 (8.3) 1 (8.3) Related AE 0 (0.0) 2 (16.7) 2 (16.7) 1 (8.3) 0 (0.0) 1 (8.3) Not related AE 2 (16.7) 1 (8.3) 2 (16.7) 1 (8.3) 3 (25.0) 2 (16.7) Discontinued due to AE 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0)

Example 3: Phase II Pharmacokinetics, Relative Bioavailability, and Tolerability Study of Three Different PA101 Formulations in Healthy Subjects (PK-02)

As shown in Table 1, PA101 contains 4% (by weight) cromolyn as the active substance, 0.2% Sodium Chloride as an osmotic agent, 0.02% EDTA as a chelating agent, 1.25% mannitol as a non-ionic osmotic agent, and purified water q.s. PA101 has an osmolality of 200 mOsm/kg. PA101-B contains 4% or 6% (by weight) cromolyn as the active substance, 0.2% sodium chloride as an osmotic agent, 0.02% EDTA as a chelating agent, and purified water q.s. PA101-B (40 mg) has an osmolality of 125 mOsm/kg. PA101-B (60 mg) has an osmolality of 135 mOsm/kg.

A primary objective of the study was to evaluate the pharmacokinetics, relative bioavailability, and tolerability three different PA101 formulations in healthy subjects.

The study was designed as a randomized, double-blind, 4 period cross-over study using 12 healthy volunteers, between 18 and 45 years old.

The treatments given were one of the following: 1) 40 mg PA101 (with mannitol), 2) 40 mg PA101B (no mannitol), 3) 60 mg PA101B (no mannitol), and 4) Placebo TID (no mannitol). All treatments administered three times per day (TID) as a single day treatment. Each study treatment separated by a washout period of minimum 24 hrs. All formulations were administered with a Pari eFlow 30L device. The placebo contained 0.2% sodium chloride as an osmotic agent, 0.02% EDTA as a chelating agent, and purified water q.s. The osmolality of the placebo was about 65 mOsm/kg.

Study demographics: 13 total subjects, 5 male and 8 female, having a mean age of 28 years old (total range of 21-40 years old).

Disposition: 13 subjects were randomized. 12 subjects completed the study whereas one subject discontinued during the treatment period 1 (subject was receiving placebo) due to an adverse event (a cough that started 1 minute post-dosing and lasted three minutes).

Table 8 provides a summary of the adverse events observed during this study.

TABLE 8 PA101 PA101-B (40) PA101-B (60) Placebo (N = 12) (N = 12) (N = 12) (N = 13) n (%) n (%) n (%) n (%) Any AE 5 (41.7) 7 (58.3) 5 (41.7) 7 (53.8) Any SAE 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0) Probably related AE 0 (0.0) 2 (16.7) 1 (8.3) 4 (30.8) Possibly related AE 0 (0.0) 0 (0.0) 1 (8.3) 1 (7.7) Unlikely related AE 1 (8.3) 1 (8.3) 2 (16.7) 1 (7.7) Not related AE 5 (41.7) 6 (50.0) 1 (8.3) 2 (15.4) Related AE* 0 (0.0) 2 (16.7) 2 (16.7) 4 (30.8) Not related AE* 5 (41.7) 6 (50.0) 3 (25.0) 3 (23.1) Discontinued due to AE 0 (0.0) 0 (0.0) 0 (0.0) 1 (7.7) Concomitant medication given 2 (16.7) 2 (16.7) 0 (0.0) 0 (0.0) AE of mild intensity 5 (41.7) 7 (58.3) 5 (41.7) 7 (53.8) AE of moderate intensity 2 (16.7) 2 (16.7) 0 (0.0) 2 (15.4) AE of severe intensity 0 (0.0) 0 (0.0) 0 (0.0) 0 (0.0)

Tables 9a and 9b provide an accounting of all adverse events observed during this study.

TABLE 9a PA101 PA101-B (40) PA101-B (60) Placebo (N = 12) (N = 12) (N = 12) (N = 13) System Organ Class Preferred term n (%) n (%) n (%) n (%) Number of subject with at least one TEAE 5 (41.7) 7 (58.3) 5 (41.7) 7 (53.8) GENERAL DISORDERS AND 4 (33.3) 2 (16.7) 2 (16.7) 2 (15.4) ADMINISTRATION SITE CONDITIONS APPLICATION SITE REACTION 1 (8.3) 1 (8.3) 1 (8.3) 0 (0.0) FEELING HOT 0 (0.0) 0 (0.0) 1 (8.3) 0 (0.0) ASTHENIA 0 (0.0) 0 (0.0) 0 (0.0) 1 (7.7) CATHETER SITE PAIN 1 (8.3) 0 (0.0) 0 (0.0) 1 (7.7) CATHETER SITE RELATED REACTION (16.7) 1 (8.3) 0 (0.0) 0 (0.0) FATIGUE 0 (0.0) 0 (0.0) 0 (0.0) 1 (7.7) GASTROINTESTINAL DISORDERS 0 (0.0) 1 (8.3) 0 (0.0) 2 (15.4) ABDOMINAL PAIN UPPER 0 (0.0) 0 (0.0) 0 (0.0) 1 (7.7) DRY MOUTH 0 (0.0) 1 (8.3) 0 (0.0) 0 (0.0) NAUSEA 0 (0.0) 0 (0.0) 0 (0.0) 1 (7.7) INVESTIGATIONS 0 (0.0) 1 (8.3) 0 (0.0) 0 (0.0) SPUTUM ABNORMAL 0 (0.0) 1 (8.3) 0 (0.0) 0 (0.0) MUSCULOSKELETAL AND 1 (8.3) 0 (0.0) 0 (0.0) 0 (0.0) CONNECTIVE TISSUE DISORDERS BACK PAIN 1 (8.3) 0 (0.0) 0 (0.0) 0 (0.0)

TABLE 9b PA101 PA101-B (40) PA101-B (60) Placebo (N = 12) (N = 12) (N = 12) (N = 13) System Organ Class Preferred term n (%) n (%) n (%) n (%) NERVOUS SYSTEM DISORDERS 1 (8.3) 4 (33.3) 3 (25.0) 1 (7.7) DIZZINESS 1 (8.3) 3 (25.0) 2 (16.7) 1 (7.7) HEADACHE 1 (8.3) 2 (16.7) 2 (16.7) 0 (0.0) RESPIRATORY, THORACIC AND 0 (0.0) 3 (25.0) 3 (25.0) 4 (30.8) MEDIASTINAL DISORDERS COUGH 0 (0.0) 0 (0.0) 2 (16.7) 4 (30.8) THROAT IRRITATION 0 (0.0) 1 (8.3) 1 (8.3) 2 (15.4) NASAL CONGESTION 0 (0.0) 1 (8.3) 0 (0.0) 0 (0.0) OROPHARYNGEAL PAIN 0 (0.0) 1 (8.3) 0 0 (0.0) SKIN AND SUBCUTANEOUS (8.3) 0 (0.0) 0 (0.0) 1 (7.7) TISSUE DISORDERS PETECHIAE 1 (8.3) 0 (0.0) 0 (0.0) 0 (0.0) SKIN REACTION 0 (0.0) 0 (0.0) 0 1 (7.7) VASCULAR DISORDERS 1 (8.3) 0 (0.0) 0 (0.0) 0 (0.0) THROMBOPHLEBITIS 1 (8.3) 0 (0.0) 0 (0.0) 0 (0.0)

Table 10 provides a summary of adverse events observed during this study related to administration of PA101 or PA101-B.

TABLE 10 PA101 PA101-B (40) PA101-B (60) Placebo (N = 12) (N = 12) (N = 12) (N = 13) System Organ Class Preferred term n (%) n (%) n (%) n (%) Number of subject with at least one TEAE 0 (0.0) 2 (16.7) 2 (16.7) 4 (30.8) RESPIRATORY, THORACIC AND 0 (0.0) 1 (8.3) 2 (16.7) 4 (30.8) MEDIASTINAL DISORDERS COUGH 0 (0.0) 0 (0.0) 1 (8.3) 4 (30.8) THROAT IRRITATION 0 (0.0) 1 (8.3) 1 (8.3) 2 (15.4) NERVOUS SYSTEM DISORDERS 0 (0.0) 0 (0.0) 1 (8.3) 0 (0.0) DIZZINESS 0 (0.0) 0 (0.0) 1 (8.3) 0 (0.0) GASTROINTESTINAL DISORDERS 0 (0.0) 1 (8.3) 0 (0.0) 1 (7.7) DRY MOUTH 0 (0.0) 1 (8.3) 0 (0.0) 0 (0.0) NAUSEA 0 (0.0) 0 (0.0) 0 (0.0) 1 (7.7)

Table 11 provides a summary of moderate adverse events observed during this study.

TABLE 11 PA101 PA101-B (40) PA101-B (60) Placebo (N = 12) (N = 12) (N = 12) (N = 13) System Organ Class Preferred term n (%) n (%) n (%) n (%) Number of subject with at least one TEAE 2 (16.7) 2 (16.7) 0 (0.0) 2 (15.4) GENERAL DISORDERS AND 0 (0.0) 1 (8.3) 0 (0.0) 1 (7.7) ADMINISTRATION SITE CONDITIONS APPLICATION SITE REACTION 0 (0.0) 1 (8.3) 0 (0.0) 0 (0.0) CATHETER SITE PAIN 0 (0.0) 0 (0.0) 0 (0.0) 1 (7.7) NERVOUS SYSTEM DISORDERS 1 (8.3) 2 (16.7) 0 (0.0) 0 (0.0) HEADACHE 1 (8.3) 2 (16.7) 0 (0.0) 0 (0.0) RESPIRATORY, THORACIC AND 0 (0.0) 0 (0.0) 0 (0.0) 1 (7.7) MEDIASTINAL DISORDERS COUGH 0 (0.0) 0 (0.0) 0 (0.0) 1 (7.7) VASCULAR DISORDERS 1 (8.3) 0 (0.0) 0 (0.0) 0 (0.0) THROMBOPHLEBITIS 1 (8.3) 0 (0.0) 0 (0.0) 0 (0.0)

Pharmacokinetic Results:

Table 12 provides the mean and standard deviation (SD) for each pharmacokinetic parameter measured for each PA101 formulation studied. As used herein, “Ke1”=is the elimination rate constant that describes the rate at which the cromolyn of PA101 or PA101-B formulations is removed from the subject's system. This measure is equivalent to the fraction of cromolyn that is removed per unit of time (T−1, or in this case 1/hours (h)).

TABLE 12 40 mg PA101 40 mg PA101-B 60 mg PA101-B Parameter Mean SD Mean SD Mean SD 1st Dose Cmax, ng/mL 76.8 31.0 75.6 29.1 119 41.0 Tmax, ha 0.56 (0.31-2.04) 0.56 (0.31-2.04) 0.56 (0.13-2.04) AUC0-6, h · ng/mL 229b   96.6b 216b   79.7b  358b 136b  2nd Dose Cmax, ng/mL 84.7 34.7 82.3 32.1 148 60.3 Tmax, ha 0.56 (0.23-2.04) 0.56 (0.13-2.06) 0.56 (0.23-1.04) AUC0-6, h · ng/mL 266b  123b  258b  101b  420 175   3rd Dose Cmax, ng/mL 92.1 30.1 92.9 35.1 157 58.2 Tmax, ha 0.56 (0.23-0.81) 0.56 (0.23-2.04) 0.56 (0.13-0.56) AUC0-t, h · ng/mL 330   142   330   140   529 257   AUC0-inf, h · ng/mL 342   147   340   145   542 262   kel, 1/h   0.281   0.0282   0.294   0.0229     0.306   0.0385 t1/2, h  2.49   0.237  2.37   0.184    2.30   0.265

The pharmacokinetic parameters of the PA101 treatments of the Phase I study (described in Example 2—PK-01) and the Phase II study (described in this Example-PK-02) are compared in Table 13 below. Note that subjects in one of the PK-01, 40 mg group was administered the formulation comprising cromolyn sodium using a Pari eFlow 40L device, while the formulations were administered to all other subjects in the study using a Pari eFlow 30L device. In the PK-01 study there were three subjects whose plasma values were very high compared to the average, and these outlier values skewed the Cmax and AUC results in the PK-01 study. If the data are analyzed by excluding these outliers, the Cmax and AUC results of the PK-01 and PK-02 studies are comparable. This was supported by the finding that the urine cromolyn levels were similar in the two studies.

TABLE 13 PK-01 PK-01 PK-01 PK-02 PK-02 PK-02 PA101 PA101 PA101 PA101 PA101B PA101B Intal Nalcrom 40 mg 40 mg 80 mg 40 mg 40 mg 60 mg 20 mg 200 mg (40L) (30L) (30L) (30L) (30L) (30L) Cmax 17.8 (10.4) 5.2 (3.1) 88.6 (45.5) 156 (104) 236 (104) 76.8 (31.0) 75.6 (29.1) 119 (41.0) (ng/mL) Tmax (h) 0.6 (0.1) 3.2 (2.1) 0.6 (0.1) 0.7 (0.1) 0.7 (0.1) 0.6 (0.3) 0.6 (0.3) 0.6 (0.1) AUC0-t 39.1 (15.1) 29.4 (10.4) 206 (94.3) 329 (144) 514 (186) 229 (97) 216 (80) 358 (136) (h · ng/mL) AUC0-∞ 40.6 (15.6) 33.3 (11.7) 212 (96.0) 338 (146) 526 (198) (h · ng/mL) T1/2(h) 2.5 (0.8) 4.3 (1.3) 2.5 (0.7) 2.2 (0.6) 2.1 (0.5)

PA101 formulations from the Phase I and Phase II studies are safe and well-tolerated. The most common adverse events, reported in at least 2 subjects, include cough, throat irritation, dizziness, headache and catheter-site reaction. Treatment-related adverse events include cough, throat irritation, dizziness, dry mouth and nausea. Both the frequency and severity of adverse events are comparable between active and placebo treatments, which was unexpected given that the PA101-B formulations (without mannitol) exhibited osmolalities that were significantly different than formulations comprising mannitol. Accordingly, the majority of adverse events have a mild intensity and transient duration. Thus, the PA101-B formulations (at both 40 mg and 60 mg dosages) are well-tolerated with an adverse event (AE) profile similar to PA101.

PA101-B formulations (at both 40 mg and 60 mg dosages) have a comparable pharmacokinetic profile to PA101.

Example 4: Phase II Safety and Efficacy Study of Cromolyn Formulations in Subjects with Chronic Cough Due to Idiopathic Pulmonary Fibrosis (IPF)

This is a randomized, double-blind, placebo-controlled, 2-period crossover, 2-cohort, multi-center, Phase 2 study in 48 subjects with chronic cough: 24 subjects exhibiting chronic cough associated with idiopathic pulmonary fibrosis (IPF, Cohort 1) and 24 subjects with chronic idiopathic cough not associated with IPF (CIC, Cohort 2) (see Example 5).

The study consists of two treatment periods of 14 days each separated by a Washout Period of 14 days (+2 days) between Period 1 and Period 2. A Screening Visit will be conducted within 14 days before the Baseline Visit of Period 1. The two periods are identical except that in Period 2, subjects are crossed over to the alternate treatment from that received in Period 1, according to a 1:1 randomization scheme. At the Screening Visit subjects with a daytime cough severity score >40 mm using a linear 100 mm visual analogue scale are placed on 24-hour objective cough count monitoring using the LCM cough monitor. Subjects with an average daytime cough count of at least 15 coughs per hour using LCM at the Screening Visit are eligible for randomization. During each period, subjects self-administer study drug (i.e., 40 mg PA101 or Placebo PA101 via eFlow) three times daily (i.e., 8:00 am±1 hour, 2:00 pm±1 hour, and 8:00 pm±1 hour) for 14 consecutive days of each period (e.g., Days 1-14). Subjects attend a Pre-study Visit (Visit 1, Day −1) at the clinic in the morning prior to the Baseline/Treatment Visit (Visit 2, Day 1) and a cough count device (LCM) is dispensed for measurement of baseline 24-hour cough count. Subjects return to the clinic next day in the morning (Visit 2, Day 1) to return the devices, assessment of quality of life measures, and to receive the first dose of the study treatment. Additional treatment visits during the Treatment Period occur on Day 7±1 day (Visit 3) and Day 15±1 day (Visit 5). Subjects visit the clinic on Day 7±1 day (Visit 3) and Day 14±1 day (Visit 4) in the morning and the LCM device is dispensed for measurement of 24-hour cough count. Study assessments include assessment of quality of life (LCQ and K-BILD), cough severity (VAS), pulmonary function tests (forced expiratory volume in one second [FEV1], forced vital capacity [FVC], and FEV1/FVC ratio), fraction of exhaled nitric oxide (FeNO), and safety assessments (AEs, vital signs, and ECG) on Days 1, 7 and 15 of each treatment period.

Pulmonary function tests and K-BILD assessment are only be performed in the IPF cohort.

A safety follow-up call is placed within 7±2 days following the last study treatment. Clinical safety laboratory samples are collected at the start and end of the treatment of each treatment period (Screening Visit and Visit 5 during the Treatment Period 1, and at Visit 2 and Visit 5 during the Treatment Period 2). All post-dose study procedures are conducted from time 0. Time 0 is defined as the start of the first study drug administration (i.e., when the nebulizer has been turned on) of each period.

In the IPF cohort, subjects are allowed to use antifibrotic therapy i.e., pirfenidone, nintedanib, and N-acetylcysteine) during the course of the study provided that the dose is stabilized at least 3 months prior to the Screening visit and throughout the study period. Subjects are not allowed to use the following drugs (i.e., prednisone, narcotic antitussives, baclofen, gabapentin, inhaled corticosteroids, benzonatate, dextromethorphan, carbetapentane, and H1 antihistamines, leukotriene modifiers, and cromolyn sodium) for at least 2 weeks prior to the Screening Visit and throughout the study. Drugs containing bronchodilators (including beta-2 agonists and anticholinergics) are not allowed for at least 1 week prior to the Baseline Visit and during the study.

As shown in Table 1, PA101 contains 4% (by weight) cromolyn as the active substance, 0.2% Sodium Chloride as an osmotic agent, 0.02% EDTA as a chelating agent, 1.25% mannitol as a non-ionic osmotic agent, and purified water q.s. PA101 has an osmolality of 200 mOsm/kg.

A primary objective of the study was to assess the effectiveness of inhaled PA101 delivered via a Pari eFlow 30L nebulizer for treating chronic cough due to IPF.

The study design is provided graphically in FIG. 4.

Inclusion Criteria: 1) male or female subjects age 40 through 79 years, inclusive of the endpoints, 2) diagnosis of Idiopathic Pulmonary Fibrosis (IPF) with the consensus of the multidisciplinary team based on the presence of definitive or possible usual interstitial pneumonia (UIP) pattern on high-resolution computed tomography (HRCT) and after excluding alternative diagnoses, including lung diseases associated with environmental and occupational exposure, with connective tissue diseases and with drugs, 3) chronic cough present for at least 8 weeks and not responsive to current therapies, 4) daytime cough severity score on visual analogue scale >40 mm at the Screening Visit, 5) daytime average cough count of at least 15 coughs per hour using objective cough count monitor at the Screening Visit, 6) transfer capacity for carbon monoxide corrected for hemoglobin (TLCOc) >25% predicted value within 12 months of the Screening Visit and Forced Vital Capacity (FVC) >50% predicted value within 1 month of the Screening Visit, and 7) willingness and ability to provide written informed consent.

Exclusion Criteria: 1) current or recent history of clinically significant medical condition, laboratory abnormality, or illness that could put the subject at risk or compromise the quality of the study data as determined by the investigator, 2) significant coronary artery disease (i.e., myocardial infarction within 6 months or unstable angina within 1 month of the Screening Visit), 3) an upper or lower respiratory tract infection within 4 weeks of the Screening Visit, 4) acute exacerbation of IPF within 3 months of the Screening Visit, 5) long-term daily oxygen therapy (>10 hours/day), 6) presence of pulmonary arterial hypertension with limitation of activity, 7) history of malignancy of any organ system, treated or untreated within the past 5 years, with the exception of localized basal cell carcinoma or cervix carcinoma in situ, 8) current or recent history (previous 12 months) of excessive use or abuse of alcohol, 9) current or recent history (previous 12 months) of abusing legal drugs or use of illegal drugs or substances, 10) participation in any other investigational drug study within 4 weeks prior to the Screening Visit, 11) use of the following drugs within 2 weeks of the Screening Visit: Prednisone, narcotic antitussives, baclofen, gabapentin, inhaled corticosteroids, benzonatate, dextromethorphan, carbetapentane, H1 antihistamines, leukotriene modifiers, and cromolyn sodium, 12) females who are pregnant or breastfeeding, or if of child-bearing potential unwilling to practice acceptable means of birth control or abstinence during the study (e.g., abstinence, combination barrier and spermicide, or hormonal), and 13) history of hypersensitivity or intolerance to cromolyn sodium.

Table 14 provides the demographics of the study participants.

TABLE 14 Number (N) 24 Age 67 ± 6 (56-79 years) Sex 15 Male/9 Female Race 2 Asian/22 White Body Mass Index (BMI) 29.1 ± 3.8  Time since IPF diagnosis 4.1 ± 3.3 (1-13 years) Time since Cough diagnosis 5.6 ± 4.2 (1-16 years) Forced Vital Capacity (FVC) - Mean (L) 2.54 ± 0.96 Forced Vital Capacity (FVC) - Predicted (%) 73 ± 15 FEV1/FVC (%) 83 ± 9  FEV1 = Volume that has been exhaled at the end of the first second of forced expiration

Safety Results:

Table 15 below summarizes the treatment emergent adverse events (AEs) reported for at least two subjects having chronic cough due to IPF.

TABLE 15 Adverse Events (AEs) PA101 (n = 23) Placebo (n = 23) Subjects with at least one 14 (61%) 14 (61%) Adverse Event (AE) Severe AEs 0 0 AEs leading to withdrawal 1 (4.3%) 1 (4.3%) Related AEs 11 (48%) 9 (39%) Unrelated AEs 3 (13%) 5 (22%) Mild AEs 11 (48%) 9 (39%) Moderate AEs 3 (13%) 5 (22%) Severe AEs 0 0 Respiratory System Cough 3 (13%) 4 (17.4%) Dyspnoea 1 (4.3%) 2 (8.7%) Exertional dyspnoea 0 1 (4.3%) Dysphonia 0 1 (4.3%) Hyperventilation 0 1 (4.3%) Idiopathic pulmonary fibrosis 0 1 (4.3%) Oropharyngeal pain 1 (4.3%) 0 Increased sputum 0 1 (4.3%) Wheezing 1 (4.3%) 0 Other Systems Headache 3 (13%) 2 (8.7%) Dizziness 1 (4.3%) 2 (8.7%) Diarrhea 2 (8.7%) 1 (4.3%) Defecation urgency 2 (8.7%) 0 Dry mouth 2 (8.7%) 0 Fatigue 1 (4.3%) 2 (8.7%) Flushing 2 (8.7%) 0

There were two participants who withdrew from the study. One participant who received the placebo treatment experienced increased shortness of breath & cough (mild, possibly related) starting on Day 1, lasting 13 days, at which time the symptoms resolved. The other participant received the PA101 treatment and contracted the common cold (moderate, unlikely to be related to the treatment), starting on day 5 and lasting 17 days, at which time the symptoms resolved.

Efficacy Results:

Cough count was measured by Leicester Cough Monitor (LCM). FIG. 5 provides a summary of the daytime average cough/hour in subjects with chronic cough due to IPF receiving either PA101 or a placebo at baseline, Day 7, and Day 14 of the study. When the change from baseline is analyzed (see FIG. 6), a statistically significant difference emerges at Days 7 and 14 for the PA101-treated subjects. To demonstrate the clinical relevance of this result, this statistically significant difference can be expressed as both a placebo-adjusted (PBO-adjusted) reduction in cough frequency (see FIG. 7) and a least squares (LS) means ratio with a 95% confidence interval (CI) (see FIGS. 7 and 8). Taken together, the data indicate that treatment with PA101 provides a reduction in cough that is not only statistically significant but also clinically meaningful.

Table 16 summarizes the data shown graphically in FIGS. 7 and 8.

TABLE 16 Treatment PA101 Placebo (N = 23) (N = 23) Day 7 Number of subjects included in 23 (100) 21 (91.3) analysis (n) (%) Adjusted Least squares 0.78 1.08 treatment mean estimate mean 95% CI 0.64, 0.96 0.87, 1.33 (LSMean) Treatment Ratio of means 0.73 Comparison p-value  0.0329 95% CI 0.54, 0.97 Day 14/ Number of subjects included in 23 (100) 21 (91.3) ET analysis (n) (%) Adjusted Least squares 0.70 1.05 treatment mean estimate mean 95% CI 0.55, 0.90 0.81, 1.36 (LSMean) Treatment Ratio of means 0.67 Comparison p-value  0.0241 95% CI 0.48, 0.94

Table 17 summarizes the data shown graphically in FIGS. 7 and 8.

TABLE 17 PA101 (n = 23) Placebo (n = 21) Mean ± SD Mean ± SD Baseline 54.7 ± 54.5 50.9 ± 36.5 Day 7 38.1 ± 24.7 56.5 ± 42.3 Change from Baseline −16.4 ± 50.5 (−30%) 5.5 ± 28.1 (11%) LS Mean Ratio (95% CI) 0.73 (0.54, 0.97) p = 0.03 Day 14 38.8 ± 29.0 51.9 ± 39.9 Change from Baseline −16.2 ± 44.9 (−29%) 1.3 ± 17.1 (2%)  LS Mean Ratio (95% CI) 0.67 (0.48-0.94) p = 0.02

FIG. 12 provides a summary of the nighttime average cough/hour in subjects with chronic cough due to IPF receiving either PA101 or a placebo at baseline, Day 7, and Day 14 of the study. Similar to the analysis performed for daytime cough, FIG. 13 provides the change from baseline for nighttime cough at Days 7 and 14 for the PA101-treated subjects.

Table 18 summarizes the data shown graphically in FIGS. 12 and 13.

TABLE 18 Treatment PA101 Placebo (N = 23) (N = 23) Day 7 Number of subjects included in 21 (91.3)  20 (87.0) analysis (n) (%) Adjusted Least squares 0.70 1.31 treatment mean estimate mean 95% CI 0.43, 1.15 0.79, 2.18 (LSMean) Treatment Ratio of means 0.53 Comparison p-value  0.0122 95% CI 0.33, 0.85 Day 14/ Number of subjects included in 23 (100.0) 20 (87.0) ET analysis (n) (%) Adjusted Least squares 0.60 0.93 treatment mean estimate mean 95% CI 0.33, 1.10 0.48, 1.81 (LSMean) Treatment Ratio of means 0.64 Comparison p-value  0.3022 95% CI 0.26, 1.57

To demonstrate clinical relevance of the change in 24-hour average cough/hour in PA101-treated subjects, this difference can be expressed as both a placebo-adjusted (PBO-adjusted) reduction in cough frequency (see FIGS. 15 and 17) and a least squares (LS) means ration with a 95% confidence interval (CI) (see FIGS. 16 and 17). Taken together, the data indicate that treatment with PA101 provides a reduction in 24-hour average cough/hour that is clinically meaningful.

Table 19 summarizes the data shown graphically in FIGS. 15-17.

TABLE 19 Treatment PA101 Placebo (N = 23) (N = 23) Day 7 Number of subjects included in 23 (100.0) 21 (91.3) analysis (n) (%) Adjusted Least squares 0.75 1.05 treatment mean estimate mean 95% CI 0.62, 0.91 0.86, 1.29 (LSMean) Treatment Ratio of means 0.71 Comparison p-value  0.0163 95% CI 0.54, 0.94 Day 14/ Number of subjects included in 23 (100.0) 21 (91.3) ET analysis (n) (%) Adjusted Least squares 0.71 1.04 treatment mean estimate mean 95% CI 0.56, 0.90 0.81, 1.34 (LSMean) Treatment Ratio of means 0.68 Comparison p-value  0.0245 95% CI 0.49, 0.95

FIG. 18 synthesizes the data from daytime, nighttime and 24-hour cough/hour analyses to generate a 24-hour cough profile for subjects in this study with chronic cough due to IPF who receive either placebo or PA101. For each treatment group, the average cough/hour rates over a 24-hour window are drawn contrasting baseline rates to those rates demonstrated following 14 days of administration of either the placebo or PA101. The PA101-treated group shows a statistically-significant and clinically relevant reduction in cough/hour rates by Day 14, particularly from the hours of 8 am to 11 pm.

In addition to the objective cough/hour data captured using the LCM, subjective data was collected from study subjects. A quality of life score was measured by the Leicester Cough Questionnaire (LCQ) and King's Brief Interstitial Lung Disease Questionnaire (K-BILD).

Table 20 summarizes the data collected using the Leicester Cough Questionnaire to assess Quality of Life changes from baseline to Days 7 and 14. Quality of Life was a secondary endpoint of this Phase II study. LCQ: worst score=3, best score=21; MID: >1.3.

TABLE 20 PA101 (n = 23) Placebo (n = 21) Mean ± SD Mean ± SD Baseline 13.6 ± 3.6 13.7 ± 3.5 Day 7 14.4 ± 3.2 14.0 ± 3.3 Change from Baseline 0.7 0.3 Day 14 14.8 ± 3.3 13.4 ± 3.7 Change from Baseline 1.1 0   LS Mean Difference 1.1 p = 0.09

Table 21 summarizes the change from baseline in the Leicester Cough Questionnaire measuring Quality of Life.

TABLE 21 Treatment PA101 Placebo (N = 23) (N = 23) Day 7 Number of subjects included in 23 (100.0) 22 (95.7)  analysis (n) (%) Adjusted Least squares 0.66  0.33 treatment mean estimate mean 95% CI −0.06, 1.38 −0.41, 1.07 (LSMean) Treatment Ratio of means 0.33 Comparison p-value  0.5057 95% CI −0.70, 1.37 Day 14/ Number of subjects included in 23 (100.0) 23 (100.0) ET analysis (n) (%) Adjusted Least squares 1.04 −0.03 treatment mean estimate mean 95% CI  0.16, 1.93 −0.92, 0.85 (LSMean) Treatment Ratio of means 1.08 Comparison p-value  0.0905 95% CI −0.18, 2.33

Table 22 summarizes the cough specific Quality of Life in Responders. Responders are defined herein as those subjects with chronic cough due to IPF who experience a greater than 30% reduction in the average daytime cough following treatment with a composition of the disclosure, or in this study, PA101.

TABLE 22 PA101 (n = 8) Placebo (n = 4) Mean ± SD Mean ± SD Baseline 14.5 ± 4.0 14.2 ± 2.9 Day 7 15.6 ± 3.2 13.6 ± 3.1 Change from Baseline 1.1 −0.6 Day 14 16.8 ± 2.8 14.4 ± 3.3 Change from Baseline 2.3  0.2

Table 23 summarizes the data collected using the King's Brief Interstitial Lung Disease Questionnaire (K-BILD) to assess Quality of Life changes from baseline to day 14. Disease-specific quality of life was a secondary efficacy endpoint of this Phase II study. LCQ: worst score=15, best score=100.

TABLE 23 PA101 (n = 23) Placebo (n = 23) Mean ± SD Mean ± SD MID Baseline - Total Score 55.4 ± 9.2  57.0 ± 11.7 Psychological 55.5 ± 13.0 57.7 ± 13.3 Breathlessness & Activities 44.1 ± 15.2 45.0 ± 20.9 Chest 61.8 ± 19.9 63.5 ± 21.7 Day 14 - Total Score 56.7 ± 9.1  55.1 ± 10.0 Psychological 57.9 ± 13.4 55.0 ± 13.7 Breathlessness & Activities 44.3 ± 18.7 44.5 ± 17.9 Chest 69.3 ± 14.3 60.8 ± 19.3 Change from Baseline 1.3 ± 4.1 −1.9 ± 7.5  (Mean ± SD) Psychological 2.4 ± 6.3 −2.7 ± 10.2 Breathlessness & Activities  0.2 ± 10.1 −0.5 ± 11.0 Chest 7.4 ± 9.9 −2.7 ± 18.2 LS Mean Difference (95% CI) Total 2.2 (−0.8, 5.2) (p = 0.11) 5 Psychological  3.9 (0.4, 7.4) (p = 0.03)* 6 Breathlessness & Activities 0.5 (−7.2, 8.2) (p = 0.88) 7 Chest  8.7 (1.4, 16.0) (p = 0.03)* 11

Table 24 summarizes the change from baseline in K-BILD psychological domain measuring Quality of Life using a mixed score model in subjects with chronic cough due to IPF treated with either placebo or PA101.

TABLE 24 Treatment PA101 Placebo (N = 23) (N = 23) Day 7 Number of subjects included in 23 (100.0) 22 (95.7)  analysis (n) (%) Adjusted Least squares 2.36 −0.94 treatment mean estimate mean 95% CI −0.63, 5.35 −3.99, 2.11 (LSMean) Treatment Ratio of means 3.30 Comparison p-value  0.0783 95% CI −0.41, 7.00 Day 14/ Number of subjects included in 23 (100.0) 23 (100.0) ET analysis (n) (%) Adjusted Least squares 1.34 −2.55 treatment mean estimate mean 95% CI −2.46, 5.14 −6.37, 1.26 (LSMean) Treatment Ratio of means 3.89 Comparison p-value  0.0322 95% CI 0.37, 7.40

Table 25 summarizes the change from baseline in K-BILD breathlessness and activities domain measuring Quality of Life using a mixed score model in subjects with chronic cough due to IPF treated with either placebo or PA101.

TABLE 25 Treatment PA101 Placebo (N = 23) (N = 23) Day 7 Number of subjects included in 23 (100.0) 22 (95.7)  analysis (n) (%) Adjusted Least squares −1.02   2.01 treatment mean estimate mean 95% CI −4.74, 2.70 −1.79, 5.80 (LSMean) Treatment Ratio of means −3.02  Comparison p-value  0.1627 95% CI −7.43, 1.38 Day 14/ Number of subjects included in 23 (100.0) 23 (100.0) ET analysis (n) (%) Adjusted Least squares 0.08 −0.41 treatment mean estimate mean 95% CI −4.39, 4.55 −4.88, 4.07 (LSMean) Treatment Ratio of means 0.49 Comparison p-value  0.8794 95% CI −7.24, 8.21

Table 26 summarizes the change from baseline in K-BILD chest symptoms domain measuring Quality of Life using a mixed score model in subjects with chronic cough due to IPF treated with either placebo or PA101.

TABLE 26 Treatment PA101 Placebo (N = 23) (N = 23) Day 7 Number of subjects included in 23 (100.0) 22 (95.7)  analysis (n) (%) Adjusted Least squares 3.47  2.58 treatment mean estimate mean 95% CI −1.36, 8.31 −2.33, 7.50 (LSMean) Treatment Ratio of means 0.89 Comparison p-value  0.7129 95% CI −4.19, 5.98 Day 14/ Number of subjects included in 23 (100.0) 23 (100.0) ET analysis (n) (%) Adjusted Least squares 6.26 −2.42 treatment mean estimate mean 95% CI  0.77, 11.75 −7.92, 3.08 (LSMean) Treatment Ratio of means 8.68 Comparison p-value  0.0270 95% CI  1.38, 15.99

Table 27 summarizes the change from baseline in K-BILD total score measuring Quality of Life using a mixed score model in subjects with chronic cough due to IPF treated with either placebo or PA101.

TABLE 27 Treatment PA101 Placebo (N = 23) (N = 23) Day 7 Number of subjects included 23 (100.0) 22 (95.7)  in analysis (n) (%) Adjusted Least squares 0.37  0.06 treatment mean estimate mean 95% CI −1.61, 2.35 −1.96, 2.08 (LSMean) Treatment Ratio of means 0.31 Comparison p-value  0.7655 95% CI −1.85, 2.46 Day 14/ Number of subjects included 23 (100.0) 23 (100.0) ET in analysis (n) (%) Adjusted Least squares 0.49 −1.73 treatment mean estimate mean 95% CI −2.22, 3.20 −4.44, 0.99 (LSMean) Treatment Ratio of means 2.22 Comparison p-value  0.1189 95% CI −0.76, 5.19

Table 28 summarizes the data from measuring cough severity by VAS in subjects with chronic cough due to IPF treated with either placebo or PA101. Cough severity was measured by Visual Analog Scale (VAS). When using the visual analogue scale (VAS), for example, to measure cough severity, the subject is asked to mark on a 100 mm scale between ‘no cough’ and ‘the worst cough severity’. VAS: worse score=100, best score=0

TABLE 28 PA101 (n = 23) Placebo (n = 21) Mean ± SD Mean ± SD Baseline 54.8 ± 20.6 55.3 ± 23.8 Day 7 52.5 ± 20.7 51.2 ± 26.5 Change from Baseline  −2.3 −3.4 Day 14 44.3 ± 26.7 53.3 ± 26.6 Change from Baseline −10.3 −1.8 LS Mean Difference −8.5 p = 0.14

Table 29 summarizes the change from baseline in cough severity by VAS in subjects with chronic cough due to IPF treated with either placebo or PA101.

TABLE 29 Treatment PA101 Placebo (N = 23) (N = 23) Day 7 Number of subjects 23 (100.0) 22 (95.7)  included in analysis (n) (%) Adjusted Least squares −2.27   −3.38 treatment mean estimate mean 95% CI −8.15, 3.61 −9.38, 2.63 (LSMean) Treatment Ratio of means 1.11  Comparison p-value 0.7914 95% CI −7.30, 9.51 Day 14/ Number of subjects 23 (100.0) 23 (100.0) ET included in analysis (n) (%) Adjusted Least squares −10.31   −1.84 treatment mean estimate mean 95% CI −18.22, −2.41 −9.74, 6.06 (LSMean) Treatment Ratio of means −8.48   Comparison p-value 0.1331 95% CI −19.65, 2.70 

Table 30 summarizes the data from measuring cough severity by VAS in Responders.

TABLE 30 PA101 (n = 8) Placebo (n = 4) Mean ± SD Mean ± SD Baseline 57.3 ± 17.2 69.0 ± 17.2 Day 7 45.3 ± 14.5 64.0 ± 20.6 Change from Baseline −12  −5 Day 14 26.6 ± 14.2 56.3 ± 12.8 Change from Baseline −31 −13

The LCQ and VAS scores in Responders were compared (see FIG. 19). The data indicate a significant difference in VAS scores in Responders treated with PA101 between baseline and day 14. This subjective measure of cough severity corroborates the objective measure of cough frequency measured by using a cough monitor. Both methods demonstrate a clinically relevant reduction in symptoms of chronic cough due to IPF following 14 days of treatment with PA101.

This phase II study confirmed that PA101 has a very good safety profile. Moreover, the data from this study demonstrate a statistically significant 30% reduction in the objective cough count vs. placebo. The data indicate positive trends in cough-specific quality of life (QoL) measurements (LCQ) and subject-reported cough severity (VAS). The data also demonstrate a minimal placebo effect, which is clinically relevant for future trials in IPF-chronic cough.

Example 5: Phase II Safety and Efficacy Study of Cromolyn Formulations in Subjects with Chronic Idiopathic Cough

This is a randomized, double-blind, placebo-controlled, 2-period crossover, 2-cohort, multi-center, Phase 2 study in 48 subjects with chronic cough: 24 subjects with idiopathic pulmonary fibrosis (IPF, Cohort 1) (see Example 4) and 24 subjects with chronic idiopathic cough (CIC, Cohort 2).

The study consists of two treatment periods of 14 days each separated by a Washout Period of 14 days (+2 days) between Period 1 and Period 2. A Screening Visit will be conducted within 14 days before the Baseline Visit of Period 1. The two periods are identical except that in Period 2, subjects were crossed over to the alternate treatment from that received in Period 1, according to a 1:1 randomization scheme. At the Screening Visit subjects with a daytime cough severity score >40 mm using a linear 100 mm visual analogue scale are placed on 24-hour objective cough count monitoring using the LCM cough monitor. Subjects with an average daytime cough count of at least 15 coughs per hour using LCM at the Screening Visit are eligible for randomization. During each period, subjects self-administer study drug (i.e., 40 mg PA101 or Placebo PA101 via a Pari eFlow 30L device) three times daily (i.e., 8:00 am±1 hour, 2:00 pm±1 hour, and 8:00 pm±1 hour) for 14 consecutive days of each period (e.g., Days 1-14). Subjects attend a Pre-study Visit (Visit 1, Day −1) at the clinic in the morning prior to the Baseline/Treatment Visit (Visit 2, Day 1) and a cough count device (LCM) is dispensed for measurement of baseline 24-hour cough count. Subjects return to the clinic next day in the morning (Visit 2, Day 1) to return the devices, assessment of quality of life measures, and to receive the first dose of the study treatment. Additional treatment visits during the Treatment Period occur on Day 7±1 day (Visit 3) and Day 15±1 day (Visit 5). Subjects visit the clinic on Day 7±1 day (Visit 3) and Day 14±1 day (Visit 4) in the morning and the LCM device is dispensed for measurement of 24-hour cough count. Study assessments include assessment of quality of life (LCQ and K-BILD), cough severity (VAS), pulmonary function tests (forced expiratory volume in one second [FEV1], forced vital capacity [FVC], and FEV1/FVC ratio), fraction of exhaled nitric oxide (FeNO), and safety assessments (AEs, vital signs, and ECG) on Days 1, 7 and 15 of each treatment period.

A safety follow-up call is placed within 7±2 days following the last study treatment. Clinical safety laboratory samples are collected at the start and end of the treatment of each treatment period (Screening Visit and Visit 5 during the Treatment Period 1, and at Visit 2 and Visit 5 during the Treatment Period 2). All post-dose study procedures are conducted from time 0. Time 0 is defined as the start of the first study drug administration (i.e., when the nebulizer has been turned on) of each period.

Subjects are not allowed to use the following drugs (i.e., prednisone, narcotic antitussives, baclofen, gabapentin, inhaled corticosteroids, benzonatate, dextromethorphan, carbetapentane, and H1 antihistamines, leukotriene modifiers, and cromolyn sodium) for at least 2 weeks prior to the Screening Visit and throughout the study. Drugs containing bronchodilators (including beta-2 agonists and anticholinergics) are not allowed for at least 1 week prior to the Baseline Visit and during the study.

As shown in Table 1, PA101 contains 4% (by weight) cromolyn as the active substance, 0.2% Sodium Chloride as an osmotic agent, 0.02% EDTA as a chelating agent, 1.25% mannitol as a non-ionic osmotic agent, and purified water q.s. PA101 has an osmolality of 200 mOsm/kg.

A primary objective of the study was to assess the effectiveness of inhaled PA101 delivered via an eFlow nebulizer for treating chronic idiopathic cough.

The study design is provided graphically in FIG. 20.

The inclusion criteria of the chronic idiopathic cough (CIC) cohort are: 1) male or female subjects age 18 through 75 years, inclusive of the endpoints, 2) chronic cough that has been present for at least 8 weeks, 3) diagnosis of chronic idiopathic cough (CIC) that is unresponsive to targeted treatment for identified underlying triggers (i.e., post-nasal drip, asthmatic/non-asthmatic eosinophilic bronchitis, and gastro-esophageal reflux disease), 4) cough score on visual analogue scale of ≥40 mm at the Screening Visit, 5) daytime average cough count of at least 15 coughs per hour using objective cough count monitor at the Screening Visit, and 6) willingness and ability to provide written informed consent.

The exclusion criteria of the chronic idiopathic cough (CIC) cohort are: 1) current or recent history of clinically significant medical condition, laboratory abnormality, or illness that could put the subject at risk or compromise the quality of the study data as determined by the investigator, 2) an upper or lower respiratory tract infection within 4 weeks of the Screening Visit, 3) history of malignancy of any organ system, treated or untreated within the past 5 years, with the exception of localized basal cell carcinoma or cervix carcinoma in situ, 4) current or recent history (previous 12 months) of excessive use or abuse of alcohol, 5) current or recent history (previous 12 months) of abusing legal drugs or use of illegal drugs or substances, 5) participation in any other investigational drug study within 4 weeks prior to the Screening Visit, 6) use of the following drugs within 2 weeks of the Screening Visit: Prednisone, narcotic antitussives, baclofen, gabapentin, inhaled corticosteroids, benzonatate, dextromethorphan, carbetapentane, H1 antihistamines, leukotriene modifiers, and cromolyn sodium, 7) females who are pregnant or breastfeeding, or if of child-bearing potential unwilling to practice acceptable means of birth control or abstinence during the study (e.g., abstinence, combination barrier and spermicide, or hormonal), and 8) history of hypersensitivity or intolerance to cromolyn sodium.

Table 31 summarizes the CIC cohort demographics.

TABLE 31 Number of subjects (N) 27 Age 62 ± 11 (23-73 years) Sex 6 Male/21 Female Race 2 African American/25 White BMI 27.7 ± 5.7 Time since Diagnosis 9.9 ± 9.8 (2-43 years)

Safety Results:

As shown in Example 4, this study demonstrates that the use of PA101 is safe as the number of adverse events observed in subjects receiving PA101 and subjects receiving the placebo is not statistically significantly different.

Table 32 summarizes the Treatment Emergent Adverse Events (AEs) observed in at least two subjects of the CIC cohort.

TABLE 32 Adverse Events (AEs) PA101 (n = 25) Placebo (n = 27) Subjects with at least one Adverse 10 (40%) 13 (48.1%) Event (AE) Severe AEs 0 0 AEs leading to withdrawal 2 (7.4%) 2 (7.4%) Related AEs 6 (24%) 6 (22.2%) Unrelated AEs 4 (16%) 7 (25.9%) Mild AEs 6 (24%) 11 (40.7%) Moderate AEs 4 (16%) 2 (7.4%) Severe AEs 0 0 Respiratory System Cough 3 (12%) 3 (11.1%) Oropharyngeal pain 2 (8%) 3 (11.1%) Dyspnoea 1 (4%) 1 (4%) Pharyngeal hypoasthesia 2 (8%) 0 Throat clearing 0 1 (4%) Increased bronchial secretion 0 1 (4%) Nasal congestion 1 (4%) 0 Other Systems Tremor 2 (8%) 0 Dry Mouth 3 (12%) 0

Two subjects receiving the placebo withdrew from the study, one subject experiencing sore throat and headache post-dose (Period 1—Day 3) and the other subject experiencing dizziness (Period 1—Day 7).

Two subjects receiving PA101 withdrew from the study, one subject experiencing angioedema (thought to be a possible allergic reaction to IP) (Period 2—Day 6) and the other subject experiencing increased cough and shortness of breath (Period 2—Day 7).

Efficacy Results:

In sharp contrast to the results of the IPF cohort provided in Example 4, this study demonstrates that PA101 is not effective for the treatment of chronic idiopathic cough. Subjects with CIC who received PA101 did not present a statistically significant or clinically relevant improvement compared to the subjects with CIC who received the placebo.

Cough count was measured by Leicester Cough Monitor (LCM). Table 33 provides a summary of the daytime average cough/hour in subjects with CIC.

TABLE 33 PA101 (n = 25) Placebo (n = 26) Mean ± SD Mean ± SD Baseline 48 ± 79 44 ± 35 Day 7 41 ± 38 34 ± 29 Change from Baseline  −8 ± 53 (−15%) −11 ± 20 (−23%) Day 14 36 ± 39 36 ± 38 Change from Baseline −12 ± 72 (−25%) −8 ± 21 −18%) LS Mean Difference 1.27 (0.798-2.06) p = 0.32

Table 34 provides a summary of the change from baseline in log transformed daytime average cough/hour in subjects with CIC. This data is provided graphically in FIG. 23.

TABLE 34 Treatment PA101 Placebo (N = 22) (N = 22) Day 7 Number of subjects included in 22 (100.0) 21 (95.5) analysis (n) (%) Adjusted Least squares 0.98 0.73 treatment mean estimate mean 95% CI 0.76, 1.25 0.57, 0.93 (LSMean) Treatment Ratio of means 1.35 Comparison p-value  0.0382 95% CI 1.02, 1.78 Day 14/ Number of subjects included in 22 (100.0) 21 (95.5) ET analysis (n) (%) Adjusted Least squares 0.87 0.68 treatment mean estimate mean 95% CI 0.58, 1.30 0.45, 1.02 (LSMean) Treatment Ratio of means 1.28 Comparison p-value  0.3370 95% CI 0.76, 2.18

The individual average daytime cough/hour rates for the CIC cohort are provided in FIG. 21 at baseline and Day 14. FIG. 22 depicts the change from baseline log transformed daytime average cough/hour for the CIC cohort. The number of subjects responsive to treatment with PA101 versus placebo for CIC is shown in FIG. 24.

The 24-hour average cough/hour rates for the CIC cohort are provided in FIG. 22 at baseline, Day7 and Day 14. FIG. 25 depicts the change from baseline log transformed daytime average cough/hour for the CIC cohort.

Table 35 provides a summary of the change from baseline in log transformed 24-hour average cough/hour in subjects with CIC.

TABLE 35 Treatment PA101 Placebo (N = 25) (N = 27) Day 7 Number of subjects included in 23 (92.0) 24 (88.9) analysis (n) (%) Adjusted Least squares 0.93 0.72 treatment mean estimate mean 95% CI 0.73, 1.18 0.57, 0.90 (LSMean) Treatment Ratio of means 1.30 Comparison p-value  0.0690 95% CI 0.98, 1.73 Day 14/ Number of subjects included in 23 (92.0) 24 (88.9) ET analysis (n) (%) Adjusted Least squares 0.86 0.66 treatment mean estimate mean 95% CI 0.60, 1.25 0.46, 0.94 (LSMean) Treatment Ratio of means 1.30 Comparison p-value  0.2553 95% CI 0.81, 2.09

In addition to the objective cough/hour data captured using the LCM, subjective data was collected from study subjects. A quality of life score was measured by the Leicester Cough Questionnaire (LCQ). Table 36 summarizes the data collected using the Leicester Cough Questionnaire to assess Quality of Life changes from baseline to Days 7 and 14. Quality of Life was a secondary endpoint of this Phase II study. LCQ: worst score=3, best score=21; MID: >1.3.

TABLE 36 PA101 (n = 25) Placebo (n = 27) Mean ± SD Mean ± SD Baseline 10.9 ± 3.4 11.1 ± 3.1 Day 7 12.5 ± 3.9 12.3 ± 3.9 Change from Baseline 1.4 1.2 Day 14 12.6 ± 4.5 12.0 ± 4.1 Change from Baseline 1.6 1.0 LS Mean Difference 0.50 (−0.80, 1.80) p = 0.42

Table 37 summarizes the changes from baseline from the Leicester Cough Questionnaire for the CIC cohort.

TABLE 37 Treatment PA101 Placebo (N = 25) (N = 27) Day 7 Number of subjects included 23 (92.0)  25 (92.6)  in analysis (n) (%) Adjusted Least squares 1.27 1.21 treatment mean estimate mean 95% CI 0.23, 2.32 0.22, 2.20 (LSMean) Treatment Ratio of means 0.06 Comparison p-value  0.9280 95% CI −1.41, 1.54  Day 14/ Number of subjects included 25 (100.0) 27 (100.0) ET in analysis (n) (%) Adjusted Least squares 1.51 1.01 treatment mean estimate mean 95% CI 0.37, 2.65 −0.09, 2.10  (LSMean) Treatment Ratio of means 0.50 Comparison p-value  0.4232 95% CI −0.80, 1.80 

The study includes an additional subjective measure: Cough Severity, provided quantitatively as a measure on a visual analogue scale (VAS). When using the visual analogue scale (VAS), for example, to measure cough severity, the subject is asked to mark on a 100 mm scale between ‘no cough’ and ‘the worst cough severity’. Table 38 provides the mean, standard deviation (SD) and median scores on the VAS for each parameter by treatment at either day 7 or day 14 of the study for the CIC cohort.

TABLE 38 PA101 (n = 25) Placebo (n = 27) Mean ± SD Mean ± SD Baseline 65 ± 21 71 ± 12 Day 7 60 ± 22 63 ± 20 Change from Baseline −5.3  −8.1 Day 14 58 ± 23 61 ± 24 Change from Baseline −7.7 −10.0 LS Mean Difference −0.40 (−9.37, 8.58) p = 0.93

Table 39 summarizes the changes from baseline for Cough Severity as measured by the VAS score for the CIC cohort.

TABLE 39 Treatment PA101 Placebo (N = 25) (N = 27) Day 7 Number of subjects included 23 (92.0) 25 (92.6)  in analysis (n) (%) Adjusted Least squares −5.81 −7.47 treatment mean estimate mean 95% CI −13.08, 1.46  −14.39, −0.55 (LSMean) Treatment Ratio of means  1.66 Comparison p-value   0.6914 95% CI  −6.86, 10.18 Day 14/ Number of subjects included 25 (100.0) 27 (100.0) ET in analysis (n) (%) Adjusted Least squares −9.20 −8.80 treatment mean estimate mean 95% CI −17.73, −0.66 −17.06, −0.54 (LSMean) Treatment Ratio of means −0.40 Comparison p-value   0.9277 95% CI −9.37, 8.58

Tables 40a and 40b summarize the treatment period effect using daytime average count for the CIC cohort.

TABLE 40a Treatment Period 1 Baseline D 7 D 14 PA101 (n = 13) 65.2 48.6 35.0 Change from baseline −46% Placebo (n = 12) 52.8 41.1 48.3 Change from baseline  −8%

TABLE 40b Treatment Period 2 Baseline D 7 D 14 PA101 (n = 13) 29 32 43 Change from baseline 47% Placebo (n = 12) 37 27 25 Change from baseline −34% 

Taken together, PA101 has a very good safety profile for the treatment of CIC.

The efficacy results for the use of PA101 in the CIC cohort do not indicate a statistically significant difference between treatment groups using objective measures of count cough rate. Those subjects who received PA101 for CIC and those subject who receive the placebo for CIC present similar results 7 and 14 days after treatment begins. There are no favorable trends in cough-specific Quality of Life measurements (LCQ) or subject-reported cough severity using the VAS scale.

The results do not support the use of PA101 for the treatment of CIC.

Example 6: Cromolyn Sodium Formulations for Treatment of Chronic Cough Due to IPF Versus Chronic Idiopathic Cough (CIC)

The data from Example 4 demonstrate that the compositions of the disclosure, of which PA101 is an exemplary formulation, are safe and efficacious for the treatment of chronic cough due to IPF. In contrast, the data from Example 5 demonstrate that the compositions of the disclosure, of which PA101 is an exemplary formulation, are safe but are not efficacious for the treatment of chronic idiopathic cough (CIC). To further demonstrate this unexpected clinical result, FIGS. 26-29 compare the results of the daytime and 24-hour average cough/hour data between subjects with chronic cough due to IPF (Example 4) versus subjects with chronic idiopathic cough (CIC) (Example 5). The pharmacokinetic parameters in subjects administered PA101 (with mannitol) and PA101-B (without mannitol) are expected to be similar in subjects with chronic cough associated with IPF.

Example 7: Stability of Cromolyn Sodium Formulations

The compositions and formulations of the disclosure are both physically and chemically stable.

As shown by the physical appearance, Table 41 demonstrates that each formulation remains clear, and, therefore, free of any precipitate, from manufacture through the 24 month time point (i.e. for at least 24 months) when the formulations are stored at 25° C. As shown by the physical appearance, Table 42 demonstrates that each formulation remains clear, and, therefore, free of any precipitate, from manufacture through the 24 month time point (i.e. for at least 24 months) when the formulations are stored at 40° C.

As shown by the chemical measures of pH and osmolality, Table 41 demonstrates that each formulation maintains consistent appearance, pH, osmolality assay and related substances from manufacture through the 24 month time point (i.e. for at least 24 months) when the formulations are stored at 25° C. As shown by the chemical measures of pH and osmolality, Table 42 demonstrates that each formulation maintains consistent appearance, pH, osmolality, assay and related substances from manufacture through the 6 month time point (i.e. for at least 6 months) when the formulations are stored at 40° C.

TABLE 41 Stability data at 25° C. Stability Duration T0 3 months 6 months 9 months 12 months 18 months 24 months Appearance PA101, 20 mg/mL Clear Clear Clear Clear Clear PA101, 40 mg/mL Clear Clear Clear Clear Clear Clear PA101B, 10 mg/mL Clear Clear Clear Clear Clear Clear PA101B, 20 mg/mL Clear Clear Clear Clear Clear Clear PA101B, 40 mg/mL Clear Clear Clear Clear Clear Clear Clear PA101B, 60 mg/mL Clear Clear Clear Clear Clear Clear KM104, 60 mg/mL Clear Clear Clear Clear Clear pH PA101, 20 mg/mL 5.3 5.6 5.5 5.8 5.8 PA101, 40 mg/mL 5.4 5.7 5.5 5.9 5.7 5.8 PA101B, 10 mg/mL 5.5 6.1 5.9 5.7 5.4 6.2 PA101B, 20 mg/mL 5.7 6.2 5.7 5.8 5.4 6 PA101B, 40 mg/mL 5.5 6 6 5.8 5.9 6.32 5.94 PA101B, 60 mg/mL 5.2 5.3 5.4 5.5 5.6 5.6 KM104, 60 mg/mL 5.6 5.6 5.8 5.9 5.8 Osmolality PA101, 20 mg/mL 195 192 194 196 195 (mOsm/kg) PA101, 40 mg/mL 204 202 203 206 205 204 PA101B, 10 mg/mL 106 108 105 106 105 110 PA101B, 20 mg/mL 117 114 117 117 116 124 PA101B, 40 mg/mL 126 126 127 126 128 125 126 PA101B, 60 mg/mL 138 138 138 142 N/A 144 KM104, 60 mg/mL 294 288 291 289 291 Assay PA101, 20 mg/mL 101.8 103.6 102.6 102.6 104.6 (% Label PA101, 40 mg/mL 102.4 102.6 101.8 96.9 100.6 104.3 claim) PA101B, 10 mg/mL 98.9 102.2 102.2 100.8 101.2 96.7 PA101B, 20 mg/mL 98.1 101.7 98.7 100.2 99.1 96.6 PA101B, 40 mg/mL 97.3 99.2 101.1 103.7 100.7 98.9 101.6 PA101B, 60 mg/mL 98.7 100.8 100.7 101.8 99.9 102.2 KM104, 60 mg/mL 100 100.4 98.7 101.1 100.8 Related PA101, 20 mg/mL 0.11 0.11 0.11 0.11 <LOD substance PA101, 40 mg/mL 0.11 0.11 0.11 0.11 <LOD <LOD (% total) PA101B, 10 mg/mL 0.11 <LOD N/A <LOD <LOD <LOD PA101B, 20 mg/mL 0.11 <LOD <LOD <LOD <LOD <LOD PA101B, 40 mg/mL 0.11 0.11 <LOD <LOD <LOD <LOD <LOD PA101B, 60 mg/mL 0.1 0.1 0.1 0.1 0.1 0.1 KM104, 60 mg/mL 0.1 0.1 0.1 0.2 <LOD

TABLE 42 Stability at 40° C. Stability Duration T0 1 month 2 months 3 months 6 months Appearance PA101, 20 mg/mL Clear Clear Clear PA101, 40 mg/mL Clear Clear Clear PA101B, 10 mg/mL Clear Clear Clear Clear Clear PA101B, 20 mg/mL Clear Clear Clear Clear Clear PA101B, 40 mg/mL Clear Clear Clear Clear Clear KM104, 60 mg/mL Clear Clear Clear  Clear* pH PA101, 20 mg/mL 5.3 5.6 5.8    5.6 PA101, 40 mg/mL 5.4 5.6 5.8    5.7 PA101B, 10 mg/mL 5.5 5.9 6.4 6.1    5.6 PA101B, 20 mg/mL 5.7 5.9 5.8 6.3    5.9 PA101B, 40 mg/mL 5.5 6.0 5.9 5.8    5.9 KM104, 60 mg/mL 5.6 5.5 5.6    5.7* Osmolality PA101, 20 mg/mL 195 206 193 192 mOsm/kg PA101, 40 mg/mL 204 206 203 205 PA101B, 10 mg/mL 106 108 107 109 105 PA101B, 20 mg/mL 117 117 117 118 117 PA101B, 40 mg/mL 126 127 128 126 128 KM104, 60 mg/mL 294 293 288  292* Assay PA101, 20 mg/mL 101.8 102.9 102.9   102.7 (% Label PA101, 40 mg/mL 102.4 102.9 102.5   101.4 claim) PA101B, 10 mg/mL 98.9 98.9 98.9 100.3   103.6 PA101B, 20 mg/mL 98.1 98.5 98.8 100.2   98.1 PA101B, 40 mg/mL 97.3 98.0 98.9 99.2   101.8 KM104, 60 mg/mL 100 100.1 99.8   99.5 Related PA101, 20 mg/mL 0.11 0.11 0.11    0.11 substance PA101, 40 mg/mL 0.11 0.11 0.11    0.11 (% total) PA101B, 10 mg/mL 0.11 0.11 <LOD <LOD <LOD PA101B, 20 mg/mL 0.11 0.11 <LOD <LOD <LOD PA101B, 40 mg/mL 0.11 0.1 0.11 0.11 <LOD KM104, 60 mg/mL 0.11 <LOD 0.11     0.11* *KM104 data are at 13 months duration

INCORPORATION BY REFERENCE

Every document cited herein, including any cross referenced or related patent or application is hereby incorporated herein by reference in its entirety unless expressly excluded or otherwise limited. The citation of any document is not an admission that it is prior art with respect to any invention disclosed or claimed herein or that it alone, or in any combination with any other reference or references, teaches, suggests or discloses any such invention. Further, to the extent that any meaning or definition of a term in this document conflicts with any meaning or definition of the same term in a document incorporated by reference, the meaning or definition assigned to that term in this document shall govern.

Other Embodiments

While particular embodiments of the disclosure have been illustrated and described, various other changes and modifications can be made without departing from the spirit and scope of the disclosure. The scope of the appended claims includes all such changes and modifications that are within the scope of this disclosure.

Claims

1.-30. (canceled)

31. A method of reducing chronic cough in a subject having idiopathic pulmonary fibrosis, comprising administering to the subject a therapeutically effective amount of a pharmaceutical composition comprising cromolyn, or a pharmaceutically acceptable salt thereof;

with an inhalation device;
wherein the subject has been diagnosed as having idiopathic pulmonary fibrosis prior to administration of the pharmaceutical composition; and
wherein the 24 hour cough count decreases by about 10% to about 80%, relative to a baseline cough count.

32. The method of claim 31, wherein the 24 hour cough count decreases by about 10% to about 70%, relative to a baseline cough count.

33. The method of claim 31, wherein the 24 hour cough count decreases by about 10% to about 60%, relative to a baseline cough count.

34. The method of claim 31, wherein the 24 hour cough count decreases by about 10% to about 50%, relative to a baseline cough count.

35. The method of claim 31, wherein the 24 hour cough count decreases by about 10% to about 40%, relative to a baseline cough count.

36. The method of claim 31, wherein the 24 hour cough count decreases by about 10% to about 30%, relative to a baseline cough count.

37. The method of claim 31, wherein the 24 hour cough count decreases by about 10% to about 20%, relative to a baseline cough count.

38. The method of claim 31, wherein the inhalation device is a nebulizer or a dry powder inhaler.

39. The method of claim 38, wherein the inhalation device is a nebulizer.

40. The method of claim 39, wherein the nebulizer is a jet nebulizer.

41. The method of claim 39, wherein the nebulizer is a high-efficiency nebulizer.

42. The method of claim 41, wherein the high-efficiency nebulizer is a vibrating mesh nebulizer.

43. The method of claim 38, wherein the inhalation device is a dry powder inhaler.

44. The method of claim 31, wherein the composition comprises a pharmaceutically acceptable salt of cromolyn.

45. The method of claim 44, wherein the composition comprises cromolyn sodium.

46. The method of claim 31, wherein the pharmaceutical composition comprises between about 2% and about 99% by weight of a pharmaceutically acceptable salt of cromolyn.

47. The method of claim 46, wherein the composition comprises comprises between about 2% and about 99% by weight cromolyn sodium.

48. The method according to claim 31, wherein the pharmaceutical composition is administered to the subject from once per day to five times per day.

49. The method according to claim 31, wherein the pharmaceutical composition is administered to the subject three times per day.

50. The method of claim 38, wherein administration of the pharmaceutical composition to the subject with the inhalation device provides in the subject a bioavailability of cromolyn between about 5% and about 60%.

51. The method of claim 38, wherein each of the doses of the pharmaceutical composition comprises from about 10 mg to about 80 mg of cromolyn sodium, and administration of the pharmaceutical composition provides in the subject an AUC(0-∞) of the cromolyn sodium of between about 100 ng*hr/mL and about 500 ng*hr/mL.

52. The method of claim 38, wherein each of the doses of the pharmaceutical composition comprises from about 20 mg to about 80 mg of cromolyn sodium, and administration of the pharmaceutical composition provides in the subject a Cmax of the cromolyn sodium between about 50 ng/mL and about 230 ng/mL.

53. The method of claim 38, wherein the nebulizer produces an aerosol of the pharmaceutical composition that exhibits a respirable fraction (≤5 μm) as measured by USP <1601> of between about 60% and about 90%.

Patent History
Publication number: 20200155452
Type: Application
Filed: Sep 19, 2019
Publication Date: May 21, 2020
Applicant: Respivant Sciences GmbH (Basel)
Inventors: Pravin Soni (Sunnyvale, CA), William Gerhart (Del Mar, CA), Ahmet Tutuncu (Del Mar, CA)
Application Number: 16/576,487
Classifications
International Classification: A61K 9/00 (20060101); A61P 37/08 (20060101); A61K 31/35 (20060101); A61P 11/00 (20060101); A61K 31/352 (20060101); A61K 47/18 (20060101); A61P 11/14 (20060101);