US20220133629A1 - Formulations for treatment of dry eye disease - Google Patents

Formulations for treatment of dry eye disease Download PDF

Info

Publication number
US20220133629A1
US20220133629A1 US17/450,623 US202117450623A US2022133629A1 US 20220133629 A1 US20220133629 A1 US 20220133629A1 US 202117450623 A US202117450623 A US 202117450623A US 2022133629 A1 US2022133629 A1 US 2022133629A1
Authority
US
United States
Prior art keywords
cyclodextrin
pharmaceutically acceptable
reproxalap
day
ophthalmic solution
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/450,623
Inventor
David Clark
Todd BRADY
Susan Macdonald
Stephen Gitu Machatha
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aldeyra Therapeutics Inc
Original Assignee
Aldeyra Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aldeyra Therapeutics Inc filed Critical Aldeyra Therapeutics Inc
Priority to US17/450,623 priority Critical patent/US20220133629A1/en
Assigned to ALDEYRA THERAPEUTICS, INC. reassignment ALDEYRA THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRADY, TODD, CLARK, DAVID, MACDONALD, SUSAN, MACHATHA, Stephen Gitu
Publication of US20220133629A1 publication Critical patent/US20220133629A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/40Cyclodextrins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/04Artificial tears; Irrigation solutions

Definitions

  • the present invention relates to ophthalmic solutions comprising reproxalap, or a pharmaceutically acceptable salt thereof, for treatment of dry eye disease (DED).
  • DED dry eye disease
  • Dry eye disease is a complex disease that results in ocular discomfort, visual disturbance, and tear film instability, which create the potential for damage to the ocular surface. It is characterized by increased osmolarity of the tear film and inflammation of the ocular surface. Estimates of the prevalence of dry eye disease vary considerably, depending on the criteria used to define the disease, but in the United States (U.S.), it has been estimated that as many as 20 million adults in the U.S. have dry eye disease.
  • Aldehydes are reactive organic molecules that bind to proteins, carbohydrates, lipids and nucleic acids (Esterbauer, Free Radical Biology and Medicine, 1991, 11(1):81-128). Free aldehydes—aldehydes not sequestered or otherwise protected in specific metabolic processes—can be toxic, and aldehyde binding to cellular constituents can lead to inflammation (Yadav, Oxidative Medicine and Cellular Longevity, 2013, Volume 2013, Article ID 690545), molecular dysfunction (O'Brien, Critical Reviews in Toxicology, 2005, 35(7):609-62), and the accumulation of indigestible metabolites, such as lipofuscin components in the retina (Boyer, J Biol Chem., 2012, 287:22276-86).
  • aldehydes are formed by a variety of processes, including the oxidation of alcohols, polyamine and glucose metabolism, and oxidative stress. In some disease states, aldehyde concentrations may be increased. Increases in aldehyde concentrations, particularly malonyldialdehyde (MDA), which is thought to be most commonly derived from lipid peroxidation, has been described in a variety of inflammatory ocular diseases, including pterygium, Behcet's Disease, Sjögren's Syndrome, anterior uveitis, and dry eye disease (Sandikci, Acta Dermato-Venereologica, 2003, 83(5): 342-6; Cejkova, Histology and Histopathology, 2007, 22(9):997-1003; Balci, Molecular Vision, 2011, 17: 443-7; Turk, Ocular Immunology and Inflammation, 2014, 22(2):127-32; Choi, Current Eye Research, 2016, 41(9): 1143-9).
  • MDA malonyldial
  • the present invention provides an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the concentration of reproxalap, or a pharmaceutically acceptable salt thereof, is about 0.5% w/v or less and about 0.1% w/v or greater.
  • the ophthalmic solution comprises about 0.15 to 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the ophthalmic solution comprises about 0.2 to 0.3% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the ophthalmic solution comprises about 0.25% w/v reproxalap, and a pharmaceutically acceptable excipient.
  • the pharmaceutically acceptable excipient comprises a cyclodextrin, particularly sulfobutylether- ⁇ -cyclodextrin or hydroxypropyl- ⁇ -cyclodextrin.
  • the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to an eye of a subject with dry eye disease a therapeutically effective amount of an ophthalmic solution of the invention.
  • a method of the invention comprises topically administering to an eye of a subject with dry eye disease an ophthalmic solution of the invention four times a day (QID).
  • a method of the invention comprises topically administering to an eye of a subject with dry eye disease an ophthalmic solution of the invention three times a day (TID).
  • a method of the invention comprises topically administering to an eye of a subject with dry eye disease an ophthalmic solution of the invention two times a day (BID) or once a day. In some embodiments, a method of the invention comprises topically administering to an eye of a subject with dry eye disease ophthalmic solution of the invention as needed (PRN).
  • a method of the invention comprises topically administering an ophthalmic solution of the invention four times a day (QID) at initiation phase, followed by a maintenance phase wherein an ophthalmic solution of the invention is administered fewer than four times a day, for example, one, two, or three times a day.
  • QID a day
  • a maintenance phase wherein an ophthalmic solution of the invention is administered fewer than four times a day, for example, one, two, or three times a day.
  • FIG. 1 depicts OD & 4-Symptom Questionnaire: Dryness (ITT Population with Observed Data Only).
  • FIG. 2 depicts OD & 4-Symptom Questionnaire: Overall Ocular Discomfort (ITT Population with Observed Data Only).
  • FIG. 3 depicts Fluorescein Staining: Conjunctival Sum Score (Nasal and Temporal) (ITT Population with Observed Data Only).
  • FIG. 4 depicts Fluorescein Staining: Nasal (ITT Population with Observed Data Only).
  • FIG. 5 depicts Tear Quantity and Quality Improved: Tear Film Break-Up Time, Schirmer's Test & Tear Osmolarity Supports Broad Activity Profile (Endpoint Specific Worst Eye: ITT Population with Observed Data Only).
  • FIG. 6 depicts that CAE Endpoints are Confounded by Clear Efficacy at Week 12 Baseline Lissamine Green Staining: Inferior (ITT Population with Observed Data Only).
  • Reproxalap topical ocular solution is being developed for treatment of ocular inflammation.
  • the drug product in various strengths, has completed a Phase 1 clinical trial, as well as a controlled, double-masked Phase 2a clinical trial in allergic conjunctivitis; a controlled, double-masked Phase 2b clinical trial in allergic conjunctivitis; a controlled, double-masked Phase 2 clinical trial in noninfectious anterior uveitis; a Phase 2a clinical trial in dry eye disease; and a controlled, double-masked Phase 2b clinical trial in dry eye disease.
  • the maximum exposure to reproxalap in these completed clinical trials has been dosing with 0.5% w/v reproxalap for six weeks, for treating subjects with noninfectious anterior uveitis.
  • the objective of the Phase 2a clinical trial in dry eye disease was to assess the safety, tolerability, and pharmacodynamic activity of Reproxalap Ophthalmic Solutions in subjects with dry eye disease (DED) for 28 days of QID dosing with one of three different formulations.
  • the formulations used were 0.1% w/v Reproxalap Ophthalmic Solution, and 0.5% w/v Reproxalap Ophthalmic Solution, and 0.5% w/v Ophthalmic Lipid Solution.
  • SAEs serious adverse events
  • Reproxalap Ophthalmic Solutions 0.25% and 0.1%) were evaluated in patients with dry eye disease (see the details in Example 1).
  • Administration of Reproxalap Ophthalmic Solutions for 12 weeks resulted in statistically significant improvements in multiple signs and symptoms.
  • the efficacy profile of Reproxalap Ophthalmic Solutions was evident as early as two weeks following treatment initiation.
  • Reproxalap Ophthalmic Solutions showed broad activity across signs, including tear quantity (Schirmer's Test), tear quality (Tear Film Breakup Time (TBUT) and tear osmolarity), and ocular surface staining.
  • TBUT Near Film Breakup Time
  • osmolarity tear osmolarity
  • a dose response between 0.1% and 0.25% reproxalap dose strengths was demonstrated in the Phase 2b clinical trial.
  • Reproxalap Ophthalmic Solutions were found to be safe and well tolerated in the 12-week study, with only mild-to-moderate adverse events, including transient ocular stinging upon instillation.
  • the present invention provides an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the concentration of reproxalap, or a pharmaceutically acceptable salt thereof, is about 0.5% w/v or less and 0.1% w/v or greater.
  • the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to an eye of a subject with dry eye disease an ophthalmic solution described.
  • pharmaceutically acceptable is defined herein to refer to those compounds, biologic agents, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues a subject e.g., a mammal or human, without excessive toxicity, irritation allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
  • treating comprises a treatment relieving, reducing or alleviating at least one symptom in a subject or affecting a delay of progression of a disease, condition and/or disorder.
  • treatment can be the diminishment of one or several signs or symptoms of a disorder or complete eradication of a disorder.
  • the term “treat” also denotes to arrest, delay the onset (e.g., the period prior to clinical manifestation of a disease) and/or reduce the risk of developing or worsening a disease.
  • subject or “patient” as used herein includes animals, such as mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats and transgenic non-human animals.
  • the subject is a human.
  • the term “about” or “approximately” shall have the meaning of within 10% of a given value or range. In some embodiments, the term “about” refers to within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% of a given value.
  • w/v refers to “gram/mL” (weight over volume), which is a concentration unit. For example, 7% w/v is equivalent to 70 mg/mL.
  • reproxalap functions as an aldehyde sequestering agent, or “trap”, which binds rapidly to aldehydes and forms a cyclic product.
  • An ophthalmic solution of the invention comprises reproxalap, or a pharmaceutically acceptable salt thereof, at a concentration suitable for effectively treating dry eye disease, in particular without causing severe or intolerable adverse effects.
  • the present invention provides an ophthalmic solution comprising about 0.1% to 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the excipient comprises a cyclodextrin, such as sulfobutylether ⁇ -cyclodextrin (SBECD) or hydroxypropyl ⁇ -cyclodextrin.
  • an ophthalmic solution comprises repoxalap and a cyclodextrin excipient in a ratio of less than 1:2.1 on a mole:mole basis.
  • the ratio of reproxalap and cyclodextrin is about 1:2.1 to about 1:25 ratio on a mole:mole basis.
  • the ratio is about 1:2.2 to 1:20, 1:2.5 to 1:20, 1:2.5 to 1:10, 1:2.75 to 1:10, 1:3 to 1:8, 1:3.5 to 1:7, 1:4 to 1:6, or 1:4 to 1:5 in a mole:mole basis.
  • the ratio is about 1:2.1, 1:2.2, 1:2.3, 1:2.4, 1:2.5, 1:2.6, 1:2.7, 1:2.8, 1:2.9, 1:3, 1:3.1, 1:3.2, 1:3.3, 1:3.4, 1:3.5, 1:3.6, 1:3.7, 1:3.8, 1:3.9, 1:4.0, 1:4.1, 1:4.2, 1:4.3, 1:4.4, 1:4.5, 1:4.6, 1:4.7, 1:4.8, 1:4.9, 1:5.0, 1:5.1, 1:5.2, 1:5.3, 1:5.4, 1:5.5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:12, 1:15, 1:20, or 1:25 on a mole:mole basis.
  • the cyclodextrin excipient is one of those described herein, such as sulfobutylether ⁇ -cyclodextrin (SBECD).
  • SBECD sulfobutylether ⁇ -cyclodextrin
  • the average degree of substitution of the SBECD is about 6.5.
  • the ratio of reproxalap to the excipient is about 1:2.1 or less on a mole:mole basis.
  • the excipient is a cyclodextrin and the ratio of reproxalap to the excipient is about 1:2.1 to about 1:25 on a mole:mole basis.
  • the excipient is a cyclodextrin and the ratio of reproxalap to the excipient is about 1:2 to about 1:5 on a mole:mole basis.
  • the present invention provides an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the concentration of reproxalap, or a pharmaceutically acceptable salt thereof, is about 0.5% w/v or less and about 0.1% w/v or greater.
  • the ophthalmic solution comprises about 0.15 to about 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the ophthalmic solution comprises about 0.2 to about 0.4% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the ophthalmic solution comprises about 0.21 to about 0.35% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the ophthalmic solution comprises about 0.22 to about 0.3% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the ophthalmic solution comprises about 0.22 to about 0.29% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the ophthalmic solution comprises about 0.25% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the ophthalmic solution comprises about 0.25% w/v reproxalap and a pharmaceutically acceptable excipient selected from a cyclodextrin. In some embodiments, the ophthalmic solution comprises about 0.5% w/v reproxalap and a pharmaceutically acceptable excipient selected from a cyclodextrin.
  • the present invention provides an ophthalmic solution comprising less than 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the present invention provides an ophthalmic solution comprising at least 0.1% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the present invention provides an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the concentration of reproxalap, or a pharmaceutically acceptable salt thereof, is less than 0.5% w/v and 0.1% w/v or greater.
  • reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.45% w/v and at least 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.4% w/v and at least 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.35% w/v and at least 0.1% w/v.
  • reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.3% w/v and at least 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.25% w/v and more than 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.2% w/v and at least 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.15% w/v and at least 0.1% w/v.
  • reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.15% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.2% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.25% w/v.
  • reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.3% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.35% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.4% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.45% w/v.
  • reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of about 0.1% to 0.5%, 0.15% to 0.45% w/v, 0.15% to 0.4% w/v, 0.15% to 0.35% w/v, 0.15% to 0.3% w/v, 0.15% to 0.25% w/v, or 0.15% to 0.2% w/v.
  • reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.2% to 0.45% w/v, 0.2% to 0.4% w/v, 0.2% to 0.35% w/v, 0.2% to 0.3% w/v, or 0.2% to 0.25% w/v.
  • reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.25% to 0.45% w/v, 0.25% to 0.4% w/v, 0.25% to 0.35% w/v, or 0.25% to 0.3% w/v.
  • reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.3% to 0.45% w/v or 0.3% to 0.4% w/v.
  • reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of about 0.1% w/v, 0.15% w/v, about 0.2% w/v, about 0.25%, about 0.3% w/v, about 0.35% w/v, about 0.4% w/v, about 0.45% w/v, or about 0.5% w/v.
  • the foregoing concentrations of reproxalap can be selected and applied to treatment regimen that includes an initiation phase, an exacerbation phase, and/or a maintenance phase.
  • a pharmaceutically acceptable excipient in an ophthalmic solution of the invention is a cyclodextrin.
  • a cyclodextrin is ⁇ -, ⁇ - and ⁇ -cyclodextrin.
  • a cyclodextrin is a pharmaceutically acceptable derivative of a cyclodextrin, including, but not limited to, the hydroxyalkyl derivatives of ⁇ -, ⁇ - and ⁇ -cyclodextrin (especially the hydroxyethyl and hydroxypropyl derivatives of ⁇ -cyclodextrin and ⁇ -cyclodextrin), randomly methylated ⁇ -cyclodextrin, sulfobutylether ⁇ -cyclodextrin, sulfobutylether ⁇ -cyclodextrin, and the so-called branched ⁇ - and ⁇ -cyclodextrin derivatives such as glucosyl- ⁇ -cyclodextrin and glucosyl- ⁇ -cyclodextrin.
  • the natural cyclodextrins are either used alone or in a mixture of two or more cyclodextrins, by way of non-limiting example, a mixture of the ⁇ -cyclodextrin and the more water-soluble hydroxypropyl ⁇ -cyclodextrin, or ⁇ -cyclodextrin and sulfobutylether ⁇ -cyclodextrin, or ⁇ -cyclodextrin and hydroxypropyl- ⁇ -cyclodextrin, or ⁇ -cyclodextrin and sulfobutylether ⁇ -cyclodextrin.
  • a cyclodextrin in an ophthalmic solution of the invention is at a concentration of 0 to 20% w/v. In some embodiments, a cyclodextrin in an ophthalmic solution of the invention is at a concentration of 1 to 18% w/v, 1 to 16% w/v, 1 to 14% w/v, 2 to 12% w/v, 4 to 10% w/v, 5 to 9% w/v, or 6 to 8% w/v. In some embodiments, the cyclodextrin in an ophthalmic solution of the invention is at a concentration of 7% to 11% w/v.
  • a cyclodextrin in an ophthalmic solution of the invention is at a concentration of about 1% w/v, 2% w/v, 3% w/v, 4% w/v, 5% w/v, 6% w/v, 7% w/v, 8% w/v, 9% w/v, 10% w/v, 11% w/v, 12% w/v, 13% w/v, 14% w/v, 15% w/v, 16% w/v, 17% w/v, 18% w/v, 19% w/v, or 20% w/v.
  • a pharmaceutically acceptable excipient in an ophthalmic solution of the invention is sulfobutylether- ⁇ -cyclodextrin, in particular at any of the specified concentrations and ranges of concentrations above, such as about 7% w/v.
  • a pharmaceutically acceptable excipient in an ophthalmic solution of the invention is hydroxypropyl- ⁇ -cyclodextrin, in particular at any of the specified concentrations and ranges of concentrations specified above, such as about 7% w/v.
  • the ophthalmic solution comprises about 0.2% to 0.4% w/v reproxalap and about 7% to 25% w/v of a cyclodextrin excipient such as SBECD. In some embodiments, the ophthalmic solution comprises about 0.2%, 0.25%, 0.3%, 0.35%, or 0.4% w/v reproxalap and about 7% to 25% w/v of a cyclodextrin excipient such as SBECD.
  • the ophthalmic solution comprises about 0.25% w/v reproxalap and about 4.7% to about 25% w/v of a cyclodextrin excipient such as SBECD.
  • the ophthalmic solution comprises about 0.25% w/v reproxalap and about 7% to 25% w/v of a cyclodextrin excipient such as SBECD.
  • the ophthalmic solution comprises about 0.25% w/v reproxalap and about 4.75% to about 11% w/v of a cyclodextrin excipient such as SBECD.
  • the ophthalmic solution comprises about 0.5% w/v reproxalap and about 9.5% to about 11% w/v of a cyclodextrin excipient such as SBECD.
  • the ratio of API to SBECD is about a mole of API per 2 moles of SBECD.
  • the ophthalmic solution comprises about 0.25% w/v reproxalap and about 7% w/v of a cyclodextrin excipient such as SBECD.
  • the ratio of API to SBECD is about a mole of API per 3 moles SBECD.
  • the ophthalmic solution comprises about 0.25% w/v reproxalap and about 11% w/v of a cyclodextrin excipient such as SBECD.
  • the ratio of API to SBECD is about a mole of API per 5 moles SBECD.
  • an ophthalmic solution of the invention comprises a pharmaceutically acceptable buffering agent.
  • a pharmaceutically acceptable buffering agent is a phosphate buffer, citrate buffer, tris buffer, histidine buffer or acetate buffer.
  • a pharmaceutically acceptable buffering agent is sodium phosphate, dibasic. In some embodiments, a pharmaceutically acceptable buffering agent is sodium phosphate, monobasic. In some embodiments, a pharmaceutically acceptable buffering agent is a mixture of sodium phosphate, dibasic, and sodium phosphate, monobasic. In some embodiments, an ophthalmic solution of the invention comprises about 0.083% w/v sodium phosphate, dibasic, and about 0.017% w/v sodium phosphate, monobasic.
  • the ophthalmic solution of the invention is at an approximately neutral pH. In some embodiments, an ophthalmic solution of the invention is at a pH of 6.5 to 8. In some embodiments, an ophthalmic solution of the invention is at a pH of 6.9 to 7.7. In some embodiments, an ophthalmic solution of the invention is at a pH of 7.1 to 7.5. In some embodiments, an ophthalmic solution of the invention is at a pH of about 7.3.
  • an ophthalmic solution of the invention comprises a pharmaceutically acceptable acid.
  • an ophthalmic solution of the invention comprises a pharmaceutically acceptable base.
  • an ophthalmic solution of the invention comprises a pharmaceutically acceptable acid and base.
  • a pharmaceutically acceptable acid is hydrochloric acid.
  • pharmaceutically acceptable base is sodium hydroxide.
  • an ophthalmic solution of the invention comprises a tonicity agent.
  • a tonicity agent is selected from the group consisting of dextrose, potassium chloride, propylene glycol, and sodium chloride.
  • an ophthalmic solution of the invention comprises a tonicity agent at a concentration of less than about 0.5% w/v.
  • an ophthalmic solution of the invention comprises a tonicity agent at a concentration of about 0.45%, 0.4%, 0.35%, 0.3%, 0.25%, 0.2%, 0.15%, or 0.1% w/v.
  • a tonicity agent is sodium chloride.
  • the ophthalmic solution comprises reproxalap at the specified concentrations, cyclodextrin, phosphate, and sodium chloride. In some embodiments. In some embodiments, the ophthalmic solution comprises reproxalap at the specified concentrations herein (e.g., 0.1% w/v, 0.25% w/v, 0.5% w/v, etc.), 5 to 9% w/v cyclodextrin (e.g., sulfobutylether- ⁇ -cyclodextrin or hydroxypropyl- ⁇ -cyclodextrin); 0.07% to 0.09% w/v sodium phosphate (dibasic), 0.015% to 0.19% w/v sodium phosphate (monobasic), and 0.2 to 0.3% w/v sodium chloride.
  • the specified concentrations herein e.g., 0.1% w/v, 0.25% w/v, 0.5% w/v, etc.
  • the ophthalmic solution comprises reproxalap at the specified concentrations herein (e.g., 0.1% w/v, 0.25% w/v, 0.5% w/v, etc.), about 7% w/v cyclodextrin (e.g., sulfobutylether- ⁇ cyclodextrin or hydroxypropyl- ⁇ -cyclodextrin); 0.07% to 0.09% w/v sodium phosphate (dibasic), 0.015% to 0.019% w/v sodium phosphate (monobasic), and 0.2 to 0.3% w/v sodium chloride.
  • the ophthalmic solution is adjusted to an appropriate pH with sodium hydroxide or HCL.
  • the ophthalmic solution comprises the following (0.5% Reproxalap Ophthalmic Solution A):
  • the ophthalmic solution comprises the following (0.5% Reproxalap Ophthalmic Solution B)
  • the ophthalmic solution comprises the following (0.25% Reproxalap Ophthalmic Solution A)
  • the ophthalmic solution comprises the following (0.25% Reproxalap Ophthalmic Solution B)
  • the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to an eye of a subject in need thereof a therapeutically effective amount of an ophthalmic solution of the invention.
  • the concentration of reproxalap in the ophthalmic solution used in the method is as described above.
  • an ophthalmic solution of the invention can be administered at different frequencies suitable for effectively treating dry eye disease, for example, without causing severe or intolerable adverse effects.
  • an ophthalmic solution of the invention can be topically administered one to six times a day.
  • a method of the invention comprises topically administering an ophthalmic solution of the invention six times a day.
  • a method of the invention comprises topically administering an ophthalmic solution of the invention five times a day.
  • a method of the invention comprises topically administering an ophthalmic solution of the invention four times a day (QID).
  • a method of the invention comprises topically administering an ophthalmic solution of the invention three times a day (TID).
  • a method of the invention comprises topically administering an ophthalmic solution of the invention two times a day (BID).
  • a method of the invention comprises topically administering an ophthalmic solution of the invention once a day (QD). In some embodiments, a method of the invention comprises topically administering an ophthalmic solution of the invention as needed (PRN).
  • a method of the invention comprises topically administering to an eye of a subject with dry eye disease a therapeutically effective amount of an ophthalmic solution of the invention six times a day, five times a day, four times a day (QID), three times a day (TID), two times a day (BID), once a day (QD), followed by administration as needed (PRN).
  • a therapeutically effective amount of an ophthalmic solution of the invention six times a day, five times a day, four times a day (QID), three times a day (TID), two times a day (BID), once a day (QD), followed by administration as needed (PRN).
  • a method of the invention comprises topically administering an ophthalmic solution of the invention at various strengths (for example, at different reproxalap concentrations and different administration frequencies, as described herein).
  • a method of the invention comprises topically administering an ophthalmic solution comprising about 0.25% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • a method of the invention comprises topically administering an ophthalmic solution comprising about 0.30% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • a method of the invention comprises topically administering an ophthalmic solution comprising about 0.35% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • a method of the invention comprises topically administering an ophthalmic solution comprising about 0.4% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • a method of the invention comprises topically administering an ophthalmic solution comprising about 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • a method of the invention comprises topically administering an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • a method of the invention comprises topically administering an ophthalmic solution comprising 0.3% to 0.4% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • a method of the invention comprises topically administering an ophthalmic solution comprising 0.2% to 0.3% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • a method of the invention comprises topically administering an ophthalmic solution comprising 0.2% to 0.4% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • a method of the invention comprises two or more phases, wherein an ophthalmic solution of the invention is topically administering at different strengths in different phases.
  • a method of the invention comprises an initiation phase and a maintenance phase, wherein the ophthalmic solution is topically administered at a higher strength in the initiation phase than in the maintenance phase.
  • a treatment cycle of a method of the invention comprising multiple phases, including an exacerbation phase during which signs and/or symptoms become worse.
  • the method of the invention comprises two or more phases, wherein an ophthalmic solution of the invention is topically administering at different strengths in different phases.
  • a method of the invention comprises an initiation phase, wherein the ophthalmic solution is topically administered at a high strength in the initiation phase, at a low strength in the maintenance phase, and at a high strength during an exacerbation of disease signs and/or symptoms.
  • an ophthalmic solution administered in an initiation phase comprises a higher concentration of reproxalap, or a pharmaceutically acceptable salt thereof, than an ophthalmic solution administered in a maintenance phase.
  • the ophthalmic solution administered in an initiation phase or an exacerbation phase and the ophthalmic solution administered in a maintenance phase comprises reproxalap, or a pharmaceutically acceptable salt, at a concentration selected from the group consisting of about 0.5% w/v, 0.45% w/v, 0.4% w/v, 0.35% w/v, 0.3% w/v, 0.25% w/v, 0.2% w/v, 0.15% w/v, and 0.1% w/v.
  • an ophthalmic solution of about 0.5% w/v reproxalap is administered in an initiation phase or exacerbation phase, and less than 0.5% w/v reproxalap administered in a maintenance phase. In some embodiments, an ophthalmic solution of about 0.4% w/v, 0.35% w/v, 0.3% w/v, 0.25% w/v, 0.2% w/v, 0.15% w/v or 0.1% w/v reproxalap is administered in the maintenance phase.
  • an ophthalmic solution of about 0.5% w/v to about 0.4% reproxalap is administered in an initiation phase or exacerbation phase, and less than 0.4% w/v reproxalap administered in a maintenance phase.
  • an ophthalmic solution of about 0.35% w/v, 0.3% w/v, 0.25% w/v, 0.2% w/v, 0.15% w/v or 0.1% w/v reproxalap is administered in the maintenance phase.
  • an ophthalmic solution of about 0.5% w/v to about 0.3% reproxalap is administered in an initiation phase or exacerbation phase, and less than 0.3% w/v reproxalap administered in a maintenance phase. In some embodiments, an ophthalmic solution of about 0.25% w/v, 0.2% w/v, 0.15% w/v or 0.1% w/v reproxalap is administered in the maintenance phase.
  • an ophthalmic solution of about 0.4% w/v to about 0.3% reproxalap is administered in an initiation phase or exacerbation phase, and less than 0.3% w/v reproxalap administered in a maintenance phase. In some embodiments, an ophthalmic solution of about 0.25% w/v, 0.2% w/v, 0.15% w/v or 0.1% w/v reproxalap is administered in the maintenance phase.
  • an ophthalmic solution of about 0.3% w/v to about 0.2% reproxalap (e.g., 0.3%, 0.25%, or 0.2% w/v) is administered in an initiation phase or exacerbation phase, and 0.25% w/v or less reproxalap administered in a maintenance phase.
  • an ophthalmic solution of about 0.25% w/v, 0.2% w/v, 0.15% w/v or 0.1% w/v reproxalap is administered in the maintenance phase.
  • an ophthalmic solution of the invention is topically administered more frequently per day in an initiation phase and an exacerbation phase than in a maintenance phase. In some embodiments, an ophthalmic solution of the invention is topically administered five times a day in an initiation phase, followed by four, three, two, or one times a day in a maintenance phase. In some embodiments, an ophthalmic solution of the invention is topically administering four times a day in an initiation phase or exacerbation phase, followed by three, two, or one times a day in a maintenance phase.
  • an ophthalmic solution of the invention is topically administering three times a day in an initiation phase or exacerbation phase, followed by two or one times a day in a maintenance phase. In some embodiments, an ophthalmic solution of the invention is topically administering two times a day in an initiation phase or exacerbation phase, followed by once daily in a maintenance phase.
  • an ophthalmic solution administered in an initiation phase or exacerbation phase is at a higher reproxalap concentration and higher administration frequency than an ophthalmic solution administered in a maintenance phase.
  • the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to the subject an ophthalmic solution comprising about 0.4% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the ophthalmic solution is administered at a higher strength in an initiation phase or exacerbation phase followed by a lower strength in a maintenance phase, wherein each of the initiation phase, exacerbation phase, and maintenance phase is as described herein.
  • a multiple phase treatment cycle can include an initiation phase or exacerbation phase of up to 12 weeks with an ophthalmic solution comprising about 0.5%, 0.4% or 0.35% w/v (e.g., 0.5% to 0.35% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is up to 12 weeks, followed by a maintenance phase.
  • an ophthalmic solution comprising about 0.5%, 0.4% or 0.35% w/v (e.g., 0.5% to 0.35% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered four times a day in an initiation phase or exacerbation phase followed by three, two, or one times a day in the maintenance phase.
  • an ophthalmic solution comprising about 0.5%, 0.4% or 0.35% w/v (e.g., 0.5% to 0.35% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered three times a day in an initiation phase or exacerbation phase followed by two or one times a day in the maintenance phase.
  • an ophthalmic solution comprising about 0.4%, 0.35% or 0.3% w/v (e.g., 0.4% to 0.3% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered four times a day in an initiation phase or exacerbation phase followed by three, two, or one times a day in the maintenance phase.
  • an ophthalmic solution comprising about 0.4%, 0.35% or 0.3% w/v (e.g., 0.4% to 0.3% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered three times a day in an initiation phase or exacerbation phase followed by two or one times a day in the maintenance phase.
  • an ophthalmic solution comprising about 0.3%, 0.25% or 0.2% w/v (e.g., 0.3% to 0.2% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered four times a day in an initiation phase or exacerbation phase followed by three, two, or one times a day in the maintenance phase.
  • an ophthalmic solution comprising about 0.3%, 0.25% or 0.2% w/v (e.g., 0.3% to 0.2% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered three times a day in an initiation phase or exacerbation phase followed by two or one times a day in the maintenance phase.
  • the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to the subject an ophthalmic solution comprising 0.35% to 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the ophthalmic solution is administered at a higher strength in an initiation phase or exacerbation phase followed by a lower strength in a maintenance phase, wherein each of the initiation phase, exacerbation phase and maintenance phase is as described herein.
  • a multiple phase treatment cycle of an ophthalmic solution comprising 0.35% to 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof is up to 12 weeks.
  • an ophthalmic solution comprising 0.35% to 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is administered four times a day in an initiation phase or exacerbation phase followed by three, two, or one times a day in maintenance phase.
  • an ophthalmic solution comprising 0.35%-0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is administered three times a day in an initiation phase or exacerbation phase followed by two or one times a day in maintenance phase.
  • an ophthalmic solution is administered QID for about 10 to 14 weeks, preferably about 12 weeks. In some embodiments, an ophthalmic solution is administration QID for about 2 to 6 weeks, preferably about 4 weeks followed by administration BID for about 6 to 10 weeks, preferably about 8 weeks. In some embodiments, the ophthalmic solution for the foregoing treatment regimen is 0.25% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and about 7% w/v SBECD.
  • an ophthalmic solution is administered QID for about 2 to 6 weeks, preferably about 4 weeks, followed by administration BID for about 6 to 10 weeks, preferably about 8 weeks.
  • the ophthalmic solution for the foregoing treatment regimen is 0.25% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and about 11% w/v SBECD.
  • the present invention provides a method for treating certain subjects with dry eye disease.
  • a subject with dry eye disease is 18 years or older.
  • a subject with dry eye disease has a history of dry eye for at least six months prior to receiving the treatment of the invention.
  • a subject with dry eye disease has a history of use or desire to use eye drops for dry eye symptoms within six months prior to receiving the treatment of the invention.
  • the present invention provides a method for treating a subject with dry eye disease, in particular moderate-to-severe dry-eye disease, comprising identifying subjects satisfying one or more of the following criteria for at least one eye, prior to receiving the treatment of the invention (for example, a screening performed at about one and/or two weeks before receiving the treatment):
  • a subject with dry eye disease is not a female patient who is pregnant, nursing, or planning a pregnancy. In some embodiments, a subject with dry eye disease has not previously used reproxalap ophthalmic solution.
  • the present invention provides a method for treating a subject with dry eye disease comprising a screening to exclude subjects having one or more of the following conditions for at least one eye, prior to receiving the treatment of the invention:
  • an ophthalmic solution of the invention can achieve an early onset of effect in subjects with dry eye disease.
  • an “early onset effect” refers to early efficacy (e.g., within 1 to 2 weeks of initiation of treatment—in initiation or exacerbation phase) in ameliorating symptoms of dry eye disease.
  • the “early onset effect” is for the same dose and frequency of administration in the initiation or exacerbation phase.
  • the present invention provides a method for treating a subject with dry eye disease comprising topically administering to the subject an ophthalmic solution of the invention, wherein the ophthalmic solution is administered at a dose strength which can achieve an early onset profile.
  • an early onset profile comprises early onset of effect for symptoms (e.g., ocular discomfort including dryness, itchiness, tearing, burning, stinging, grittiness, cloudy vision, sensitivity to environment, stringy ocular secretion).
  • an early onset profile comprises early onset of effect for signs (e.g., ocular vital staining, tear film break-up time, tear osmolarity, tear volume).
  • a dose strength which can achieve an early onset of effect comprises topically administering an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, at a concentration as described herein. In some embodiments, a dose strength which can achieve an early onset of effect comprises topically administering an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, at a frequency at described herein. In some embodiments, a dose strength which can achieve an early onset of effect comprises topically administering an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, at a concentration and a frequency at described herein.
  • a method of the invention can achieve an onset of effect in about two weeks.
  • a method of the invention can achieve an onset in fewer than about two weeks.
  • a method of the invention can achieve an onset in about 14, 13, 12, 11, ten, nine, or eight days.
  • a method of the invention can achieve an onset in about one week or less.
  • a method of the invention can achieve an onset in about seven, six, five, four, three, two, or one days.
  • the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to the subject an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the ophthalmic solution is administered three, two, or one times a day.
  • an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof is administered three times a day.
  • an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof is administered two times a day.
  • an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof is administered once daily.
  • the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to the subject an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the ophthalmic solution is administered at a higher strength in an initiation phase or exacerbation phase, followed by a lower strength in a maintenance phase, wherein each of the initiation phase, exacerbation phase, and maintenance phase is as described herein.
  • an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof is administered four times a day in an initiation phase or exacerbation phase followed by three, two, or one times a day in a maintenance phase.
  • an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof is administered three times a day in an initiation phase or exacerbation phase followed by two or one times a day in a maintenance phase.
  • an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof is administered two times a day in an initiation phase followed by one time a day in a maintenance phase.
  • an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof is topically administered in an initiation phase or exacerbation phase, followed by topical administration of an ophthalmic solution comprising less than about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, in a maintenance phase, wherein the administration frequency of each ophthalmic solution is selected from those as described above.
  • reproxalap was formulated as an ophthalmic solution as described in the specification.
  • a subject's participation was estimated to be approximately 14 weeks (98 days).
  • Dosage/Dose Regimen/Instillation/Application/Use Screening: Between Visits 1 and 2, all subjects received 14 consecutive days ( ⁇ 2) of Run-in (vehicle) ocular drops self-administered QID in both eyes.
  • Reproxalap Ophthalmic Solution at concentrations of 0.1%, 0.25%, or vehicle ophthalmic solution was administered QID by bilateral topical ocular dosing. Subjects were randomized to one of three treatment groups (1:1:1) to receive study drug after the Post-CAE® assessments at Visit 2.
  • Sample Size The study sample size of 100 per group was selected based on prior Phase 2 and 3 clinical trial results using the DED Hybrid CAE study design with other development programs and the effect size seen in Phase 2a with reproxalap on change from baseline after four weeks of treatment. This sample size was deemed sufficient to assess the effect size on the DED sign and symptom endpoints with reproxalap vs vehicle, to confirm the endpoint selection and sample size needed for Phase 3 studies with reproxalap.
  • phase 2b data are shown in FIGS. 1 through 9 and Tables 1 through 3.
  • Ocular Discomfort Scale ⁇ ⁇ ⁇ OD & 4-Symptom Overall Ocular (0-5) ⁇ ⁇ ⁇ c ⁇ a Questionnaire Discomfort Burning (0-5) ⁇ N/A b N/A Dryness (0-5) ⁇ ⁇ ⁇ ⁇ c a Grittiness (0-5) ⁇ ⁇ ⁇ c Stinging (0-5) ⁇ ⁇ b c c a Ocular Surface (0-100) ⁇ ⁇ N/A Disease Index (OSDI) SANDE Severity (0-100 mm) ⁇ ⁇ N/A ⁇ b Questionnaire Frequency (0-100 mm) ⁇ ⁇ ⁇ Sign Measures Lisamine Green Total Score (0-20; ⁇ 5x) ⁇ ⁇ ⁇ ⁇ Staining (all five regions) Corneal Sum Score (0-12; ⁇ 3x) ⁇ ⁇ ⁇ ⁇ c (

Abstract

The present invention provides a reproxalap ophthalmic solution, and methods of using the same for treating dry eye disease.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 16/582,720, filed Sep. 25, 2019, which claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application No. 62/736,417, filed Sep. 25, 2018, and U.S. Provisional Application No. 62/824,233, filed Mar. 26, 2019; the entire contents of each of which is incorporated herein by reference.
  • TECHNICAL FIELD
  • The present invention relates to ophthalmic solutions comprising reproxalap, or a pharmaceutically acceptable salt thereof, for treatment of dry eye disease (DED).
  • BACKGROUND
  • Dry eye disease is a complex disease that results in ocular discomfort, visual disturbance, and tear film instability, which create the potential for damage to the ocular surface. It is characterized by increased osmolarity of the tear film and inflammation of the ocular surface. Estimates of the prevalence of dry eye disease vary considerably, depending on the criteria used to define the disease, but in the United States (U.S.), it has been estimated that as many as 20 million adults in the U.S. have dry eye disease. It has been projected that there will be a 40% increase in number of patients affected by 2030 (Schaumberg, Advances in Experimental Medicine and Biology, 2002, 506:989-98; Schaumberg, American Journal of Ophthalmology, 2003, 136:318-26; Schaumberg, Archives of Ophthalmology, 2009, 127:763-8). With the aging population in the U.S. and other countries of the developed world, and increasing computer use, dry eye disease is expected to become more prevalent. Thus, finding a treatment is becoming more important (Brewitt, Survey of Ophthalmology, 2001, 45 Suppl 2:S199-202).
  • Aldehydes are reactive organic molecules that bind to proteins, carbohydrates, lipids and nucleic acids (Esterbauer, Free Radical Biology and Medicine, 1991, 11(1):81-128). Free aldehydes—aldehydes not sequestered or otherwise protected in specific metabolic processes—can be toxic, and aldehyde binding to cellular constituents can lead to inflammation (Yadav, Oxidative Medicine and Cellular Longevity, 2013, Volume 2013, Article ID 690545), molecular dysfunction (O'Brien, Critical Reviews in Toxicology, 2005, 35(7):609-62), and the accumulation of indigestible metabolites, such as lipofuscin components in the retina (Boyer, J Biol Chem., 2012, 287:22276-86).
  • In biological systems, aldehydes are formed by a variety of processes, including the oxidation of alcohols, polyamine and glucose metabolism, and oxidative stress. In some disease states, aldehyde concentrations may be increased. Increases in aldehyde concentrations, particularly malonyldialdehyde (MDA), which is thought to be most commonly derived from lipid peroxidation, has been described in a variety of inflammatory ocular diseases, including pterygium, Behcet's Disease, Sjögren's Syndrome, anterior uveitis, and dry eye disease (Sandikci, Acta Dermato-Venereologica, 2003, 83(5): 342-6; Cejkova, Histology and Histopathology, 2007, 22(9):997-1003; Balci, Molecular Vision, 2011, 17: 443-7; Turk, Ocular Immunology and Inflammation, 2014, 22(2):127-32; Choi, Current Eye Research, 2016, 41(9): 1143-9).
  • SUMMARY
  • In some embodiments, the present invention provides an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the concentration of reproxalap, or a pharmaceutically acceptable salt thereof, is about 0.5% w/v or less and about 0.1% w/v or greater. In some embodiments, the ophthalmic solution comprises about 0.15 to 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the ophthalmic solution comprises about 0.2 to 0.3% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • In some embodiments, the ophthalmic solution comprises about 0.25% w/v reproxalap, and a pharmaceutically acceptable excipient.
  • In some embodiments, the pharmaceutically acceptable excipient comprises a cyclodextrin, particularly sulfobutylether-β-cyclodextrin or hydroxypropyl-β-cyclodextrin.
  • In some embodiments, the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to an eye of a subject with dry eye disease a therapeutically effective amount of an ophthalmic solution of the invention. In some embodiments, a method of the invention comprises topically administering to an eye of a subject with dry eye disease an ophthalmic solution of the invention four times a day (QID). In some embodiments, a method of the invention comprises topically administering to an eye of a subject with dry eye disease an ophthalmic solution of the invention three times a day (TID). In some embodiments, a method of the invention comprises topically administering to an eye of a subject with dry eye disease an ophthalmic solution of the invention two times a day (BID) or once a day. In some embodiments, a method of the invention comprises topically administering to an eye of a subject with dry eye disease ophthalmic solution of the invention as needed (PRN).
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution of the invention four times a day (QID) at initiation phase, followed by a maintenance phase wherein an ophthalmic solution of the invention is administered fewer than four times a day, for example, one, two, or three times a day.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts OD & 4-Symptom Questionnaire: Dryness (ITT Population with Observed Data Only).
  • FIG. 2 depicts OD & 4-Symptom Questionnaire: Overall Ocular Discomfort (ITT Population with Observed Data Only).
  • FIG. 3 depicts Fluorescein Staining: Conjunctival Sum Score (Nasal and Temporal) (ITT Population with Observed Data Only).
  • FIG. 4 depicts Fluorescein Staining: Nasal (ITT Population with Observed Data Only).
  • FIG. 5 depicts Tear Quantity and Quality Improved: Tear Film Break-Up Time, Schirmer's Test & Tear Osmolarity Supports Broad Activity Profile (Endpoint Specific Worst Eye: ITT Population with Observed Data Only).
  • FIG. 6 depicts that CAE Endpoints are Confounded by Clear Efficacy at Week 12 Baseline Lissamine Green Staining: Inferior (ITT Population with Observed Data Only).
  • FIG. 7 depicts Fluorescein Staining: Inferior Total Population (N=100/100/100) vs High Median Subgroup (N=68/69/66) (ITT Population with Observed Data Only).
  • FIG. 8 depicts Fluorescein Staining: Nasal Total Population (N=100/100/100) vs High Median Subgroup (N=59/56/62) (ITT Population with Observed Data Only).
  • FIG. 9 depicts Fluorescein Staining: Conjunctival Sum Score (Nasal and Temporal) Total Population (N=100/100/100) vs High Median Subgroup (N=55/56/60) (ITT Population with Observed Data Only).
  • DETAILED DESCRIPTION 1. General Description of Certain Embodiments of the Invention
  • Reproxalap topical ocular solution is being developed for treatment of ocular inflammation. The drug product, in various strengths, has completed a Phase 1 clinical trial, as well as a controlled, double-masked Phase 2a clinical trial in allergic conjunctivitis; a controlled, double-masked Phase 2b clinical trial in allergic conjunctivitis; a controlled, double-masked Phase 2 clinical trial in noninfectious anterior uveitis; a Phase 2a clinical trial in dry eye disease; and a controlled, double-masked Phase 2b clinical trial in dry eye disease. The maximum exposure to reproxalap in these completed clinical trials has been dosing with 0.5% w/v reproxalap for six weeks, for treating subjects with noninfectious anterior uveitis.
  • The objective of the Phase 2a clinical trial in dry eye disease was to assess the safety, tolerability, and pharmacodynamic activity of Reproxalap Ophthalmic Solutions in subjects with dry eye disease (DED) for 28 days of QID dosing with one of three different formulations. The formulations used were 0.1% w/v Reproxalap Ophthalmic Solution, and 0.5% w/v Reproxalap Ophthalmic Solution, and 0.5% w/v Ophthalmic Lipid Solution. No serious adverse events (SAEs) were observed during the 28-day treatment with any of the three reproxalap formulations, and no clinically significant change in visual acuity (VA), intraocular pressure (IOP), slit lamp biomicroscopic findings, or undilated funduscopic findings were observed. Drop comfort was less well tolerated with the 0.5% w/v Reproxalap Ophthalmic Solution and Ophthalmic Lipid Solution than with the 0.1% w/v Reproxalap Ophthalmic Solution. Statistically significant efficacy in within-subject improvement was observed over a broad array of DED signs and symptoms assessed as exploratory pharmacodynamics endpoints.
  • In a Phase 2b clinical trial, the efficacy of Reproxalap Ophthalmic Solutions (0.25% and 0.1%) were evaluated in patients with dry eye disease (see the details in Example 1). Administration of Reproxalap Ophthalmic Solutions for 12 weeks resulted in statistically significant improvements in multiple signs and symptoms. The efficacy profile of Reproxalap Ophthalmic Solutions was evident as early as two weeks following treatment initiation. Reproxalap Ophthalmic Solutions showed broad activity across signs, including tear quantity (Schirmer's Test), tear quality (Tear Film Breakup Time (TBUT) and tear osmolarity), and ocular surface staining. In addition, a dose response between 0.1% and 0.25% reproxalap dose strengths was demonstrated in the Phase 2b clinical trial. Reproxalap Ophthalmic Solutions were found to be safe and well tolerated in the 12-week study, with only mild-to-moderate adverse events, including transient ocular stinging upon instillation.
  • Accordingly, in some embodiments, the present invention provides an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the concentration of reproxalap, or a pharmaceutically acceptable salt thereof, is about 0.5% w/v or less and 0.1% w/v or greater. In some embodiments, the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to an eye of a subject with dry eye disease an ophthalmic solution described.
  • 2. Definitions
  • The general terms used herein are defined with the following meanings, unless explicitly stated otherwise.
  • The term “comprising” and “including” are used herein in their open-ended and non-limiting sense unless otherwise noted. It is to be further understood that where descriptions of various embodiments use the term “comprising” or “including,” those skilled in the art would understand that in some specific instances, an embodiment can be alternatively described using language “consisting essentially of” or “consisting of.”
  • The terms “a” and “an” and “the” and similar references in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Where the plural form is used for compounds, salts, and the like, this is taken to mean also a single compound, salt, or the like.
  • The term “pharmaceutically acceptable” is defined herein to refer to those compounds, biologic agents, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues a subject e.g., a mammal or human, without excessive toxicity, irritation allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
  • The term “treating” or “treatment” as used herein comprises a treatment relieving, reducing or alleviating at least one symptom in a subject or affecting a delay of progression of a disease, condition and/or disorder. For example, treatment can be the diminishment of one or several signs or symptoms of a disorder or complete eradication of a disorder. Within the meaning of the present invention, the term “treat” also denotes to arrest, delay the onset (e.g., the period prior to clinical manifestation of a disease) and/or reduce the risk of developing or worsening a disease.
  • The term “subject” or “patient” as used herein includes animals, such as mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats and transgenic non-human animals. In some embodiments, the subject is a human.
  • The term “about” or “approximately” shall have the meaning of within 10% of a given value or range. In some embodiments, the term “about” refers to within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% of a given value.
  • The term “w/v” as used herein refers to “gram/mL” (weight over volume), which is a concentration unit. For example, 7% w/v is equivalent to 70 mg/mL.
  • Reproxalap is of formula
  • Figure US20220133629A1-20220505-C00001
  • Without wishing to be bound by any particular theory, reproxalap functions as an aldehyde sequestering agent, or “trap”, which binds rapidly to aldehydes and forms a cyclic product.
  • 3. Ophthalmic Solutions
  • An ophthalmic solution of the invention comprises reproxalap, or a pharmaceutically acceptable salt thereof, at a concentration suitable for effectively treating dry eye disease, in particular without causing severe or intolerable adverse effects. In some embodiments, the present invention provides an ophthalmic solution comprising about 0.1% to 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the excipient comprises a cyclodextrin, such as sulfobutylether β-cyclodextrin (SBECD) or hydroxypropyl β-cyclodextrin.
  • In some embodiments, an ophthalmic solution comprises repoxalap and a cyclodextrin excipient in a ratio of less than 1:2.1 on a mole:mole basis. In some embodiments, the ratio of reproxalap and cyclodextrin is about 1:2.1 to about 1:25 ratio on a mole:mole basis. In some embodiments, the ratio is about 1:2.2 to 1:20, 1:2.5 to 1:20, 1:2.5 to 1:10, 1:2.75 to 1:10, 1:3 to 1:8, 1:3.5 to 1:7, 1:4 to 1:6, or 1:4 to 1:5 in a mole:mole basis. In some embodiments, the ratio is about 1:2.1, 1:2.2, 1:2.3, 1:2.4, 1:2.5, 1:2.6, 1:2.7, 1:2.8, 1:2.9, 1:3, 1:3.1, 1:3.2, 1:3.3, 1:3.4, 1:3.5, 1:3.6, 1:3.7, 1:3.8, 1:3.9, 1:4.0, 1:4.1, 1:4.2, 1:4.3, 1:4.4, 1:4.5, 1:4.6, 1:4.7, 1:4.8, 1:4.9, 1:5.0, 1:5.1, 1:5.2, 1:5.3, 1:5.4, 1:5.5, 1:6, 1:7, 1:8, 1:9, 1:10, 1:12, 1:15, 1:20, or 1:25 on a mole:mole basis.
  • In some embodiments, the cyclodextrin excipient is one of those described herein, such as sulfobutylether β-cyclodextrin (SBECD). The average degree of substitution of the SBECD is about 6.5.
  • In some embodiments, the ratio of reproxalap to the excipient is about 1:2.1 or less on a mole:mole basis.
  • In some embodiments, the excipient is a cyclodextrin and the ratio of reproxalap to the excipient is about 1:2.1 to about 1:25 on a mole:mole basis.
  • In some embodiments, the excipient is a cyclodextrin and the ratio of reproxalap to the excipient is about 1:2 to about 1:5 on a mole:mole basis.
  • In some embodiments, the present invention provides an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the concentration of reproxalap, or a pharmaceutically acceptable salt thereof, is about 0.5% w/v or less and about 0.1% w/v or greater. In some embodiments, the ophthalmic solution comprises about 0.15 to about 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the ophthalmic solution comprises about 0.2 to about 0.4% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the ophthalmic solution comprises about 0.21 to about 0.35% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the ophthalmic solution comprises about 0.22 to about 0.3% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the ophthalmic solution comprises about 0.22 to about 0.29% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the ophthalmic solution comprises about 0.25% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the ophthalmic solution comprises about 0.25% w/v reproxalap and a pharmaceutically acceptable excipient selected from a cyclodextrin. In some embodiments, the ophthalmic solution comprises about 0.5% w/v reproxalap and a pharmaceutically acceptable excipient selected from a cyclodextrin.
  • In some embodiments, the present invention provides an ophthalmic solution comprising less than 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the present invention provides an ophthalmic solution comprising at least 0.1% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the present invention provides an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the concentration of reproxalap, or a pharmaceutically acceptable salt thereof, is less than 0.5% w/v and 0.1% w/v or greater.
  • In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.45% w/v and at least 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.4% w/v and at least 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.35% w/v and at least 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.3% w/v and at least 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.25% w/v and more than 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.2% w/v and at least 0.1% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of less than 0.15% w/v and at least 0.1% w/v.
  • In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.15% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.2% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.25% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.3% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.35% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.4% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.5% w/v or less and at least 0.45% w/v.
  • In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of about 0.1% to 0.5%, 0.15% to 0.45% w/v, 0.15% to 0.4% w/v, 0.15% to 0.35% w/v, 0.15% to 0.3% w/v, 0.15% to 0.25% w/v, or 0.15% to 0.2% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.2% to 0.45% w/v, 0.2% to 0.4% w/v, 0.2% to 0.35% w/v, 0.2% to 0.3% w/v, or 0.2% to 0.25% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.25% to 0.45% w/v, 0.25% to 0.4% w/v, 0.25% to 0.35% w/v, or 0.25% to 0.3% w/v. In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of 0.3% to 0.45% w/v or 0.3% to 0.4% w/v.
  • In some embodiments, reproxalap, or a pharmaceutically acceptable salt thereof, in an ophthalmic solution of the invention is at a concentration of about 0.1% w/v, 0.15% w/v, about 0.2% w/v, about 0.25%, about 0.3% w/v, about 0.35% w/v, about 0.4% w/v, about 0.45% w/v, or about 0.5% w/v.
  • In some embodiments, as further described herein, the foregoing concentrations of reproxalap can be selected and applied to treatment regimen that includes an initiation phase, an exacerbation phase, and/or a maintenance phase.
  • In some embodiments, a pharmaceutically acceptable excipient in an ophthalmic solution of the invention is a cyclodextrin. In some embodiments, a cyclodextrin is α-, β- and γ-cyclodextrin. In some embodiments, a cyclodextrin is a pharmaceutically acceptable derivative of a cyclodextrin, including, but not limited to, the hydroxyalkyl derivatives of α-, β- and γ-cyclodextrin (especially the hydroxyethyl and hydroxypropyl derivatives of β-cyclodextrin and γ-cyclodextrin), randomly methylated β-cyclodextrin, sulfobutylether β-cyclodextrin, sulfobutylether γ-cyclodextrin, and the so-called branched β- and γ-cyclodextrin derivatives such as glucosyl-β-cyclodextrin and glucosyl-γ-cyclodextrin. The natural cyclodextrins are either used alone or in a mixture of two or more cyclodextrins, by way of non-limiting example, a mixture of the γ-cyclodextrin and the more water-soluble hydroxypropyl γ-cyclodextrin, or γ-cyclodextrin and sulfobutylether γ-cyclodextrin, or β-cyclodextrin and hydroxypropyl-β-cyclodextrin, or β-cyclodextrin and sulfobutylether β-cyclodextrin.
  • In some embodiments, a cyclodextrin in an ophthalmic solution of the invention is at a concentration of 0 to 20% w/v. In some embodiments, a cyclodextrin in an ophthalmic solution of the invention is at a concentration of 1 to 18% w/v, 1 to 16% w/v, 1 to 14% w/v, 2 to 12% w/v, 4 to 10% w/v, 5 to 9% w/v, or 6 to 8% w/v. In some embodiments, the cyclodextrin in an ophthalmic solution of the invention is at a concentration of 7% to 11% w/v. In some embodiments, a cyclodextrin in an ophthalmic solution of the invention is at a concentration of about 1% w/v, 2% w/v, 3% w/v, 4% w/v, 5% w/v, 6% w/v, 7% w/v, 8% w/v, 9% w/v, 10% w/v, 11% w/v, 12% w/v, 13% w/v, 14% w/v, 15% w/v, 16% w/v, 17% w/v, 18% w/v, 19% w/v, or 20% w/v.
  • In some embodiments, a pharmaceutically acceptable excipient in an ophthalmic solution of the invention is sulfobutylether-β-cyclodextrin, in particular at any of the specified concentrations and ranges of concentrations above, such as about 7% w/v. In some embodiments, a pharmaceutically acceptable excipient in an ophthalmic solution of the invention is hydroxypropyl-β-cyclodextrin, in particular at any of the specified concentrations and ranges of concentrations specified above, such as about 7% w/v.
  • In some embodiments, the ophthalmic solution comprises about 0.2% to 0.4% w/v reproxalap and about 7% to 25% w/v of a cyclodextrin excipient such as SBECD. In some embodiments, the ophthalmic solution comprises about 0.2%, 0.25%, 0.3%, 0.35%, or 0.4% w/v reproxalap and about 7% to 25% w/v of a cyclodextrin excipient such as SBECD.
  • In some embodiments, the ophthalmic solution comprises about 0.25% w/v reproxalap and about 4.7% to about 25% w/v of a cyclodextrin excipient such as SBECD.
  • In some embodiments, the ophthalmic solution comprises about 0.25% w/v reproxalap and about 7% to 25% w/v of a cyclodextrin excipient such as SBECD.
  • In some embodiments, the ophthalmic solution comprises about 0.25% w/v reproxalap and about 4.75% to about 11% w/v of a cyclodextrin excipient such as SBECD.
  • In some embodiments, the ophthalmic solution comprises about 0.5% w/v reproxalap and about 9.5% to about 11% w/v of a cyclodextrin excipient such as SBECD. In some embodiments, the ratio of API to SBECD is about a mole of API per 2 moles of SBECD.
  • In some embodiments, the ophthalmic solution comprises about 0.25% w/v reproxalap and about 7% w/v of a cyclodextrin excipient such as SBECD. In some embodiments, the ratio of API to SBECD is about a mole of API per 3 moles SBECD.
  • In some embodiments, the ophthalmic solution comprises about 0.25% w/v reproxalap and about 11% w/v of a cyclodextrin excipient such as SBECD. In some embodiments, the ratio of API to SBECD is about a mole of API per 5 moles SBECD.
  • In some embodiments, an ophthalmic solution of the invention comprises a pharmaceutically acceptable buffering agent. In some embodiments, a pharmaceutically acceptable buffering agent is a phosphate buffer, citrate buffer, tris buffer, histidine buffer or acetate buffer.
  • In some embodiments, a pharmaceutically acceptable buffering agent is sodium phosphate, dibasic. In some embodiments, a pharmaceutically acceptable buffering agent is sodium phosphate, monobasic. In some embodiments, a pharmaceutically acceptable buffering agent is a mixture of sodium phosphate, dibasic, and sodium phosphate, monobasic. In some embodiments, an ophthalmic solution of the invention comprises about 0.083% w/v sodium phosphate, dibasic, and about 0.017% w/v sodium phosphate, monobasic.
  • In some embodiments, the ophthalmic solution of the invention is at an approximately neutral pH. In some embodiments, an ophthalmic solution of the invention is at a pH of 6.5 to 8. In some embodiments, an ophthalmic solution of the invention is at a pH of 6.9 to 7.7. In some embodiments, an ophthalmic solution of the invention is at a pH of 7.1 to 7.5. In some embodiments, an ophthalmic solution of the invention is at a pH of about 7.3.
  • Pharmaceutically acceptable acids and/or bases may be used in the ophthalmic solution to adjust pH. In some embodiments, an ophthalmic solution of the invention comprises a pharmaceutically acceptable acid. In some embodiments, an ophthalmic solution of the invention comprises a pharmaceutically acceptable base. In some embodiments, an ophthalmic solution of the invention comprises a pharmaceutically acceptable acid and base. In some embodiments, a pharmaceutically acceptable acid is hydrochloric acid. In some embodiments, pharmaceutically acceptable base is sodium hydroxide.
  • In some embodiments, an ophthalmic solution of the invention comprises a tonicity agent. In some embodiments, a tonicity agent is selected from the group consisting of dextrose, potassium chloride, propylene glycol, and sodium chloride. In some embodiments, an ophthalmic solution of the invention comprises a tonicity agent at a concentration of less than about 0.5% w/v. In some embodiments, an ophthalmic solution of the invention comprises a tonicity agent at a concentration of about 0.45%, 0.4%, 0.35%, 0.3%, 0.25%, 0.2%, 0.15%, or 0.1% w/v. In some embodiments, a tonicity agent is sodium chloride.
  • In some embodiments, the ophthalmic solution comprises reproxalap at the specified concentrations, cyclodextrin, phosphate, and sodium chloride. In some embodiments. In some embodiments, the ophthalmic solution comprises reproxalap at the specified concentrations herein (e.g., 0.1% w/v, 0.25% w/v, 0.5% w/v, etc.), 5 to 9% w/v cyclodextrin (e.g., sulfobutylether-β-cyclodextrin or hydroxypropyl-β-cyclodextrin); 0.07% to 0.09% w/v sodium phosphate (dibasic), 0.015% to 0.19% w/v sodium phosphate (monobasic), and 0.2 to 0.3% w/v sodium chloride. In some embodiments, the ophthalmic solution comprises reproxalap at the specified concentrations herein (e.g., 0.1% w/v, 0.25% w/v, 0.5% w/v, etc.), about 7% w/v cyclodextrin (e.g., sulfobutylether-β cyclodextrin or hydroxypropyl-β-cyclodextrin); 0.07% to 0.09% w/v sodium phosphate (dibasic), 0.015% to 0.019% w/v sodium phosphate (monobasic), and 0.2 to 0.3% w/v sodium chloride. In some embodiments, the ophthalmic solution is adjusted to an appropriate pH with sodium hydroxide or HCL.
  • In some embodiments, the ophthalmic solution comprises the following (0.5% Reproxalap Ophthalmic Solution A):
  • Component Amount (% w/v) Grade
    ADX-102 (reproxalap) 0.5% GMP
    Sulfobutylether-beta-cyclodextrin 9.5% USP
    (SBECD)
    Sodium phosphate, dibasic, anhydrous 0.083% USP
    Sodium phosphate, monobasic, 0.017% USP
    monohydrate
    Sodium hydroxide or Hydrochloric acid pH adjustment USP/NF
    Sterile Water for Injection (WFI) Dilute to volume USP
  • In some embodiments, the ophthalmic solution comprises the following (0.5% Reproxalap Ophthalmic Solution B)
  • Component Amount (% w/v) Grade
    ADX-102 (reproxalap) 0.5% GMP
    Sulfobutylether-beta-cyclodextrin 9.5% USP
    (SBECD)
    Sodium hydroxide or Hydrochloric acid pH adjustment USP/NF
    Sterile Water for Injection (WFI) Dilute to volume USP
  • In some embodiments, the ophthalmic solution comprises the following (0.25% Reproxalap Ophthalmic Solution A)
  • Component Amount (% w/v) Grade
    ADX-102 (reproxalap) 0.25% GMP
    Sulfobutylether-beta-cyclodextrin 7.0% USP
    (SBECD)
    Sodium phosphate, dibasic, anhydrous 0.083% USP
    Sodium phosphate, monobasic, 0.017% USP
    monohydrate
    Sodium chloride 0.24% USP
    Sodium chloride Tonicity adjustment USP
    Sodium hydroxide or Hydrochloric acid pH adjustment USP/NF
    Sterile Water for Injection (WFI) Dilute to volume USP
  • In some embodiments, the ophthalmic solution comprises the following (0.25% Reproxalap Ophthalmic Solution B)
  • Component Amount (% w/v) Grade
    ADX-102 (reproxalap) 0.25% GMP
    Sulfobutylether-beta-cyclodextrin 7.0% USP
    (SBECD)
    Sodium chloride Tonicity adjustment USP
    Sodium hydroxide or Hydrochloric acid pH adjustment USP/NF
    Sterile Water for Injection (WFI) Dilute to volume USP
  • It is to be understood that variations of the ophthalmic solutions within the scope of the disclosure may be prepared given the guidance provided herein.
  • 4. Methods of Treatment
  • In one aspect, the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to an eye of a subject in need thereof a therapeutically effective amount of an ophthalmic solution of the invention. In some embodiments, the concentration of reproxalap in the ophthalmic solution used in the method is as described above.
  • In some embodiments, an ophthalmic solution of the invention can be administered at different frequencies suitable for effectively treating dry eye disease, for example, without causing severe or intolerable adverse effects.
  • In some embodiments, an ophthalmic solution of the invention can be topically administered one to six times a day. In some embodiments, a method of the invention comprises topically administering an ophthalmic solution of the invention six times a day. In some embodiments, a method of the invention comprises topically administering an ophthalmic solution of the invention five times a day. In some embodiments, a method of the invention comprises topically administering an ophthalmic solution of the invention four times a day (QID). In some embodiments, a method of the invention comprises topically administering an ophthalmic solution of the invention three times a day (TID). In some embodiments, a method of the invention comprises topically administering an ophthalmic solution of the invention two times a day (BID). In some embodiments, a method of the invention comprises topically administering an ophthalmic solution of the invention once a day (QD). In some embodiments, a method of the invention comprises topically administering an ophthalmic solution of the invention as needed (PRN).
  • In some embodiments, a method of the invention comprises topically administering to an eye of a subject with dry eye disease a therapeutically effective amount of an ophthalmic solution of the invention six times a day, five times a day, four times a day (QID), three times a day (TID), two times a day (BID), once a day (QD), followed by administration as needed (PRN).
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution of the invention at various strengths (for example, at different reproxalap concentrations and different administration frequencies, as described herein).
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution comprising about 0.25% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution comprising about 0.30% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution comprising about 0.35% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution comprising about 0.4% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution comprising about 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution comprising 0.3% to 0.4% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution comprising 0.2% to 0.3% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • In some embodiments, a method of the invention comprises topically administering an ophthalmic solution comprising 0.2% to 0.4% w/v reproxalap, or a pharmaceutically acceptable salt thereof, four times a day, three times a day, or two times a day.
  • In some embodiments, a method of the invention comprises two or more phases, wherein an ophthalmic solution of the invention is topically administering at different strengths in different phases. In some embodiments, a method of the invention comprises an initiation phase and a maintenance phase, wherein the ophthalmic solution is topically administered at a higher strength in the initiation phase than in the maintenance phase. In some embodiments, a treatment cycle of a method of the invention comprising multiple phases, including an exacerbation phase during which signs and/or symptoms become worse.
  • In some embodiments, the method of the invention comprises two or more phases, wherein an ophthalmic solution of the invention is topically administering at different strengths in different phases. In some embodiments, a method of the invention comprises an initiation phase, wherein the ophthalmic solution is topically administered at a high strength in the initiation phase, at a low strength in the maintenance phase, and at a high strength during an exacerbation of disease signs and/or symptoms.
  • In some embodiments, an ophthalmic solution administered in an initiation phase comprises a higher concentration of reproxalap, or a pharmaceutically acceptable salt thereof, than an ophthalmic solution administered in a maintenance phase. In some embodiments, the ophthalmic solution administered in an initiation phase or an exacerbation phase and the ophthalmic solution administered in a maintenance phase, comprises reproxalap, or a pharmaceutically acceptable salt, at a concentration selected from the group consisting of about 0.5% w/v, 0.45% w/v, 0.4% w/v, 0.35% w/v, 0.3% w/v, 0.25% w/v, 0.2% w/v, 0.15% w/v, and 0.1% w/v.
  • In some embodiments, an ophthalmic solution of about 0.5% w/v reproxalap is administered in an initiation phase or exacerbation phase, and less than 0.5% w/v reproxalap administered in a maintenance phase. In some embodiments, an ophthalmic solution of about 0.4% w/v, 0.35% w/v, 0.3% w/v, 0.25% w/v, 0.2% w/v, 0.15% w/v or 0.1% w/v reproxalap is administered in the maintenance phase.
  • In some embodiments, an ophthalmic solution of about 0.5% w/v to about 0.4% reproxalap is administered in an initiation phase or exacerbation phase, and less than 0.4% w/v reproxalap administered in a maintenance phase. In some embodiments, an ophthalmic solution of about 0.35% w/v, 0.3% w/v, 0.25% w/v, 0.2% w/v, 0.15% w/v or 0.1% w/v reproxalap is administered in the maintenance phase.
  • In some embodiments, an ophthalmic solution of about 0.5% w/v to about 0.3% reproxalap is administered in an initiation phase or exacerbation phase, and less than 0.3% w/v reproxalap administered in a maintenance phase. In some embodiments, an ophthalmic solution of about 0.25% w/v, 0.2% w/v, 0.15% w/v or 0.1% w/v reproxalap is administered in the maintenance phase.
  • In some embodiments, an ophthalmic solution of about 0.4% w/v to about 0.3% reproxalap is administered in an initiation phase or exacerbation phase, and less than 0.3% w/v reproxalap administered in a maintenance phase. In some embodiments, an ophthalmic solution of about 0.25% w/v, 0.2% w/v, 0.15% w/v or 0.1% w/v reproxalap is administered in the maintenance phase.
  • In some embodiments, an ophthalmic solution of about 0.3% w/v to about 0.2% reproxalap (e.g., 0.3%, 0.25%, or 0.2% w/v) is administered in an initiation phase or exacerbation phase, and 0.25% w/v or less reproxalap administered in a maintenance phase. In some embodiments, an ophthalmic solution of about 0.25% w/v, 0.2% w/v, 0.15% w/v or 0.1% w/v reproxalap is administered in the maintenance phase.
  • In some embodiments, an ophthalmic solution of the invention is topically administered more frequently per day in an initiation phase and an exacerbation phase than in a maintenance phase. In some embodiments, an ophthalmic solution of the invention is topically administered five times a day in an initiation phase, followed by four, three, two, or one times a day in a maintenance phase. In some embodiments, an ophthalmic solution of the invention is topically administering four times a day in an initiation phase or exacerbation phase, followed by three, two, or one times a day in a maintenance phase. In some embodiments, an ophthalmic solution of the invention is topically administering three times a day in an initiation phase or exacerbation phase, followed by two or one times a day in a maintenance phase. In some embodiments, an ophthalmic solution of the invention is topically administering two times a day in an initiation phase or exacerbation phase, followed by once daily in a maintenance phase.
  • In some embodiments, an ophthalmic solution administered in an initiation phase or exacerbation phase is at a higher reproxalap concentration and higher administration frequency than an ophthalmic solution administered in a maintenance phase.
  • In some embodiments, the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to the subject an ophthalmic solution comprising about 0.4% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the ophthalmic solution is administered at a higher strength in an initiation phase or exacerbation phase followed by a lower strength in a maintenance phase, wherein each of the initiation phase, exacerbation phase, and maintenance phase is as described herein.
  • In some embodiments, a multiple phase treatment cycle can include an initiation phase or exacerbation phase of up to 12 weeks with an ophthalmic solution comprising about 0.5%, 0.4% or 0.35% w/v (e.g., 0.5% to 0.35% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is up to 12 weeks, followed by a maintenance phase. In some embodiments, an ophthalmic solution comprising about 0.5%, 0.4% or 0.35% w/v (e.g., 0.5% to 0.35% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered four times a day in an initiation phase or exacerbation phase followed by three, two, or one times a day in the maintenance phase. In some embodiments, an ophthalmic solution comprising about 0.5%, 0.4% or 0.35% w/v (e.g., 0.5% to 0.35% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered three times a day in an initiation phase or exacerbation phase followed by two or one times a day in the maintenance phase.
  • In some embodiments, an ophthalmic solution comprising about 0.4%, 0.35% or 0.3% w/v (e.g., 0.4% to 0.3% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered four times a day in an initiation phase or exacerbation phase followed by three, two, or one times a day in the maintenance phase. In some embodiments, an ophthalmic solution comprising about 0.4%, 0.35% or 0.3% w/v (e.g., 0.4% to 0.3% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered three times a day in an initiation phase or exacerbation phase followed by two or one times a day in the maintenance phase.
  • In some embodiments, an ophthalmic solution comprising about 0.3%, 0.25% or 0.2% w/v (e.g., 0.3% to 0.2% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered four times a day in an initiation phase or exacerbation phase followed by three, two, or one times a day in the maintenance phase. In some embodiments, an ophthalmic solution comprising about 0.3%, 0.25% or 0.2% w/v (e.g., 0.3% to 0.2% w/v) reproxalap, or a pharmaceutically acceptable salt thereof, is administered three times a day in an initiation phase or exacerbation phase followed by two or one times a day in the maintenance phase.
  • In some embodiments, the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to the subject an ophthalmic solution comprising 0.35% to 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the ophthalmic solution is administered at a higher strength in an initiation phase or exacerbation phase followed by a lower strength in a maintenance phase, wherein each of the initiation phase, exacerbation phase and maintenance phase is as described herein. In some embodiments, a multiple phase treatment cycle of an ophthalmic solution comprising 0.35% to 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is up to 12 weeks. In some embodiments, an ophthalmic solution comprising 0.35% to 0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is administered four times a day in an initiation phase or exacerbation phase followed by three, two, or one times a day in maintenance phase. In some embodiments, an ophthalmic solution comprising 0.35%-0.45% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is administered three times a day in an initiation phase or exacerbation phase followed by two or one times a day in maintenance phase.
  • In some embodiments, an ophthalmic solution is administered QID for about 10 to 14 weeks, preferably about 12 weeks. In some embodiments, an ophthalmic solution is administration QID for about 2 to 6 weeks, preferably about 4 weeks followed by administration BID for about 6 to 10 weeks, preferably about 8 weeks. In some embodiments, the ophthalmic solution for the foregoing treatment regimen is 0.25% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and about 7% w/v SBECD.
  • In some embodiments, an ophthalmic solution is administered QID for about 2 to 6 weeks, preferably about 4 weeks, followed by administration BID for about 6 to 10 weeks, preferably about 8 weeks. In some embodiments, the ophthalmic solution for the foregoing treatment regimen is 0.25% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and about 11% w/v SBECD.
  • In some embodiments, the present invention provides a method for treating certain subjects with dry eye disease. In some embodiments, a subject with dry eye disease is 18 years or older. In some embodiments, a subject with dry eye disease has a history of dry eye for at least six months prior to receiving the treatment of the invention. In some embodiments, a subject with dry eye disease has a history of use or desire to use eye drops for dry eye symptoms within six months prior to receiving the treatment of the invention.
  • In some embodiments, the present invention provides a method for treating a subject with dry eye disease, in particular moderate-to-severe dry-eye disease, comprising identifying subjects satisfying one or more of the following criteria for at least one eye, prior to receiving the treatment of the invention (for example, a screening performed at about one and/or two weeks before receiving the treatment):
      • having a Schirmer's Test score of ≤10 mm and ≥1 mm;
      • having a tear film break-up time (TFBUT®)≤5 seconds;
      • having a corneal fluorescein staining score of ∝2 in at least one region (e.g., inferior, superior, or central);
      • having a sum corneal fluorescein staining score of ≥4 based on the sum of the inferior, superior, and central regions; and
      • having a total Lissamine green conjunctival score of ≥2 based on the sum of the temporal and nasal regions.
  • In some embodiments, a subject with dry eye disease is not a female patient who is pregnant, nursing, or planning a pregnancy. In some embodiments, a subject with dry eye disease has not previously used reproxalap ophthalmic solution.
  • In some embodiments, the present invention provides a method for treating a subject with dry eye disease comprising a screening to exclude subjects having one or more of the following conditions for at least one eye, prior to receiving the treatment of the invention:
      • having any clinically significant slit lamp findings that may include active blepharitis, meibomian gland dysfunction (MGD), lid margin inflammation, or active ocular allergies that may require therapeutic treatment;
      • having an ongoing ocular infection (bacterial, viral, or fungal), or active ocular inflammation;
      • having previously had laser-assisted in situ keratomileusis (LASIK) surgery within the last 12 months;
      • having any planned ocular and/or lid surgeries over the study period or any ocular surgery within six months; and
      • having a known allergy and/or sensitivity to an ophthalmic solution of the invention or its components.
  • As described herein, an ophthalmic solution of the invention can achieve an early onset of effect in subjects with dry eye disease. As used herein, an “early onset effect” refers to early efficacy (e.g., within 1 to 2 weeks of initiation of treatment—in initiation or exacerbation phase) in ameliorating symptoms of dry eye disease. In some embodiments, the “early onset effect” is for the same dose and frequency of administration in the initiation or exacerbation phase. Accordingly, in some embodiments, the present invention provides a method for treating a subject with dry eye disease comprising topically administering to the subject an ophthalmic solution of the invention, wherein the ophthalmic solution is administered at a dose strength which can achieve an early onset profile. In some embodiments, an early onset profile comprises early onset of effect for symptoms (e.g., ocular discomfort including dryness, itchiness, tearing, burning, stinging, grittiness, cloudy vision, sensitivity to environment, stringy ocular secretion). In some embodiments, an early onset profile comprises early onset of effect for signs (e.g., ocular vital staining, tear film break-up time, tear osmolarity, tear volume).
  • In some embodiments, a dose strength which can achieve an early onset of effect comprises topically administering an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, at a concentration as described herein. In some embodiments, a dose strength which can achieve an early onset of effect comprises topically administering an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, at a frequency at described herein. In some embodiments, a dose strength which can achieve an early onset of effect comprises topically administering an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, at a concentration and a frequency at described herein.
  • In some embodiments, a method of the invention can achieve an onset of effect in about two weeks. At different dose strengths (for example, different concentration and administering frequency), a method of the invention can achieve an onset in fewer than about two weeks. For example, in some embodiments, a method of the invention can achieve an onset in about 14, 13, 12, 11, ten, nine, or eight days. At a certain dose strength, a method of the invention can achieve an onset in about one week or less. In some embodiments, a method of the invention can achieve an onset in about seven, six, five, four, three, two, or one days.
  • In some embodiments, the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to the subject an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the ophthalmic solution is administered three, two, or one times a day. In some embodiments, an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is administered three times a day. In some embodiments, an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is administered two times a day. In some embodiments, an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is administered once daily.
  • In some embodiments, the present invention provides a method for treating dry eye disease in a subject, comprising topically administering to the subject an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, wherein the ophthalmic solution is administered at a higher strength in an initiation phase or exacerbation phase, followed by a lower strength in a maintenance phase, wherein each of the initiation phase, exacerbation phase, and maintenance phase is as described herein. In some embodiments, an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is administered four times a day in an initiation phase or exacerbation phase followed by three, two, or one times a day in a maintenance phase. In some embodiments, an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is administered three times a day in an initiation phase or exacerbation phase followed by two or one times a day in a maintenance phase. In some embodiments, an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is administered two times a day in an initiation phase followed by one time a day in a maintenance phase. In some embodiments, an ophthalmic solution comprising about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, is topically administered in an initiation phase or exacerbation phase, followed by topical administration of an ophthalmic solution comprising less than about 0.5% w/v reproxalap, or a pharmaceutically acceptable salt thereof, in a maintenance phase, wherein the administration frequency of each ophthalmic solution is selected from those as described above.
  • Exemplification
  • The following examples are intended to illustrate the invention and are not to be construed as being limitations thereon. Reproxalap can be synthesized as reported previously, for example, in WO 2006/127945, the entire content of which is incorporated herein by reference.
  • Abbreviations
  • CAE: controlled adverse environment
  • GMP: Good Manufacturing Practice
  • ICH: International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use
  • OD: right eye
  • OS: left eye
  • OU: both eyes
  • PRN: as needed
  • QD: once daily
  • QID: Four times daily
  • QS: as much as will suffice
  • EXAMPLE 1 A Multi-Center, Phase 2b, Randomized, Double-Masked, Parallel-Group, Vehicle-Controlled, Clinical Study to Assess the Safety and Efficacy of Reproxalap Ophthalmic Solution (0.25% and 0.1%) Compared to Vehicle in Subjects with Dry Eye Disease
  • Objectives:
      • To evaluate the efficacy of Reproxalap Ophthalmic Solutions (0.25% and 0.1%) on baseline to weeks 2, 4, 8, and 12 change scores for sign and symptom endpoints of dry eye disease.
      • To evaluate effect sizes for efficacy endpoints of Reproxalap Ophthalmic Solutions (0.25% and 0.1%) vs vehicle for the treatment of the signs and symptoms of dry eye disease to confirm the endpoint selection and sample size for Phase 3 studies.
      • To evaluate the safety and tolerability of Reproxalap Ophthalmic Solutions (0.25% and 0.1%) to vehicle for the treatment of the signs and symptoms of dry eye disease.
  • Investigational Product:
      • 1) Reproxalap Ophthalmic Solution (0.25%)
      • 2) Reproxalap Ophthalmic Solution (0.1%)
      • 3) Vehicle Ophthalmic Solution
  • In the Phase 2b study, reproxalap was formulated as an ophthalmic solution as described in the specification.
  • Duration: A subject's participation was estimated to be approximately 14 weeks (98 days).
  • Dosage/Dose Regimen/Instillation/Application/Use: Screening: Between Visits 1 and 2, all subjects received 14 consecutive days (±2) of Run-in (vehicle) ocular drops self-administered QID in both eyes.
  • Treatment: During the 12-week (84 ±3 days) treatment period, Reproxalap Ophthalmic Solution at concentrations of 0.1%, 0.25%, or vehicle ophthalmic solution was administered QID by bilateral topical ocular dosing. Subjects were randomized to one of three treatment groups (1:1:1) to receive study drug after the Post-CAE® assessments at Visit 2.
  • Summary of Visit Schedule: Six visits over the course of approximately 14 weeks
      • Visit 1=Day −14±2, CAE® Screening
      • Visit 2=Day 1, CAE® Confirmation/Baseline
      • Visit 3=Day 15±2, 2-Week Follow-Up
      • Visit 4=Day 29±2, 4-Week Follow-Up
      • Visit 5=Day 57±3, 8-Week Follow-Up
      • Visit 6=Day 85±3, 12-Week CAE® Follow-Up & Study Exit
  • Condition/Disease: Dry Eye Disease (DED)
  • Inclusion Criteria: Subjects for treatment were based on the following criteria:
      • 1 Been at least 18 years of age of either gender and any race;
      • 2 Provide written informed consent and sign the Health Information Portability and Accountability Act (HIPAA) form;
      • 3 Had a reported history of dry eye for at least six months prior to Visit 1;
      • 4 Had a history of use or desire to use eye drops for dry eye symptoms within six months of Visit 1;
      • 5 Reported a score of ≥2 on the Ora Calibra® Ocular Discomfort & 4-Symptom Questionnaire in at least one symptom at Visit 1 and Visit 2 Pre-CAE®;
      • 6 Had a Schirmer's Test score of ≤10 mm and ≥1 mm at Visit 1 and Visit 2;
      • 7 Had a tear film break-up time (TFBUT®) ≤5 seconds at Visit 1 and Visit 2 Pre-CAE®;
      • 8 Had a corneal fluorescein staining score of ≥2 in at least one region (e.g., inferior, superior, or central) at Visit 1 and Visit 2 Pre-CAE®;
      • 9 Have a sum corneal fluorescein staining score of ≥4, based on the sum of the inferior, superior, and central regions, at Visit 1 and Visit 2 Pre-CAE®;
      • 10 Had a total Lissamine green conjunctival score of ≥2, based on the sum of the temporal and nasal regions at Visit 1 and Visit 2 Pre-CAE®;
      • 11 Demonstrated a response to the CAE® at Visits 1 and 2 as defined by:
        • A. Having at least a ≥1 point increase in fluorescein staining in the inferior region in at least one eye following CAE® exposure;
        • B. Reporting an Ocular Discomfort score ≥3 at two or more consecutive time points in at least one eye during CAE® exposure (if a subject had an Ocular Discomfort rating of 3 at time=0 for an eye, s/he must have reported an Ocular Discomfort rating of 4 for two consecutive measurements for that eye). Note: a subject could not have an Ocular Discomfort score of 4 at time=0);
  • 12 Had at least one eye, the same eye, satisfy all criteria for 6, 7, 8, 9, 10, and 11 above.
  • Exclusion Criteria: Subject were excluded based on the following criteria:
      • 1 Had any clinically significant slit lamp findings at Visit 1 that may have included active blepharitis, meibomian gland dysfunction (MGD), lid margin inflammation, or active ocular allergies that require therapeutic treatment, and/or in the opinion of the investigator, might have interfered with study parameters;
      • 2 Been diagnosed with an ongoing ocular infection (bacterial, viral, or fungal), or active ocular inflammation at Visit 1;
      • 3 Worn contact lenses within seven days of Visit 1 or anticipate using contact lenses during the study;
      • 4 Used any eye drops within 2 hours of Visit 1;
      • 5 Had laser-assisted in situ keratomileusis (LASIK) surgery within the last 12 months;
      • 6 Used cyclosporine 0.05% or lifitegrast 5.0% ophthalmic solution within 90 days of Visit 1;
      • 7 Had any planned ocular and/or lid surgeries over the study period or any ocular surgery within 6 months of Visit 1;
      • 8 Been using or anticipated using temporary punctal plugs during the study that had not been stable within 30 days of Visit 1;
      • 9 Been currently taking any topical ophthalmic prescription (including medications for glaucoma) or over-the-counter (OTC) solutions, artificial tears, gels or scrubs, and cannot discontinue these medications for the duration of the trial (excluding medications allowed for the conduct of the study);
      • 10 Had corrected visual acuity greater than or equal to logarithm of the minimum angle of resolution (logMAR) +0.7 as assessed by Early Treatment of Diabetic Retinopathy Study (ETDRS) scale in both eyes at Visit 1;
      • 11 Been a woman who is pregnant, nursing, or planning a pregnancy;
      • 12 Been unwilling to submit a urine pregnancy test at Visit 1 and Visit 6 (or early termination visit) if of childbearing potential. Non-childbearing potential was defined as a woman who is permanently sterilized (e.g., has had a hysterectomy or tubal ligation), or was postmenopausal (without menses for
      • 12 consecutive months);
      • 13 Been a man or woman of childbearing potential who was not using an acceptable means of birth control; acceptable methods of contraception include: hormonal—oral, implantable, injectable, or transdermal contraceptives; mechanical—spermicide in conjunction with a barrier such as a diaphragm or condom; intrauterine device (IUD); or surgical sterilization of partner. For non-sexually active males or females, abstinence may have been regarded as an adequate method of birth control; however, if the subject became sexually active during the study, he/she must have agreed to use adequate birth control as defined above for the remainder of the study;
      • 14 Had a known allergy and/or sensitivity to the test article or its components;
      • 15 Had a condition or be in a situation which the investigator feels may have put the subject at significant risk, confounded the study results, or interfered significantly with the subject's participation in the study;
      • 16 Been currently enrolled in an investigational drug or device study or have used an investigational drug or device within 30 days of Visit 1;
      • 17 Previously used reproxalap ophthalmic solution;
      • 18 Been currently using any medication known to cause ocular drying that was not used on a stable dosing regimen for at least 30 days prior to Visit 1;
      • 19 Been unable or unwilling to follow instructions, including participation in all study assessments and visits.
  • The following efficacy measures and endpoints were used in the study:
      • Lissamine green staining (Ora Calibra® scale); regions: inferior, superior, central, temporal, nasal, corneal sum, conjunctival sum, and total eye score)
      • Fluorescein staining (Ora Calibra® scale); regions: central, superior, inferior, temporal, nasal, corneal sum, conjunctival sum, and total eye score)
      • Tear film break-up time
      • Unanesthetized Schirmer's Test
      • Ora Calibra® Ocular Discomfort Scale
      • Ora Calibra® Ocular Discomfort & 4-Symptom Questionnaire
      • Ocular Surface Disease Index (OSDI)®
      • SANDE questionnaire
      • Tear Osmolarity
  • Safety Measures:
      • Visual acuity
      • Slit-lamp evaluation
      • Adverse event query
      • Intraocular Pressure (TOP)
      • Dilated fundoscopy
  • General Statistical Methods and Types of Analyses
  • Sample Size: The study sample size of 100 per group was selected based on prior Phase 2 and 3 clinical trial results using the DED Hybrid CAE study design with other development programs and the effect size seen in Phase 2a with reproxalap on change from baseline after four weeks of treatment. This sample size was deemed sufficient to assess the effect size on the DED sign and symptom endpoints with reproxalap vs vehicle, to confirm the endpoint selection and sample size needed for Phase 3 studies with reproxalap. A sample size of 100 per group provided 90% power at α=0.05 to detect an effect size of 0.26 for inferior Lissamine green staining (Ora Calibra® scale), assuming a common standard deviation of 0.56 and an effect size of 0.44 for ocular discomfort assessed with the Ora Calibra® Ocular Discomfort Scale assuming a common standard deviation of 0.97.
  • Efficacy Analysis
      • Evaluated baseline to weeks 2, 4, 8 and 12 change scores with reproxalap on DED sign and symptom endpoints (both pre-CAE and CAE endpoints). Each endpoint was analyzed at a two-sided alpha level of 0.05, and the overall type I error was not controlled for in this investigative study.
      • Evaluated effect size of baseline to weeks 2, 4, 8 and 12 change scores of reproxalap vs vehicle on DED sign and symptom endpoints (both pre-CAE and CAE endpoints) to confirm the endpoint selection for primary outcome parameters and sample size for Phase 3 studies with reproxalap.
      • Sub-group analyses on effect size of baseline to weeks 2, 4, 8 and 12 change scores of reproxalap vs vehicle on DED sign and symptom endpoints (both pre-CAE and CAE endpoints) [Subgroups were prospectively detailed in the Statistical Analysis Plan (SAP)].
  • TABLE 1
    Summary of Subject Disposition
    Reproxalap (0.1%) Reproxalap (0.25%) Vehicle All Subjects
    N = 100 N = 100 N= 100 N = 300
    Intent-to-Treat Population 100 (100.0%) 100 (100.0%) 100 (100.0%) 300 (100.0%)
    Per Protocol Population 97 (97.0%) 86 (86.0%) 98 (98.0%) 281 (93.7%)
    Safety Population 100 (100.0%) 100 (100.0%) 100 (100.0%) 300 (100.0%)
    Study Completion
    Completed 97 (97.0%) 88 (88.0%) 99 (99.0%) 284 (94.7%)
    Discontinued 3 (3.0%) 12 (12.0%) 1 (1.0%) 16 (5.3%)
    Reason for Study Withdrawal
    Adverse Events 2 (2.0%) 10 (10.0%) 0 12 (4.0%)
    Administrative Reasons 1 (1.0%) 0 0 1 (0.3%)
    Withdrawal by Subject 0 1 (1.0%) 1 (1.0%) 2 (0.7%)
    Other 0 1 (1.0%) 0 1 (0.3%)
  • TABLE 2
    Phase 2b AE Summary
    Reproxalap 0.1% Reproxalap 0.25% Vehicle All Subjects
    (N = 100) (N = 100) (N = 100) (N = 300)
    Number of Ocular TEAEs 47  111  15  173
    Number of Subjects with Ocular TEAEs 38 (41.0%) 93 (93.0%) 13 (13.0%) 144 (48.0%)
    Mild 37 (37.0%) 84 (84.0%) 12 (12.0%) 133 (44.3%)
    Moderate 1 (1.0%) 8 (8.0%) 1 (1.0%) 10 (3.3%)
    Severe 0 1 (1.0%) 0 1 (0.3%)
    Instillation Site Pain Total 37 (37.0%) 93 (93.0%) 2 (2.0%) 132 (44.0%)
    Prior to Day 15 31 (31.0%) 89 (89.0%) 1 (1.0%) 121 (40.3%)
    Day 15-Day 28 4 (4.0%) 4 (4.0%) 0 8 (2.7%)
    Day 29-Day 56 2 (2.0%) 0 1 (1.0%) 3 (1.0%)
    After Day 56 0 0 0  0
    Number of Subjects with TEAEs 1 10 
    leading to Discontinuation
    Prior to Day 15  1*  7**
    Day 15-Day 28 0   2*** 0 11 (3.7%)
    Day 29-Day 56 0 0
    After Day 56 0 1 (SAE)
    Number of SAEs 1 (peripheral 1 (chest pain, 0 2 (0.7%)
    vertigo, not not related)
    related, stayed
    on study)
    *Subject discontinued at Day 12
    **Subjects discontinued on the following Days: 2, 3, 5, 5, 5, 12, 14
    ***Subjects discontinued on the following Days: 15, 16
  • The phase 2b data are shown in FIGS. 1 through 9 and Tables 1 through 3.
  • Key Observations From Phase 2b Clinical Trial
      • 1. Early onset of effect from Phase 2b evidenced across multiple signs and symptoms
        • Majority (>50-100%) of effect vs vehicle seen at the first study endpoint (Week 2 or 4) in 0.25% group:
        • Positive early onset for 3 out of 4 symptom endpoints: ODS, OD4SQ, OSDI
          • Negative for SANDE
        • Positive early onset for 3 out of 4 sign endpoints: Lissamine green total score, fluorescein total score, tear osmolarity
          • Negative for TFBUT® (met definition at week 4)
          • Schirmer's Test only assessed at week 12
      • 2. Dose response was demonstrated between 0.1% and 0.25% dose strengths
      • 3. 0.1% reproxalap matched higher dose effects at later time points
        • Clearest effect with signs, especially ocular staining
        • Compliance poorest in 0.25% group (8% non-compliant vs 3% in the 0.1% group and 1% in the vehicle group)
      • 4. Vehicle effect increased with study duration
        • Clearest effect was observed with signs, especially ocular staining
        • Normal pattern in DED with plateau around two to three months
        • QID vehicle in Phase 2b was expected to have increased this effect
  • TABLE 3
    Phase 2b Clinical Trial Results Heat Map: Broad Phase 2a Activity Reproduced
    Reproxalap Phase 2b DED Results Heat Map
    Intent to Treat (ITT) Population with Observed Data Only
    Non-CAE and Pre-CAE
    Endpoint-Specific Worst Eye (where applicable)
    Reproxalap 0.1% Reproxalap 0.25%
    Effect vs. Vehicle Effect vs. Vehicle
    Drug Dose Size p-value ANCOVA Size p-value ANCOVA
    Trend? Trend? >0.5 V3 V4 V5 V6 >0.5 V3 V4 V5 V6
    Symptom Measures
    Ocular Discomfort Scale (0-4)
    OD & 4-Symptom Overall Ocular (0-5) c a
    Questionnaire Discomfort
    Burning (0-5) N/A b N/A
    Dryness (0-5) c a
    Grittiness (0-5) c
    Stinging (0-5) b c c a
    Ocular Surface  (0-100) N/A
    Disease Index
    (OSDI)
    SANDE Severity (0-100 mm) N/A b
    Questionnaire Frequency (0-100 mm)
    Sign Measures
    Lisamine Green Total Score (0-20; Σ 5x)
    Staining (all five regions)
    Corneal Sum Score (0-12; Σ 3x) c
    (Inferior, Superior,
    Central)
    Conjunctival Sum  (0-8; Σ 2x) c
    Score (Nasal and
    Temporal)
    Inferior (0-4)
    Superior (0-4) N/A a N/A N/A
    Central (0-4) N/A N/A N/A
    Temporal (0-4) b
    Nasal (0-4) N/A N/A b c
    Fluorescein Total Score (0-20; Σ 5x) b
    Staining (all five regions)
    Corneal Sum Score (0-12; Σ 3x) a N/A
    (Inferior, Superior,
    and Central)
    Conjunctival Sum  (0-8; Σ 2x) N/A b b
    Score (Nasal and
    Temporal)
    Inferior (0-4) c
    Superior (0-4) N/A N/A N/A
    Central (0-4) N/A N/A N/A
    Temporal (0-4) N/A N/A b b
    Nasal (0-4) N/A a c a b
    Tear Film Break-Up Time (sec) N/A N/A N/A N/A
    Schirmer's Test (mm)
    Osmolarity (mOsm/L)
    P-value Key
    P < 0.05 a
    P < 0.10 b
    P < 0.15 c {circumflex over ( )}wrong signal defined as an effect less than
    wrong signal{circumflex over ( )} N/A Vehicle, or a worsening vs. baseline
  • While we have described a number of embodiments of this invention, it is apparent that our basic examples may be altered to provide other embodiments that utilize the compounds and methods of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments that have been represented by way of example.
  • All publications, patents, patent applications and other documents cited in this application are hereby incorporated by reference in their entireties for all purposes to the same extent as if each individual publication, patent, patent application or other document were individually indicated to be incorporated by reference for all purposes.

Claims (29)

1-10. (canceled)
11. A method of treating dry eye disease in a subject, comprising topically administering to an eye of a subject with dry eye disease a therapeutically effective amount of an ophthalmic solution of claim 1 comprising reproxalap, or a pharmaceutically acceptable salt thereof, at about 0.2% w/v to about 0.3% w/v, and a pharmaceutically acceptable excipient comprising a cyclodextrin, wherein the cyclodextrin is selected from a hydroxyalkyl derivative of β-or γ-cyclodextrin, randomly methylated β-cyclodextrin, sulfobutylether β-cyclodextrin, sulfobutylether γ-cyclodextrin, a branched β- or γ-cyclodextrin derivative, and mixtures thereof, or a pharmaceutically acceptable salt thereof, wherein the reproxalap and cyclodextrin are present in a ratio of about 1:2, about 1:3, about 1:4, or about 1:5 on a mole:mole basis, wherein the ophthalmic solution is topically administered to the eye with dry eye disease four times a day in an initiation phase or exacerbation phase followed by three times a day, twice a day, or once a day, or as needed in a maintenance phase.
12. (canceled)
13. The method of claim 11, wherein the ophthalmic solution is topically administered to the eye with dry eye disease three times a day in the maintenance phase.
14. The method of claim 11, wherein the ophthalmic solution is topically administered to the eye with dry eye disease two times a day in the maintenance phase.
15. The method of claim 11, wherein the ophthalmic solution is topically administered to the eye with dry eye disease once a day in the maintenance phase.
16. The method of claim 11, wherein the treatment is for at least 12 weeks.
17. The method of claim 11, wherein the ophthalmic solution is topically administered to the eye with dry eye disease four times a day for about 2 to 6 weeks in an initiation phase or exacerbation phase followed by administration three times a day, two times a day, once a day, or as needed in a maintenance phase.
18. The method of claim 17, wherein the maintenance phase is administration three times a day, two times a day, once a day, or as needed for about 6 to 10 weeks.
19. The method of claim 18, wherein the maintenance phase is administration two times a day.
20. A method of treating dry eye disease in a subject, comprising:
topically administering to an eye of a subject with dry eye disease a therapeutically effective amount of an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient comprising a cyclodextrin,
wherein the reproxalap is at about 0.2% w/v to about 0.3% w/v, and the cyclodextrin is selected from a hydroxyalkyl derivative of β- or γ-cyclodextrin, randomly methylated β-cyclodextrin, sulfobutylether β-cyclodextrin, sulfobutylether γ-cyclodextrin, and a branched β- or γ-cyclodextrin derivative, and mixtures thereof, or a pharmaceutically acceptable salt thereof, wherein the reproxalap and cyclodextrin are present in a ratio of about 1:2, about 1:3, about 1:4, or about 1:5 on a mole:mole basis, and
wherein the ophthalmic solution is administered four times a day.
21. The method of claim 20, wherein reproxalap is at about 0.25% w/v and the pharmaceutically acceptable excipient comprises sulfobutylether-β-cyclodextrin hydroxypropyl-β-cyclodextrin, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein the sulfobutylether-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, or a mixture thereof, or a pharmaceutically acceptable salt thereof, is at about 7% w/v.
22. The method of claim 20, wherein reproxalap is at a concentration of 0.25% w/v and the pharmaceutically acceptable excipient comprises sulfobutylether-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein the sulfobutylether-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, or a mixture thereof, or a pharmaceutically acceptable salt thereof, is at about 11% w/v.
23. A method of treating dry eye disease in a subject, comprising topically administering to an eye of a subject with dry eye disease a therapeutically effective amount of an ophthalmic solution comprising reproxalap, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient comprising a cyclodextrin,
wherein the reproxalap is at about 0.2% w/v to about 0.3% w/v, and the cyclodextrin is selected from a hydroxyalkyl derivative of β- or γ-cyclodextrin, randomly methylated β-cyclodextrin, sulfobutylether β-cyclodextrin, sulfobutylether γ-cyclodextrin, and a branched β- or γ-cyclodextrin derivative, and mixtures thereof, or a pharmaceutically acceptable salt thereof, wherein the reproxalap and cyclodextrin are present in a ratio of about 1:2, about 1:3, about 1:4, or about 1:5 on a mole:mole basis, and
wherein the ophthalmic solution is administered at least four times a day in an initiation phase or exacerbation phase, followed by three times a day, two times a day, once a day, or as needed in a maintenance phase.
24. The method of claim 23, wherein the ophthalmic solution is administered two times a day in the maintenance phase.
25. The method of claim 23, wherein reproxalap is at about 0.25% w/v and the pharmaceutically acceptable excipient comprises sulfobutylether-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein the sulfobutylether-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, or a mixture thereof; or a pharmaceutically acceptable salt thereof of, is at about 7% w/v.
26. The method of claim 23, wherein reproxalap is at a concentration of 0.25% w/v and the pharmaceutically acceptable excipient comprises sulfobutylether-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, or a mixture thereof, or a pharmaceutically acceptable salt thereof, wherein the sulfobutylether-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, or a mixture thereof, or a pharmaceutically acceptable salt thereof, is at about 11% w/v.
27. The method of claim 23, wherein the ophthalmic solution is administered at a dose strength to achieve an early onset of effect.
28. The method of claim 27, wherein the early onset effect is for dry eye symptoms of one or more of: ocular dryness, itchiness, tearing, burning, stinging, grittiness, cloudy vision, sensitivity to environment, and stringy ocular secretion.
29. The method of claim 27, wherein the early onset of effect is for dry eye signs of one or more of: ocular vital staining, tear film break-up time, tear osmolarity, and tear volume.
30. The method of claim 11, wherein the ophthalmic solution further comprises a tonicity agent selected from the group consisting of dextrose, potassium chloride, propylene glycol, and sodium chloride.
31. The method of claim 30, wherein the tonicity agent is at a concentration of about 0.45%, 0.4%, 0.35%, 0.3%, 0.25%, 0.2%, 0.15%, or 0.1% w/v.
32. The method of claim 30, wherein the tonicity agent comprises about 0.2 to about 0.3% w/v sodium chloride.
33. The method of claim 11, wherein the reproxalap and cyclodextrin are present in a ratio of about 1:3 on a mole:mole basis.
34. The method of claim 11, wherein the reproxalap and cyclodextrin are present in a ratio of about 1:4 on a mole:mole basis.
35. The method of claim 11, wherein reproxalap, or a pharmaceutically acceptable salt thereof, is at about 0.25% w/v, and the cyclodextrin comprises sulfobutylether-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, or a mixture thereof; or a pharmaceutically acceptable salt thereof, at about 7% w/v.
36. The method of claim 11, wherein reproxalap, or a pharmaceutically acceptable salt thereof, is at about 0.25% w/v, and the cyclodextrin comprises sulfobutylether-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, or a mixture thereof; or a pharmaceutically acceptable salt thereof, at about 11% w/v.
37. The method of claim 30, wherein the reproxalap and cyclodextrin are present in a ratio of about 1:4 on a mole:mole basis.
38. The method of claim 11, wherein the cyclodextrin is selected from a hydroxyethyl or hydroxypropyl derivative of β-cyclodextrin or γ-cyclodextrin, glucosyl-β-cyclodextrin, glucosyl-γ-cyclodextrin, sulfobutylether-β-cyclodextrin, and hydroxypropyl-β-cyclodextrin; or a pharmaceutically acceptable salt thereof.
US17/450,623 2018-09-25 2021-10-12 Formulations for treatment of dry eye disease Pending US20220133629A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/450,623 US20220133629A1 (en) 2018-09-25 2021-10-12 Formulations for treatment of dry eye disease

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201862736417P 2018-09-25 2018-09-25
US201962824233P 2019-03-26 2019-03-26
US16/582,720 US11197821B2 (en) 2018-09-25 2019-09-25 Formulations for treatment of dry eye disease
US17/450,623 US20220133629A1 (en) 2018-09-25 2021-10-12 Formulations for treatment of dry eye disease

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/582,720 Continuation US11197821B2 (en) 2018-09-25 2019-09-25 Formulations for treatment of dry eye disease

Publications (1)

Publication Number Publication Date
US20220133629A1 true US20220133629A1 (en) 2022-05-05

Family

ID=69949487

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/582,720 Active 2040-01-12 US11197821B2 (en) 2018-09-25 2019-09-25 Formulations for treatment of dry eye disease
US17/450,623 Pending US20220133629A1 (en) 2018-09-25 2021-10-12 Formulations for treatment of dry eye disease

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/582,720 Active 2040-01-12 US11197821B2 (en) 2018-09-25 2019-09-25 Formulations for treatment of dry eye disease

Country Status (6)

Country Link
US (2) US11197821B2 (en)
EP (1) EP3856478A4 (en)
JP (1) JP2022502499A (en)
CN (2) CN115869258A (en)
CA (1) CA3113462A1 (en)
WO (1) WO2020068986A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11583529B2 (en) 2017-10-10 2023-02-21 Aldeyra Therapeutics, Inc. Treatment of inflammatory disorders
US11701331B2 (en) 2013-01-23 2023-07-18 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US11786518B2 (en) 2019-03-26 2023-10-17 Aldeyra Therapeutics, Inc. Ophthalmic formulations and uses thereof

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1888548B1 (en) 2005-05-26 2012-08-22 Neuron Systems, Inc Quinoline derivative for the treatment of retinal diseases
JP6947406B2 (en) 2015-08-21 2021-10-13 アルデイラ セラピューティクス, インコーポレイテッド Deuterated compounds and their use
CN109152774A (en) 2016-05-09 2019-01-04 奥尔德拉医疗公司 The combined therapy of ophthalmic inflammatory illness and disease
CN112714762A (en) 2018-08-06 2021-04-27 奥尔德拉医疗公司 Polymorphic compounds and uses thereof
US20230228744A1 (en) * 2020-06-04 2023-07-20 Aldeyra Therapeutics, Inc. Dry eye disease biomarkers and their use for treatment
US20220211691A1 (en) * 2021-01-07 2022-07-07 Aldeyra Therapeutics, Inc. Treatment of dry eye disease
WO2022150580A1 (en) * 2021-01-07 2022-07-14 Aldeyra Therapeutics, Inc. Treatment of dry eye disease

Family Cites Families (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1435721A (en) 1972-05-18 1976-05-12 Lilly Industries Ltd Benzoxazole derivatives
JPS6192592A (en) 1984-10-12 1986-05-10 Norin Suisansyo Shokuhin Sogo Kenkyusho Production of branched cyclodextrin
NZ225045A (en) 1987-07-01 1990-06-26 Janssen Pharmaceutica Nv Antiviral pharmaceutical compositions containing cyclodextrin and an antiviral agent
US5002935A (en) 1987-12-30 1991-03-26 University Of Florida Improvements in redox systems for brain-targeted drug delivery
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
CA2072001C (en) 1990-10-22 1998-11-03 Nimai C. De Method and composition for cleaning contact lenses
US5668117A (en) 1991-02-22 1997-09-16 Shapiro; Howard K. Methods of treating neurological diseases and etiologically related symptomology using carbonyl trapping agents in combination with previously known medicaments
JP2682353B2 (en) 1991-11-20 1997-11-26 武田薬品工業株式会社 Oral pharmaceutical composition and method for producing the same
US6444221B1 (en) 1992-06-30 2002-09-03 Howard K. Shapiro Methods of treating chronic inflammatory diseases using carbonyl trapping agents
US20050090553A1 (en) 1992-06-30 2005-04-28 Shapiro Howard K. Compositions and method for treatment of chronic inflammatory diseases
US5472954A (en) 1992-07-14 1995-12-05 Cyclops H.F. Cyclodextrin complexation
TW401300B (en) 1992-12-25 2000-08-11 Senju Pharma Co Antiallergic composition for ophthalmic or nasal use
JP2746832B2 (en) 1993-05-14 1998-05-06 大鵬薬品工業株式会社 Ocular topical antiallergic agent
US5767109A (en) 1993-10-20 1998-06-16 Sanchez; Robert A. Complexing urushiols
US5576311A (en) 1994-11-30 1996-11-19 Pharmos Corporation Cyclodextrins as suspending agents for pharmaceutical suspensions
JP3297969B2 (en) 1994-12-26 2002-07-02 ライオン株式会社 Eye drops
FR2742053B1 (en) 1995-12-06 1998-03-06 Chauvin Lab Sa PHARMACEUTICAL COMPOSITIONS BASED ON MEQUITAZINE
JP3736916B2 (en) 1996-02-19 2006-01-18 株式会社サンコンタクトレンズ Disinfecting composition for hydrous soft contact lens and its use
WO1998005661A1 (en) 1996-08-06 1998-02-12 Pfizer Inc. Substituted pyrido- or pyrimido-containing 6,6- or 6,7-bicyclic derivatives
US6191127B1 (en) 1997-05-02 2001-02-20 Schering Aktiengesellschaft Substituted heterocycles and their use in medicaments
BR9814458A (en) 1997-09-02 2001-10-23 Du Pont Pharm Co Compound, pharmaceutical composition and method of treating affective disorders
JP3568108B2 (en) 1999-07-28 2004-09-22 松下電器産業株式会社 Method for transmitting location information of digital map and apparatus for implementing the method
AU1735001A (en) 1999-12-10 2001-06-18 Senju Pharmaceutical Co., Ltd. Cyclodextrin-containing pharmaceutical composition
JP4748289B2 (en) 2000-06-23 2011-08-17 ライオン株式会社 Eye drops, ophthalmic composition, and adsorption suppression method
US20040198828A1 (en) 2003-01-17 2004-10-07 Abelson Mark B. Combinational use of long-acting and short-acting anti-histamines for ocular allergies
US20060111318A1 (en) 2003-04-18 2006-05-25 Advanced Medicine Research Institute Agent for treating eye diseases
ATE488240T1 (en) 2003-04-18 2010-12-15 Advanced Medicine Res Inst AGENTS FOR TREATING DISEASES FOR APPLICATION TO THE EYE
CN100558360C (en) 2003-11-20 2009-11-11 奥特拉控股公司 The purposes of azanol compositions in the medicine of preparation alleviation degeneration of macula and other ophthalmic diseasess
JP2005187407A (en) 2003-12-25 2005-07-14 Lion Corp Ophthalmic composition for allergic eye disease
US20050234018A1 (en) 2004-04-15 2005-10-20 Allergan, Inc. Drug delivery to the back of the eye
JP2006008568A (en) 2004-06-24 2006-01-12 Cyclochem:Kk IgE ANTIBODY-SUPPRESSING AGENT AND FOOD
WO2006002473A1 (en) 2004-07-02 2006-01-12 Adelaide Research & Innovation Pty Ltd Method of controlling damage mediated by alpha, beta-unsaturated aldehydes
EP1893174A2 (en) 2005-05-10 2008-03-05 Cytophil, Inc. Injectable hydrogels and methods of making and using same
EP1888548B1 (en) 2005-05-26 2012-08-22 Neuron Systems, Inc Quinoline derivative for the treatment of retinal diseases
EP1967186B1 (en) 2005-12-27 2015-03-11 Lion Corporation Composition for soft contact lens and adsorption suppressing method
JP4466875B2 (en) 2006-04-05 2010-05-26 ライオン株式会社 Eye drops for soft contact lenses
JP5194218B2 (en) 2006-06-05 2013-05-08 株式会社メニコンネクト Preservation method of hydrous contact lens and hydrous contact lens preserved by the preservation method
US8158609B1 (en) 2006-11-02 2012-04-17 Novartis Ag Use of cyclodextrins as an active ingredient for treating dry AMD and solubilizing drusen
NZ584599A (en) 2007-10-05 2012-07-27 Acucela Inc Alkoxy compounds for disease treatment
CA2733966A1 (en) 2008-08-12 2010-02-18 Sirtris Pharmaceuticals, Inc. Benzoxazoles, benzthiazoles and related analogs as sirtuin modulators
JP5773877B2 (en) 2008-10-22 2015-09-02 アキュセラ インコーポレイテッド Compounds for treating eye diseases and disorders
JP2012520880A (en) 2009-03-17 2012-09-10 アーシエックス セラピューティックス, インコーポレイテッド Ketotifen ophthalmic formulation and method of use
US20110105450A1 (en) 2009-06-05 2011-05-05 Aciex Therapeutics, Inc. Ophthalmic formulations of fluticasone and methods of use
WO2011008202A1 (en) 2009-07-15 2011-01-20 Vanderbilt University Isoketal scavengers and mitigation of disorders involving oxidative injury
EP2509596B1 (en) 2009-12-08 2019-08-28 Case Western Reserve University Gamma aminoacids for treating ocular disorders
CA2782015C (en) 2009-12-11 2020-08-25 Neuron Systems, Inc. Topical ophthalmic compositions and methods for the treatment of macular degeneration
KR20120131162A (en) 2010-02-26 2012-12-04 제논 파마슈티칼스 인크. Pharmaceutical compositions of spiro-oxindole compound for topical administration and their use as therapeutic agents
US10463687B2 (en) 2011-01-20 2019-11-05 Cornell University Treatments for retinal disorders
US9302013B2 (en) 2011-01-31 2016-04-05 Termira Ab Active principle for mitigating undesired medical conditions
TWI544922B (en) 2011-05-19 2016-08-11 愛爾康研究有限公司 High concentration olopatadine ophthalmic composition
US20130190500A1 (en) 2011-12-12 2013-07-25 Neuron Systems, Inc. Process to prepare 6-chloro-3-amino-2-(2-hydroxypropyl)-1-azanaphthalene
US20130165419A1 (en) 2011-12-21 2013-06-27 Insite Vision Incorporated Combination anti-inflammatory ophthalmic compositions
GB201200192D0 (en) 2012-01-06 2012-02-22 Rosemont Pharmaceuticals Ltd Methotrexate composition
MX2015005839A (en) 2012-11-08 2015-12-17 Clearside Biomedical Inc Methods and devices for the treatment of ocular diseases in human subjects.
MX2015007916A (en) 2012-12-20 2016-04-07 Aldeyra Therapeutics Inc Peri-carbinols.
KR102243169B1 (en) 2013-01-23 2021-04-22 알데이라 테라퓨틱스, 아이엔씨. Toxic aldehyde related diseases and treatment
EP2948436A4 (en) 2013-01-25 2016-07-13 Aldeyra Therapeutics Inc Novel traps in the treatment of macular degeneration
US20160151410A1 (en) 2013-07-02 2016-06-02 The Trustees Of Columbia University In The City Of New York Clearance of bioactive lipids from membrane structures by cyclodextrins
EP3151818A4 (en) 2014-06-04 2018-05-30 Case Western Reserve University Compositions and methods of treating diabetic retinopathy
US10363231B2 (en) 2014-11-24 2019-07-30 Case Western Reserve University Compounds and methods of treating ocular disorders
JP6947406B2 (en) 2015-08-21 2021-10-13 アルデイラ セラピューティクス, インコーポレイテッド Deuterated compounds and their use
EP3337470A4 (en) 2015-08-21 2019-02-27 Aldeyra Therapeutics, Inc. Aldehyde conjugates and uses thereof
US10426790B2 (en) 2016-02-28 2019-10-01 Aldeyra Therapeutics, Inc. Treatment of allergic eye conditions with cyclodextrins
CN109152774A (en) * 2016-05-09 2019-01-04 奥尔德拉医疗公司 The combined therapy of ophthalmic inflammatory illness and disease
CA3032521A1 (en) 2016-08-22 2018-03-01 Aldeyra Therapeutics, Inc. Aldehyde trapping compounds and uses thereof
AU2017317529A1 (en) 2016-08-22 2019-02-21 Aldeyra Therapeutics, Inc. Aldehyde trapping compounds and methods of use thereof
AU2017336765B2 (en) 2016-09-28 2023-06-08 Medicon Pharmaceuticals, Inc. Compositions and methods for treating ophthalmic conditions
EP3596040B1 (en) 2017-03-16 2023-10-11 Aldeyra Therapeutics, Inc. Polymorphic salt of 6-chloro-3-amino-2(2-hydroxypropyl)quinoline and uses thereof
MX2020003425A (en) 2017-10-10 2020-07-29 Aldeyra Therapeutics Inc Treatment of inflammatory disorders.
WO2020018498A1 (en) 2018-07-16 2020-01-23 Aldeyra Therapeutics, Inc. Cyclodextrin formulations
WO2020028820A1 (en) 2018-08-03 2020-02-06 Aldeyra Therapeutics, Inc. Topical compositions and methods of preparation and use
CN112714762A (en) 2018-08-06 2021-04-27 奥尔德拉医疗公司 Polymorphic compounds and uses thereof
WO2020198064A1 (en) 2019-03-26 2020-10-01 Aldeyra Therapeutics, Inc. Ophthalmic formulations and uses thereof
EP3962894A4 (en) 2019-05-02 2023-01-11 Aldeyra Therapeutics, Inc. Polymorphic compounds and uses thereof
US20220089542A1 (en) 2019-05-02 2022-03-24 Aldeyra Therapeutics, Inc. Process for preparation of aldehyde scavenger and intermediates

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11701331B2 (en) 2013-01-23 2023-07-18 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US11771664B2 (en) 2013-01-23 2023-10-03 Aldeyra Therapeutics, Inc. Toxic aldehyde related diseases and treatment
US11583529B2 (en) 2017-10-10 2023-02-21 Aldeyra Therapeutics, Inc. Treatment of inflammatory disorders
US11786518B2 (en) 2019-03-26 2023-10-17 Aldeyra Therapeutics, Inc. Ophthalmic formulations and uses thereof

Also Published As

Publication number Publication date
CN113056353A (en) 2021-06-29
WO2020068986A1 (en) 2020-04-02
CN113056353B (en) 2022-11-01
JP2022502499A (en) 2022-01-11
EP3856478A4 (en) 2022-06-08
EP3856478A1 (en) 2021-08-04
US11197821B2 (en) 2021-12-14
CN115869258A (en) 2023-03-31
US20200121591A1 (en) 2020-04-23
CA3113462A1 (en) 2020-04-02

Similar Documents

Publication Publication Date Title
US11197821B2 (en) Formulations for treatment of dry eye disease
US10561607B2 (en) Pharmaceutical compositions comprising gels and methods for fabricating thereof
US11786518B2 (en) Ophthalmic formulations and uses thereof
US20100311688A1 (en) Ophthalmic formulations, methods of manufacture, and methods of using same
RU2746592C1 (en) Composition for the prevention or treatment of visual impairment containing ursodeoxycholic acid
US20230228744A1 (en) Dry eye disease biomarkers and their use for treatment
US10076522B2 (en) Systems and methods for treating bacterial infection
AU2011334617B2 (en) Folic acid - Ramipril combination: cellprotective, neuroprotective and retinoprotective ophtalmologic compositions
US20220211691A1 (en) Treatment of dry eye disease
KR20210127197A (en) Formulation of 4-(7-hydroxy-2-isopropyl-4-oxo-4H-quinazolin-3-yl)-benzonitrile
US20170112936A1 (en) Pharmaceutical compositions comprising gels and methods for fabricating thereof
US20200155450A1 (en) Pharmaceutical compositions comprising gels and methods for fabricating thereof
WO2022150580A1 (en) Treatment of dry eye disease
US20190328772A1 (en) Ophthalmic compositions comprising a cyclodextrin as sole active agent
US11433021B2 (en) Palonosetron for the treatment or prevention of nausea and vomiting
US20200345637A1 (en) Suspension compositions of multi-target inhibitors
Korenfeld et al. OCS-01 (Novel Topical Dexamethasone Formulation) in Inflammation and Pain Post Cataract Surgery: A Randomized, Double-Masked, Vehicle-Controlled Study
JP7417531B2 (en) Methods of using selective SYK inhibitors and pharmaceutical compositions
WO2022112511A1 (en) Pharmaceutical composition for treatment of inner ear disorders through local administration in the tympanic area
WO2022187306A1 (en) Compositions and methods for treatment of blepharitis
Maggs Managing chronic ocular conditions in dogs and cats.

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALDEYRA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CLARK, DAVID;BRADY, TODD;MACDONALD, SUSAN;AND OTHERS;REEL/FRAME:057777/0548

Effective date: 20191001

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION