EP2648711A2 - Compositions et méthodes pour le traitement d'affections ophtalmiques - Google Patents

Compositions et méthodes pour le traitement d'affections ophtalmiques

Info

Publication number
EP2648711A2
EP2648711A2 EP11847841.1A EP11847841A EP2648711A2 EP 2648711 A2 EP2648711 A2 EP 2648711A2 EP 11847841 A EP11847841 A EP 11847841A EP 2648711 A2 EP2648711 A2 EP 2648711A2
Authority
EP
European Patent Office
Prior art keywords
fenretinide
composition
micron
powder
rbp
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.)
Withdrawn
Application number
EP11847841.1A
Other languages
German (de)
English (en)
Inventor
Mark Edgar
Jay Lichter
Nathan L. Mata
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.)
Acucela Inc
Original Assignee
Revision 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 Revision Therapeutics Inc filed Critical Revision Therapeutics Inc
Publication of EP2648711A2 publication Critical patent/EP2648711A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/145Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • 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

Definitions

  • compositions described herein are directed to the treatment of ophthalmic conditions.
  • G Geographic atrophy
  • RPE retinal pigment epithelium
  • End plates shed by photoreceptors require processing by the RPE to move visual chromophore material derived from retinol through the visual cycle and return it to the photoreceptors.
  • Vitamin A retinol
  • RBP retinol binding protein
  • transthyretin One of the effects of the visual cycle is production of toxic end products lipofuscin and A2E.
  • compositions comprising (a) fenretinide powder in an amount therapeutically effective to reduce the serum RBP level in a human subject by at least about 60% or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject, and (b) one or more pharmaceutically acceptable excipients, wherein the diameter of at least 80% of the particles in the fenretinide powder in the composition is about or less than 10 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder in the composition is in the range of about 2 micron to about 10 micron.
  • the diameter of at least 80% of the particles in the fenretinide powder in the composition is in the range of about 2 micron to about 6 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder in the composition is less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder in the composition is about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron.
  • the fenretinide powder in the composition is substantially free of particles having a diameter of 10 micron or more. In certain embodiments, the diameter of at least 90% of the particles in the fenretinide powder in the composition is less than about 10 micron.
  • compositions comprising (a) fenretinide powder that has been produced from a suspension comprising corn oil and/or polysorbate in an amount therapeutically effective to reduce the serum RBP level by at least about 60% in a human subject or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject, and (b) one or more pharmaceutically acceptable excipients.
  • compositions comprising (a) a non-micronized fenretinide powder in an amount therapeutically effective to reduce the serum RBP level by at least about 60% in a human subject or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject, and (b) one or more pharmaceutically acceptable excipients.
  • compositions comprising (a) fenretinide powder, characterized by an X-ray powder diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1, in an amount therapeutically effective to reduce the serum RBP level by at least about 60% in a human subject or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject, and (b) one or more pharmaceutically acceptable excipients.
  • compositions comprising (a) fenretinide powder, characterized by a DSC phase transition at a temperature of about 164°C, in an amount therapeutically effective to reduce the serum RBP level by at least about 60% in a human subject or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject, and (b) one or more pharmaceutically acceptable excipients.
  • suspension pharmaceutical compositions prepared by (a) premixing corn oil, polysorbate 80, and fenretinide powder, and (b) high shear mixing the premixed corn oil, polysorbate 80, and fenretinide powder.
  • the fenretinide powder does not aggregate in the suspension.
  • the diameter of at least 80% of the particles in the fenretinide powder in the composition is in the range of about 2 micron to about 10 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder in the composition is less than about 10 micron.
  • the one or more pharmaceutically acceptable excipients are selected from the group consisting of corn oil, polysorbate, micro crystalline cellulose, starch, hydro xypropylcellulose, sodium starch glycolate, isopropyl alcohol, magnesium stearate, and combinations thereof.
  • the composition comprises corn oil. In some embodiments, the composition is administered orally.
  • the composition is in a solid dosage form. In some embodiments, the solid dosage form is a tablet or a capsule. In certain embodiments, the composition is in suspension form. In some embodiments ,the diameter of at least 80% of the particles in the fenretinide powder in the composition is in the range of about 2 micron to about 6 micron. In some embodiments, the diameter of at least 80% of the particles in the fenretinide powder in the composition is about 6 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder in the composition is greater than about 2 micron.
  • the fenretinide powder is characterized by a predominant endotherm at about 164°C as measured by DSC and a powder x-ray diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1.
  • the therapeutically effective amount of fenretinide powder is at least about 100 mg. In some embodiments, the
  • therapeutically effective amount of fenretinide powder is at least about 300 mg.
  • kits for reducing the serum RBP level by at least about 60% in a human subject or reducing the rate of geographic atrophy lesion size increase in an eye of the human subject comprising administering the invention pharmaceutical composition to a human subject in need thereof.
  • methods for reducing the serum RBP level of a human subject by at least about 2 mg/dL to about 2.5 mg/dL comprising administering the invention pharmaceutical composition to a human subject in need thereof.
  • methods for treating Stargardt disease comprising administering the invention pharmaceutical composition to a human subject in need thereof.
  • the effective amount of the pharmaceutical composition is administered orally to the human.
  • the methods comprise multiple administrations of the effective amount of the pharmaceutical composition.
  • the methods induce or cause delayed dark adaptation after administration of the composition.
  • the methods reduce at least about 40% the rate of geographic atrophy lesion size increase after administration of the composition for 12 months. In certain embodiments, the methods reduce about 60% to about 80% the rate of geographic atrophy lesion size. In certain embodiments, the methods reduce the rate of geographic atrophy lesion size increase to less than about 1.0 mm 2 per year after administration of the composition for 12 months. In certain embodiments, the methods reduce the rate of geographic atrophy lesion size increase in less than about 0.5 mm 2 per year after administration of the composition for 12 months. In certain embodiments, the methods reduce the rate of geographic atrophy lesion size increase to about 0.5 mm 2 to about 1.0 mm 2 per year after administration of the composition for 12 months.
  • serum RBP monitoring kits for determining the therapeutically effective amount of fenretinide to reduce the serum RBP level by at least about 60% in a human subject or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject comprising an RBP assay unit, and a means for obtaining a fluid sample from a patient who has been administered fenretinide.
  • the means for obtaining a fluid sample comprises a needle and the fluid is blood or serum.
  • compositions or methods for treating geographic atrophy in a mammal comprising reducing the serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP level in the mammal by a desired percentage.
  • the desired percentage of serum retinol, serum retinol binding protein (RBP), and/or serum retinol- RBP reduction is relative to pre-therapeutic levels; in alternative embodiments, the desired percentage of serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP reduction is relative to a pre-determined threshold level.
  • the desired percentage of serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP reduction is at least about 60%, at least about 70%, or at least about 80%.
  • the desired percentage of serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP reduction is maintained for at least 1 week, for at least 1 month, for at least 6 months, for at least 12 months, for at least 18 months, for at least 24 months, for the lifetime of the mammal.
  • compositions or methods are also used to treat a lipofuscin-based retinal disease by reducing the serum level of retinol, RBP, and/or retinol-RBP in the body of a mammal, including embodiments wherein (a) the lipofuscin-based retinal disease is juvenile macular degeneration, including Stargardt Disease; (b) the lipofuscin-based retinal disease is dry form age-related macular degeneration; (c) the lipofuscin-based retinal disease is cone-rod dystrophy; (d) the lipofuscin-based retinal disease is retinitis pigmentosa; (e) the lipofuscin-based retinal disease is wet-form age-related macular degeneration; (f) the lipofuscin-based retinal disease is or presents geographic atrophy and/or photoreceptor degeneration; or (g) the lipofuscin-based retinal disease is a lipofuscin-based retinal disease by reducing the serum level of
  • compositions or methods for treating a lipofusin-based retinal disease in a mammal comprising reducing the serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP level in the mammal by a desired percentage.
  • the desired percentage of serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP reduction is relative to pre-therapeutic levels; in alternative embodiments, the desired percentage of serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP reduction is relative to a pre-determined threshold level.
  • the desired percentage of serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP reduction is at least about 60%, at least about 70%, or at least about 80%. In certain embodiments, the desired percentage of serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP reduction is maintained for at least 1 week, for at least 1 month, for at least 6 months, for at least 1 year, for the lifetime of the mammal.
  • compositions or methods for treating a lipofuscin-based retinal disease in a mammal comprising maintaining the serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP level in the mammal within a desired range.
  • the desired range of serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP is greater than a level that leads to diseases or conditions associated with Vitamin A deficiency and less than a level that increases the accumulation of A2E in at least one eye of the mammal.
  • the level of serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP that increases the accumulation of A2E in at least one eye of the mammal is at least about 60%, at least about 70%, or at least about 80% of the pre- therapy serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP level.
  • the desired percentage of serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP reduction is maintained for at least 1 week, for at least 1 month, for at least 6 months, for at least 1 year, for the lifetime of the mammal.
  • the serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP level in the mammal is measured at periodic levels to make sure that the serum retinol, serum retinol binding protein (RBP), and/or serum retinol-RBP level is maintained within a desired range.
  • compositions or methods for treating a lipofuscin-based retinal disease in a mammal comprising reducing the retinol level in at least one RPE of the mammal by a desired percentage.
  • the desired percentage of retinol reduction is relative to pre-therapeutic levels; in alternative embodiments, the desired percentage of retinol reduction is relative to a pre-determined threshold level.
  • the desired percentage of retinol reduction is at least about 60%, at least about 70%, or at least about 80%.
  • the desired percentage of retinol reduction is maintained for at least 1 week, for at least 1 month, for at least 6 months, for at least 1 year, for the lifetime of the mammal.
  • serum retinol refers to any one or all of serum retinol, serum RBP, and serum retinol-RBP.
  • kits for reducing the formation of lipofuscin in an eye of a mammal comprising modulating the serum retinol level in the mammal by administering to the mammal at least once an effective amount of invention compositions.
  • kits for reducing the formation of drusen in an eye of a mammal comprising modulating the serum retinol level in the mammal by administering to the mammal at least once an effective amount of invention compositions.
  • neovascularization in the eye of a mammal comprising modulating the serum retinol levels in the mammal by administering to the mammal at least once an effective amount of invention compositions.
  • kits for reducing the formation of abnormal blood vessel growth beneath the macula in an eye of a mammal comprising modulating the serum RBP level in the mammal by administering to the mammal at least once an effective amount of invention compositions.
  • kits for protecting an eye of a mammal from light comprising modulating the serum RBP level in the mammal by administering to the mammal at least once an effective amount of invention compositions.
  • any of the aforementioned aspects are further embodiments comprising multiple administrations of the effective amount of the compound, including further embodiments in which (i) the time between multiple administrations is at least one week; (ii) the time between multiple administrations is at least one day; and (iii) the compound is administered to the mammal on a daily basis; or (iv) the compound is administered to the mammal every 12 hours.
  • any of the aforementioned aspects are further embodiments comprising (a) monitoring serum RBP levels; (b) monitoring formation of drusen in the eye of the mammal; (c) measuring levels of lipofuscin in the eye of the mammal by auto fluorescence; (d) measuring visual acuity in the eye of the mammal; (e) conducting a visual field examination on the eye of the mammal, including embodiments in which the visual field examination is a Humphrey visual field exam and/or microperimetry; (f) measuring the auto fluorescence or absorption spectra of N-retinylidene-phosphatidylethanolamine, dihydro-N-retinylidene-N-retinyl- phosphatidylethanolamine, N-retinylidene-N-retinyl-phosphatidylethanolamine, dihydro-N- retinylidene-N-retinyl-ethanolamine, and/or N-retinylidene-
  • any of the aforementioned aspects are further embodiments comprising an additional treatment for retinal degeneration.
  • pharmaceutically acceptable carrier comprises corn oil and a non-ionic surfactant; (b) the pharmaceutically acceptable carrier further comprises flour, a sweetener, and a humectant; (c) the pharmaceutically acceptable carrier comprises lysophosphatidylcholine, monoglyceride and a fatty acid; (d) the pharmaceutically acceptable carrier comprises dimyristoyl
  • the pharmaceutically acceptable carrier comprises ethanol, alkoxylated caster oil, and a non- ionic surfactant;
  • pharmaceutically acceptable carrier comprises an extended release formulation; or (g) the pharmaceutically acceptable carrier comprises a rapid release formulation.
  • [0029] in another aspect is a polymorphic form of fenretinide characterized by a powder x- ray diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1.
  • fenretinide in another aspect is a polymorphic form of fenretinide characterized by: (i) a powder x-ray diffraction pattern wherein said x-ray diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1 and (ii) a predominant endotherm at about 164°C as measured by DSC.
  • a polymorphic form of fenretinide prepared by precipitation of ethanol water solution at suitable temperature.
  • a solid dosage form wherein the invention polymorphic form of fenretinide is present in at least about 50% to about 99.99% of the total weight of the fenretinide.
  • FIG. 1A-1C show particle size difference between lot No. 1 and lot No. 2 of fenretinide powders before and after formulation in a suspension comprising corn oil and polysorbate.
  • FIG 1 A shows the appearance of both lots after retrieval from the formulation.
  • FIG. IB illustrate the particle size distribution of both lots after formulation and
  • FIG. 1C illustrates the particle size distribution before formulation.
  • FIG. 2 illustrates XRPD diagrams of lot No. 1 of fenretinide powders before and after formulation (suspension in corn oil and polysorbate).
  • FIG. 3 illustrates DSC diagrams of lot No. 1 of fenretinide powders before (3B) and after (3A) formulation (suspension in corn oil and polysorbate).
  • FIGS. 4A-4D illustrate pharmacokinetic analysis of three lots of fenretinide in rat.
  • Concentrations of serum fenretinide ( ⁇ ) for lot No. 1, lot No. 2 and lot No. 3 (2.4 micron particle size) were recorded for 24 hours following a single oral dose of the indicated lot (10 mg/kg) (FIG. 4A and 4C).
  • concentrations of serum retinol ( ⁇ ) for lot No. 1, lot No. 2 and lot No. 3 were recorded (FIG. 4B and 4D).
  • FIG. 5 illustrates temporal analysis of serum RBP levels of patients with lot No. 1 (300 mg "Early EnroUers”) vs. patients with lot No. 2 of fenretinide (300 mg "Late EnroUers”) for 24 months.
  • FIG. 6A illustrates results of median GA lesion size growth over 24 months among all patients.
  • FIG. 6B illustrates results of median GA lesion size growth among patients who received only lot No 1 for the entire 24 months.
  • FIG. 7A-7F show changes in lesion size (mm 2 ) or as a percentage change from baseline vs. RBP reduction among patients in the placebo and the 300 mg cohorts ("Early EnroUers” received only lot No. 1, "Late EnroUers” received a mixture of lot No. 1 and lot No. 2) at 24 months.
  • the y-axis of FIG. 7A-7C is lesion size in mm 2 while the y-axis of 7D-7F is lesion size increase in percentage.
  • FIGS. 8A-8D show correlation between RBP reduction (% from baseline) and lesion size growth.
  • FIG. 8A and 8B show results from the placebo group and
  • FIG. 8C and 8D show results from the 300 mg cohorts.
  • FIG. 9 shows mean visual acuity by ETDRS letters lost between patients who took 300 mg lot No. 1 ("Early EnroUers”) of fenretinide and lot No. 2 (“Late EnroUers”).
  • FIG. 10 shows mean visual acuity by ETDRS letters read between patients who took 300mg lot No. 1 ("Early EnroUers") of fenretinide and lot No. 2 ("Late EnroUers”).
  • FIG. 11 shows results of fenretinide decreasing the emergence of choroidal neovascularization (CNV) by 50%. The incidence of CNV at 24 months in placebo patients was 22% (15/68), in patients receiving 100 mg fenretinide 13.5% (7/52), and in patients receiving 300 mg fenretinide 13.8% (8/58).
  • CNV choroidal neovascularization
  • Fenretinide can be used to provide benefit to patients suffering from or susceptible to various macular degenerations and dystrophies, including but not limited to dry- form age-related macular degeneration and Stargardt Disease. Specifically, fenretinide provide at least some of the following benefits to such human patients: reduction in the amount of all-trans-retinal (atRAL), reduction in the formation of A2E, reduction in the formation of lipofuscin, reduction in the formation of drusen, and reduction in light sensitivity. There is a reduced tendency to form A2E in ophthalmic and ocular tissues caused, in part, by a reduction in the over- accumulation of all-trans-retinal in these tissues.
  • atRAL all-trans-retinal
  • A2E itself is cytotoxic to the retinal pigment epithelium (RPE) (which can lead to retina cell death)
  • administration of the invention fenretinide powder reduces the rate of accumulation of A2E, a cytotoxic agent, thus providing patient benefit.
  • A2E is the major fluorophore of lipofuscin
  • reduced quantities of A2E in ophthalmic and ocular tissues also results in a reduced tendency to accumulate lipofuscin in such tissues.
  • the methods and compositions described herein can be considered to be lipofuscin-based treatments because administration of the invention fenretinide powder (alone, or in combination with other agents, as described herein) reduces, lowers or otherwise impacts the accumulation of lipofuscin in ophthalmic and/or ocular tissues.
  • a reduction in the rate of accumulation of lipofuscin in ophthalmic and/or ocular tissues benefits patients that have diseases or conditions such as macular degenerations and/or dystrophies.
  • fenretinide can also be used as a preventative therapy for this latter ophthalmic condition.
  • fenretinide may provide further therapeutic effect for wet-form age-related macular degeneration because such compounds additionally have anti-angiogenic activity.
  • Macular or Retinal Degenerations and Dystrophies are a disease of the eye that involves deterioration of the macula, the central portion of the retina. Approximately 85% to 90% of the cases of macular degeneration are the "dry" (atrophic or non-neovascular) type. In dry macular degeneration, the deterioration of the retina is associated with the formation of small yellow deposits, known as drusen, under the macula; in addition, the accumulation of lipofuscin in the RPE leads to photoreceptor degeneration and geographic atrophy. This phenomena leads to a thinning and drying out of the macula.
  • the location and amount of thinning in the retina caused by the drusen directly correlates to the amount of central vision loss.
  • Degeneration of the pigmented layer of the retina and photoreceptors overlying drusen become atrophic and can cause a slow loss of central vision.
  • loss of retinal pigment epithelium and underlying photoreceptor cells results in geographic atrophy.
  • Administration of fenretinide to a mammal can reduce the formation of, or limit the spread of, photoreceptor degeneration and/or geographic atrophy in the eye of the mammal.
  • HPR hydro xypinaco lone retinoate
  • MPR N-(4-methoxyphenyl) retinamide
  • wet macular degeneration new blood vessels form (i.e., neovascularization) to improve the blood supply to retinal tissue, specifically beneath the macula, a portion of the retina that is responsible for our sharp central vision.
  • the new vessels are easily damaged and sometimes rupture, causing bleeding and injury to the surrounding tissue.
  • wet macular degeneration only occurs in about 10 percent of all macular degeneration cases, it accounts for approximately 90% of macular degeneration-related blindness.
  • Neovascularization can lead to rapid loss of vision and eventual scarring of the retinal tissues and bleeding in the eye. This scar tissue and blood produces a dark, distorted area in the vision, often rendering the eye legally blind.
  • Wet macular degeneration usually starts with distortion in the central field of vision. Straight lines become wavy.
  • VEGF vascular endothelial growth factor
  • Administration of fenretinide and the like to a mammal can reduce the formation of, or limit the spread of, wet-form age-related macular degeneration in the eye of the mammal.
  • administration of HPR and/or MPR to a mammal can be used to treat wet-form age-related macular degeneration in the eye of the mammal.
  • fenretinide and the like can be used to treat choroidal neovascularization and the formation of abnormal blood vessels beneath the macula of the eye of a mammal.
  • Such therapeutic effect can result from a number of effects: lowering of serum retinol and thus ocular retinol levels; anti-angiogenic activity, and/or the quelling of geographic atrophy.
  • Geographic atrophy Geographic atrophy the "dry” form of advanced AMD, results from atrophy to the retinal pigment epithelial layer below the retina, which causes vision loss through loss of photoreceptors (rods and cones) in the central part of the eye. Atrophy refers to the degeneration of the deepest cells, the retinal pigment epithelium (RPE), of the retina.
  • RPE retinal pigment epithelium
  • kits for reducing the serum RBP level by at least about 60% in a human subject or reducing the rate of geographic atrophy lesion size increase in an eye of the human subject comprising administering the invention
  • the effective amount of the pharmaceutical composition is administered orally to the human.
  • the methods comprise multiple administrations of the effective amount of the pharmaceutical composition.
  • the methods reduce visual acuity after administration of the composition for 18 months.
  • the methods reduce at least about 40% the rate of geographic atrophy lesion size increase after administration of the composition for 12 months.
  • the methods reduce about 60% to about 80% the rate of geographic atrophy lesion size increase.
  • fenretinide powder refers to the fenretinide solid in a particulate form, in which the diameter of at least 80% of the particles in the solid is about or less than 10 micron. In some embodiments, the diameter is less than 9 micron, less than 8 micron, less than 7 micron, less than 6 micron, less than 5 micron, less than 4 micron, or less than 2 micron. In some embodiments, the diameter of at least 80% of the particles in the solid is about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 4 micron, about 2 micron, or about 1 micron.
  • the fenretinide powder may be prepared by precipitation, by crystallization or by micronization. Micronization is the reduction in particle size. In some embodiments, the invention fenretinide powder has been produced from a suspension
  • Stargardt Disease is a macular dystrophy that manifests as a recessive form of macular degeneration with an onset during childhood. See e.g., Allikmets et al., Science, 277: 1805-07 (1997); Lewis et al, Am. J. Hum. Genet., 64:422-34 (1999); Stone et al, Nature Genetics, 20:328-29 (1998); Allikmets, Am. J. Hum. Gen., 67:793-799 (2000); Klevering, et al, Ophthalmology, 111 :546-553 (2004). Stargardt Disease is characterized clinically by
  • macular degenerations that affect children, teenagers or adults that are commonly known as early onset or juvenile macular degeneration. Many of these types are hereditary and are looked upon as macular dystrophies instead of degeneration. Some examples of macular dystrophies include: Cone-Rod Dystrophy, Corneal Dystrophy, Fuch's Dystrophy, Sorsby's Macular Dystrophy, Best Disease, and Juvenile
  • N-oxides include the use of N-oxides, crystalline forms (also known as polymorphs), or pharmaceutically acceptable salts of the fenretinide powder, as well as active metabolites of these compounds having the same type of activity.
  • a known metabolite of fenretinide is N-(4-methoxyphenyl)retinamide, also known as 4-MPR or MPR.
  • Another known metabolite of fenretinide is 4-oxo fenretinide.
  • compounds may exist as tautomers. All tautomers are included within the scope of the compounds presented herein.
  • the compounds described herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In some instances, the compounds described herein can exist in solvated forms with solvents such as pyridine, MEK, and the like. The solvated forms of the compounds presented herein are also considered to be disclosed herein.
  • compositions comprising (a) fenretinide powder in an amount therapeutically effective to reduce the serum RBP level by at least about 60% in a human subject or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject, and (b) one or more pharmaceutically acceptable excipients, wherein the diameter of at least 80% of the particles in the fenretinide powder is about or less than 10 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder is less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron.
  • the diameter of at least 80% of the particles in the fenretinide powder is about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron. In certain embodiments, the fenretinide powder is substantially free of particles having the diameter of 10 micron or more. In some embodiments, the diameter of at least 80% of the particles in the fenretinide powder in the pharmaceutical composition is less than about 10 micron, less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron.
  • the diameter of at least 80% of the particles in the fenretinide powder in the pharmaceutical composition is about 10 micron, about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron.
  • the fenretinide powder in the pharmaceutical composition is substantially free of particles having the diameter of 10 micron or more.
  • the fenretinide powder of the composition is characterized by a predominant endotherm at about 164°C as measured by DSC and a powder x-ray diffraction pattern wherein said x-ray diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1.
  • compositions comprising (a) fenretinide powder that has been produced from a suspension comprising corn oil and/or polysorbate in an amount therapeutically effective to reduce the serum RBP level by at least about 60% in a human subject or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject, and (b) one or more pharmaceutically acceptable excipients.
  • the diameter of at least 80% of the particles in the fenretinide powder is about or less than 10 micron.
  • the diameter of at least 80% of the particles in the fenretinide powder is less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder is about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron. In certain embodiments, the fenretinide powder is substantially free of particles having the diameter of 10 micron or more.
  • the diameter of at least 80% of the particles in the fenretinide powder in the pharmaceutical composition is less than about 10 micron, less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder in the pharmaceutical composition is about 10 micron, about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron. In certain embodiments, the fenretinide powder in the pharmaceutical composition is substantially free of particles having the diameter of 10 micron or more.
  • the fenretinide powder of the composition is characterized by a predominant endotherm at about 164°C as measured by DSC and a powder x-ray diffraction pattern wherein said x-ray diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1.
  • Polysorbates are oily liquids derived from PEG-ylated sorbitan (a derivative of sorbitol) esterified with fatty acids.
  • Exemplary polysorbates include, but not limited to, Polysorbate 20 (Tween 20 or polyoxy ethylene (20) sorbitan monolaurate), Polysorbate 40 (Tween 40 or polyoxy ethylene (20) sorbitan monopalmitate), Polysorbate 60 (Tween 60 or polyoxy ethylene (20) sorbitan monostearate), Polysorbate 80 (Tween 80 or polyoxy ethylene (20) sorbitan monooleate).
  • the number 20 following the polyoxyethylene part refers to the total number of oxyethylene -(CH 2 CH 2 O)- groups found in the molecule.
  • the number following the polysorbate part is related to the type of fatty acid associated with the polyoxyethylene sorbitan part of the molecule. Monolaurate is indicated by 20, monopalmitate is indicated by 40, monostearate by 60 and monooleate by 80.
  • the invention fenretinide powder is produced from any portion of PEG-ylated sorbitan esterified with fatty acids.
  • the invention fenretinide powder is produced from a composition comprising
  • Polysorbate 20 Polysorbate 40, Polysorbate 60, Polysorbate 80, or the like and/or combinations thereof.
  • Refined corn oil is 99%> triglyceride, with proportions of approximately 55%> polyunsaturated fatty acid, 30% monounsaturated fatty acid, and 15% saturated fatty acid.
  • saturated fatty acids 80% are palmitic acid (lipid number of C16:0), 14% stearic acid (C18:0), and 3% arachidic acid (C20:0).
  • oleic acid CI 8: 1 c).
  • the invention fenretinide powder is produced from any composition of corn oil or component thereof.
  • pharmaceutical compositions comprising (a) a non-micronized fenretinide powder in an amount therapeutically effective to reduce the serum RBP level by at least about 60% in a human subject or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject, and (b) one or more
  • the diameter of at least 80% of the particles in the fenretinide powder is about or less than 10 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder is less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder is about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron.
  • the fenretinide powder is substantially free of particles having the diameter of 10 micron or more.
  • the diameter of at least 80% of the particles in the fenretinide powder in the pharmaceutical composition is less than about 10 micron, less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron.
  • the diameter of at least 80% of the particles in the fenretinide powder in the pharmaceutical composition is about 10 micron, about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron.
  • the fenretinide powder in the pharmaceutical composition is substantially free of particles having the diameter of 10 micron or more.
  • the fenretinide powder of the composition is characterized by a predominant endotherm at about 164°C as measured by DSC and a powder x-ray diffraction pattern wherein said x-ray diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1.
  • the particle gains charges due to constant grinding and/or interacting between particles.
  • the charge is magnified in a very non- polar environment such as corn oil and/or polysorbate.
  • the particle aggregates and agglomerates into spheres and creates a larger particle.
  • the process of micronization provides a build-up of charges (e.g. ionizes) on the surface of the fenretinide powder, as such, in some embodiments, such surface charges promote agglomeration in nonpolar liquids, such as corn oil.
  • compositions comprising (a) fenretinide powder, characterized by an X-ray powder diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1, in an amount therapeutically effective to reduce the serum RBP level by at least about 60% in a human subject or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject, and (b) one or more
  • the diameter of at least 80% of the particles in the fenretinide powder is about or less than 10 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder is less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder is about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron.
  • the diameter of at least 80% of the particles in the fenretinide powder in the pharmaceutical composition is less than about 10 micron, less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder in the pharmaceutical composition is about 10 micron, about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron. In certain embodiments, the fenretinide powder in the pharmaceutical composition is substantially free of particles having the diameter of 10 micron or more. In certain embodiments, the fenretinide powder of the composition is characterized by a predominant endotherm at about 164°C as measured by DSC.
  • compositions comprising (a) fenretinide powder, characterized by a DSC phase transition at a temperature of about 164°C, in an amount therapeutically effective to reduce the serum RBP level by at least about 60% in a human subject or reduce the rate of geographic atrophy lesion size increase in an eye of the human subject, and (b) one or more pharmaceutically acceptable excipients.
  • the diameter of at least 80% of the particles in the fenretinide powder is about or less than 10 micron.
  • the diameter of at least 80% of the particles in the fenretinide powder is less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron. In certain embodiments, the diameter of at least 90% of the particles in the fenretinide powder is about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron.
  • the diameter of at least 80% of the particles in the fenretinide powder in the pharmaceutical composition is less than about 10 micron, less than about 9 micron, less than about 8 micron, less than about 7 micron, less than about 6 micron, less than about 5 micron, less than about 4 micron, less than about 2 micron. In certain embodiments, the diameter of at least 80% of the particles in the fenretinide powder in the pharmaceutical composition is about 10 micron, about 9 micron, about 8 micron, about 7 micron, about 6 micron, about 5 micron, about 4 micron, about 2 micron, or about 1 micron. In certain embodiments, the fenretinide powder in the pharmaceutical composition is substantially free of particles having the diameter of 10 micron or more.
  • the fenretinide powder of the composition is characterized by a powder x- ray diffraction pattern wherein said x-ray diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1.
  • the one or more pharmaceutically acceptable excipients of the invention compositions are selected from the group consisting of corn oil, polysorbate, micro crystalline cellulose, starch, hydroxypropylcellulose, sodium starch glycolate, isopropyl alcohol, magnesium stearate, and combinations thereof.
  • the one or more pharmaceutically acceptable excipients comprise corn oil.
  • solid dosage forms of the invention fenretinide powder, wherein the polymorphic form of fenretinide is present in at least about 50% to about 99.99% of the total weight of fenretinide.
  • dilute refers to chemical compounds that are used to dilute the compound of interest prior to delivery. Diluents can also be used to stabilize compounds because they can provide a more stable environment. Salts dissolved in buffered solutions (which also can provide pH control or maintenance) are utilized as diluents in the art, including, but not limited to a phosphate buffered saline solution.
  • physiologically acceptable refers to a material, such as a carrier or diluent, that does not abrogate the biological activity or properties of the compound, and is nontoxic.
  • a "metabolite” of a compound disclosed herein is a derivative of that compound that is formed when the compound is metabolized.
  • active metabolite refers to a biologically active derivative of a compound that is formed when the compound is metabolized.
  • metabolized refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism. Thus, enzymes may produce specific structural alterations to a compound.
  • cytochrome P450 catalyzes a variety of oxidative and reductive reactions while uridine diphosphate glucuronyltransferases catalyze the transfer of an activated glucuronic-acid molecule to aromatic alcohols, aliphatic alcohols, carboxylic acids, amines and free sulphydryl groups. Further information on metabolism may be obtained from The
  • Metabolites of the compounds disclosed herein can be identified either by administration of compounds to a host and analysis of tissue samples from the host, or by incubation of compounds with hepatic cells in vitro and analysis of the resulting compounds. Both methods are well known in the art.
  • MPR is a known metabolite of HPR. MPR accumulates systemically in patients that have been chronically treated with HPR. One of the reasons that MPR accumulates systemically is that MPR is only (if at all) slowly metabolized, whereas HPR is metabolized to MPR. In addition, MPR may undergo relatively slow clearance. Thus, (a) the pharmacokinetics and pharmacodynamics of MPR must be taken into consideration when administering and determining the bioavailability of HPR, (b) MPR is more stable to
  • MPR can be more immediately bio available than HPR following absorption.
  • Another known metabolite of fenretinide is 4-oxo fenretinide.
  • MPR may also be considered an active metabolite.
  • MPR (like HPR) can bind to Retinol Binding Protein (RBP) and prevent the binding of RBP to transthyretin (TTR).
  • RBP Retinol Binding Protein
  • TTR transthyretin
  • MPR can (a) serve as an inhibitor of retinol binding to RBP, (b) serve as an inhibitor of RBP to TTR, (c) limit the transport of retinol to certain tissues, including ophthalmic tissues, and (d) be transported by RBP to certain tissues, including ophthalmic tissues.
  • MPR appears to bind more weakly to RBP than HPR, and is thus a less strong inhibitor of retinol binding to RBP. Nevertheless, both MPR and HPR are expected to inhibit, approximately equivalently, the binding of RBP to TTR. MPR has, in these respects, the same mode of action as HPR and can serve as a therapeutic agent in the methods and compositions described herein.
  • the invention fenretinide powder described herein can be administered to a human patient per se, or in pharmaceutical compositions where they are mixed with other active ingredients, as in combination therapy, or suitable carrier(s) or excipient(s).
  • suitable carrier(s) or excipient(s) include butyl alcohol
  • invention compositions are administered orally.
  • invention compositions are in a solid dosage form.
  • the solid dosage form is a tablet or a capsule.
  • invention compositions are in suspension form.
  • the suspension comprises corn oil or a component thereof and a non-ionic surfactant.
  • invention compositions may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • invention compositions may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Pharmaceutical Sciences, above.
  • compositions can be administered in a variety of ways, including systemically, such as orally.
  • compositions can illustratively take the form of a liquid where the agents are present in solution, in suspension or both. Typically when the composition is administered as a solution or suspension a first portion of the agent is present in solution and a second portion of the agent is present in particulate form, in suspension in a liquid matrix.
  • a liquid composition may include a gel formulation. In other embodiments, the liquid composition is aqueous.
  • the composition can take the form of an ointment.
  • Useful aqueous suspension can also contain one or more polymers as suspending agents.
  • Useful polymers include water-soluble polymers such as cellulosic polymers, e.g., hydroxypropyl methylcellulose, and water-insoluble polymers such as cross-linked carboxyl- containing polymers.
  • Useful compositions can also comprise an acceptable mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer), poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran.
  • compositions may also include solubilizing agents to aid in the solubility of invention fenretinide power.
  • solubilizing agent generally includes agents that result in formation of a micellar solution or a true solution of the agent.
  • Certain acceptable nonionic surfactants for example polysorbate 80, can be useful as solubilizing agents, as can acceptable glycols, polyglycols, e.g., polyethylene glycol 400, and glycol ethers.
  • Useful compositions may also include one or more pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris-hydroxymethylaminomethane; and buffers such as
  • citrate/dextrose sodium bicarbonate and ammonium chloride.
  • Such acids, bases and buffers are included in an amount required to maintain pH of the composition in an acceptable range.
  • Useful compositions may also include one or more acceptable salts in an amount required to bring osmolality of the composition into an acceptable range.
  • acceptable salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
  • compositions may also include one or more acceptable preservatives to inhibit microbial activity.
  • Suitable preservatives include mercury-containing substances such as merfen and thiomersal; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride, cetyltrimethylammonium bromide and cetylpyridinium chloride.
  • compositions may include one or more acceptable surfactants to enhance physical stability or for other purposes.
  • Suitable nonionic surfactants include polyoxy ethylene fatty acid glycerides and vegetable oils, e.g., polyoxy ethylene (60)
  • compositions may include one or more antioxidants to enhance chemical stability where required.
  • Suitable antioxidants include, by way of example only, ascorbic acid and sodium metabisulfite.
  • Aqueous suspension compositions can be packaged in single-dose non-reclosable containers.
  • multiple-dose reclosable containers can be used, in which case it is typical to include a preservative in the composition.
  • compositions are, by way of example only, positively, negatively or neutrally charged phospholipids, or bile salt/phosphatidylcholine mixed lipid aggregate systems, such as those described in Li, C.Y., et al., Pharm. Res. 13:907-913 (1996).
  • An additional formulation that can be used for the same purpose with the invention compositions involves use of a solvent comprising an alcohol, such as ethanol, in combination with an alkoxylated caster oil. See, e.g., U.S. Patent Publication Number 2002/0183394.
  • a formulation comprising the invention compositions is an emulsion composed of a lipoid dispersed in an aqueous phase, a stabilizing amount of a non-ionic surfactant, optionally a solvent, and optionally an isotonic agent.
  • a formulation comprising the invention compositions includes corn oil and a non-ionic surfactant. See U.S. Patent No. 4,665,098.
  • Still another formulation comprising the invention compositions includes lysophosphatidylcholine, monoglyceride and a fatty acid. See U.S. Patent No. 4,874,795.
  • Still another formulation comprising the invention compositions includes flour, a sweetener, and a humectant. See International Publication No. WO
  • compositions includes dimyristoyl phosphatidylcholine, soybean oil, t-butyl alcohol and water. See U.S. Patent Application Publication No. US 2002/0143062.
  • compositions can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers or excipients well known in the art.
  • Such carriers enable the compounds described herein to be formulated as tablets, powders, pills, dragees, capsules, liquids, gels, syrups, elixirs, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained by mixing one or more solid excipient with one or more of the compounds described herein, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as: for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose, micro crystalline cellulose,
  • hydroxypropylmethylcellulose sodium carboxymethylcellulose; or others such as:
  • polyvinylpyrrolidone PVP or povidone
  • calcium phosphate a polyvinylpyrrolidone
  • disintegrating agents such as the cross-linked croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc,
  • polyvinylpyrrolidone carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different
  • compositions which can be used orally include push- fit capsules made of gelatin, including by way of example only, soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol; or hard-gel capsules or tablets.
  • the push- fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • a pharmaceutical carrier for the invention compositions is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant (e.g. a polysorbate), a water- miscible organic polymer, and an aqueous phase.
  • the cosolvent system may be a 10% ethanol, 10%) polyethylene glycol 300, 10%> polyethylene glycol 40 castor oil (PEG-40 castor oil) with 70%o aqueous solution.
  • PEG-40 castor oil polyethylene glycol 40 castor oil
  • This cosolvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration. Naturally, the proportions of a cosolvent system may be varied considerably without destroying its solubility and toxicity characteristics.
  • cosolvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of PEG-40 castor oil, the fraction size of polyethylene glycol 300 may be varied; other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides maybe included in the aqueous solution.
  • hydrophobic pharmaceutical compounds may be employed.
  • Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs.
  • Certain organic solvents such as N-methylpyrrolidone also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein stabilization may be employed.
  • compositions have been used with fenretinide powder in accordance with the present invention in the treatment of
  • Lym-X-SorbTM This formulation, which comprises an organized lipid matrix that includes lysophosphatidylcholine, monoglyceride and fatty acid, is designed to improve the oral availability of fenretinide.
  • a formulation i.e., an oral formulation that includes lysophosphatidylcholine, monoglyceride and fatty acid, is proposed to also provide improved bioavailability of the invention compositions for the treatment of ophthalmic and ocular diseases and conditions, including but not limited to the macular degenerations and dystrophies.
  • This formulation can be used in a range of orally- administered compositions, including by way of example only, a capsule and a powder that can be suspended in water to form a drinkable composition.
  • All of the formulations described herein may benefit from antioxidants, metal chelating agents, thiol containing compounds and other general stabilizing agents.
  • stabilizing agents include, but are not limited to: (a) about 0.5% to about 2% w/v glycerol, (b) about 0.1%) to about 1% w/v methionine, (c) about 0.1%> to about 2% w/v monothioglycerol, (d) about 1 mM to about 10 mM EDTA, (e) about 0.01% to about 2% w/v ascorbic acid, (f) 0.003% to about 0.02% w/v polysorbate 80, (g) 0.001% to about 0.05% w/v.
  • polysorbate 20 (h) arginine, (i) heparin, (j) dextran sulfate, (k) cyclodextrins, (1) pentosan polysulfate and other heparinoids, (m) divalent cations such as magnesium and zinc; or (n) combinations thereof.
  • subject or "mammal” means all mammals including humans. Mammals include, by way of example only, humans, non-human primates, cows, dogs, cats, goats, sheep, pigs, rats, mice and rabbits.
  • an effective amount refers to that amount of the compound being administered which will relieve to some extent one or more of the symptoms of the disease, condition or disorder being treated.
  • compositions containing the compound(s) described herein can be administered for prophylactic and/or therapeutic treatments.
  • treating is used to refer to either prophylactic and/or therapeutic treatments. In therapeutic applications, the
  • compositions are administered to a patient already suffering from a disease, condition or disorder, in an amount sufficient to cure or at least partially arrest the symptoms of the disease, disorder or condition. Amounts effective for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician. It is considered well within the skill of the art for one to determine such therapeutically effective amounts by routine experimentation (e.g., a dose escalation clinical trial).
  • compositions containing the compounds described herein are administered to a patient susceptible to or otherwise at risk of a particular disease, disorder or condition. Such an amount is defined to be a "prophylactically effective amount or dose.” In this use, the precise amounts also depend on the patient's state of health, weight, and the like. It is considered well within the skill of the art for one to determine such prophylactically effective amounts by routine experimentation (e.g., a dose escalation clinical trial).
  • the terms “enhance” or “enhancing” means to increase or prolong either in potency or duration a desired effect.
  • the term “enhancing” refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system.
  • An “enhancing-effective amount,” as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system. When used in a patient, amounts effective for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician.
  • the administration of the compounds may be administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease or condition.
  • the administration of the compounds may be given continuously or temporarily suspended for a certain length of time (i.e., a "drug holiday").
  • a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, can be reduced, as a function of the symptoms, to a level at which the improved disease, disorder or condition is retained. Patients can, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms.
  • the amount of a given agent that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the subject or host in need of treatment, but can nevertheless be routinely determined in a manner known in the art according to the particular circumstances surrounding the case, including, e.g., the specific agent being administered, the route of administration, the condition being treated, and the subject or host being treated.
  • doses employed for adult human treatment will typically be in the range of 0.02-5000 mg per day, preferably 1-1500 mg per day.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered simultaneously (or over a short period of time) or at appropriate intervals, for example as two, three, four or more sub-doses per day.
  • the pharmaceutical compositions provided herein are used for reducing serum RBP levels by at least about 60% in a human subject.
  • serum RBP levels are reduced by at least 30%, at least 40%, at least 50%>, at least 60%>, at least 70%), or at least 80%>.
  • serum RBP levels are reduced by about 30%>, about 40%, about 50%, about 60%, about 70%, or about 80%.
  • the pharmaceutical compositions provided herein are used for reducing the rate of geographic atrophy lesion size increase in an eye by at least about 60% in a human subject.
  • the rate of geography atrophy lesion size increase in an eye is reduced by at least 30%>, at least 40%>, at least 50%>, at least 60%>, at least 70%>, or at least 80%.
  • the rate of geography atrophy lesion size increase in an eye is reduced by about 30%, about 40%, about 50%, about 60%>, about 70%, or about 80%.
  • the pharmaceutical compositions provided herein are used for reducing the serum RBP level of a human by about 2 mg/dL to about 2.5 mg/dL.
  • the serum RBP levels of a human are reduced by about 1.0 mg/dL, about 1.2 mg/dL, about 1.4 mg/dL, about 1.5 mg/dL, about 1.6 mg/dL, about 1.8 mg/dL, about 2.0 mg/dL, about 2.2 mg/dL, about 2.4 mg/dL, about 2.5 mg/dL, about 2.6 mg/dL, about 2.8 mg/dL, or about 3.0 mg/dL.
  • compositions provided herein are used for treating Stargardt Disease.
  • the therapeutically effective amount of fenretinide powder in the invention compositions is at least about 100 mg. In other embodiments, the therapeutically effective amount of fenretinide powder is at least about 300 mg.
  • the invention fenretinide powder (or a pharmaceutically acceptable salt, ester, amide, or solvate) in combination with another therapeutic agent.
  • another therapeutic agent such as fenretinide powder (or a pharmaceutically acceptable salt, ester, amide, or solvate) in combination with another therapeutic agent.
  • the benefit of experienced by a patient may be increased by administering one of the compounds described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
  • another therapeutic agent which also includes a therapeutic regimen
  • increased therapeutic benefit may result by also providing the patient with other therapeutic agents or therapies for macular degeneration.
  • the overall benefit experienced by the patient may simply be additive of the two therapeutic agents or the patient may experience a synergistic benefit.
  • the invention fenretinide powder or other agents that result in the reduction of serum retinol levels can be administered with (meaning before, during or after) agents that treat or alleviate side effects arising from serum retinol reduction.
  • Such side effects include dry skin and dry eye.
  • agents that alleviate or treat either dry skin or dry eye may be administered with the invention fenretinide powder or other agents that reduce serum retinol levels.
  • the compound and compositions disclosed herein can also be used in assays for detecting perturbations in RBP or TTR availability through conventional means.
  • a subject may be treated with invention fenretinide powder or compositions disclosed herein, and RBP or TTR levels quantified using conventional assay techniques. See Sundaram, M., et al, Biochem. J. 362:265-271 (2002).
  • a typical non-competitive sandwich assay is an assay disclosed in U.S. Pat. No. 4,486,530, incorporated herein by reference.
  • a sandwich complex for example an immune complex, is formed in an assay medium.
  • the complex comprises the analyte, a first antibody, or binding member, that binds to the analyte and a second antibody, or binding member that binds to the analyte or a complex of the analyte and the first antibody, or binding member.
  • the sandwich complex is detected and is related to the presence and/or amount of analyte in the sample.
  • the sandwich complex is detected by virtue of the presence in the complex of a label wherein either or both the first antibody and the second antibody, or binding members, contain labels or substituents capable of combining with labels.
  • the sample may be plasma, blood, feces, tissue, mucus, tears, saliva, or urine, for example for detecting modulation of clearance rates for RBP or TTR.
  • the sample in a suitable medium is contacted with labeled antibody or binding member for the analyte and incubated for a period of time. Then, the medium is contacted with a support to which is bound a second antibody, or binding member, for the analyte. After an incubation period, the support is separated from the medium and washed to remove unbound reagents. The support or the medium is examined for the presence of the label, which is related to the presence or amount of analyte.
  • the modulators disclosed herein may also be used in in vitro assays for detecting perturbations in RBP or TTR activity.
  • the modulator may be added to a sample comprising RBP, TTR and retinol to detect complex disruption.
  • a component for example, RBP, TTR, retinol or the modulator, may be labeled to determine if disruption of complex formation occurs.
  • Complex formation and subsequent disruption may be detected and/or measured through conventional means, such as the sandwich assays disclosed above.
  • Other detection systems may also be used to detect modulation of RBP or TTR binding, for example, FRET detection of RBP-TTR-retinol complex formation. See U.S. Provisional Patent
  • hybridization patterns can be compared to determine differential gene expression.
  • hybridization patterns from samples treated with the modulators may be compared to hybridization patterns from samples which have not been treated or which have been treated with a different compound or with different amounts of the same compound.
  • the samples may be analyzed using DNA array technology, see U.S. Patent No. 6,040,138, herein incorporated by reference in its entirety.
  • RBP or TTR activity may also be analyzed using recombinant DNA technology by analyzing the expression of reporter proteins driven by RBP or TTR promoter regions in an in vitro assay. See, e.g., Rapley and Walker, Molecular Biomethods Handbook (1998); Wilson and Walker, Principals and Techniques of Practical Biochemistry (2000), hereby incorporated by reference in its entirety.
  • In vitro translation assays may also be used to detect modulation or translation of RBP or TTR by the modulators disclosed herein.
  • modulation of translation by the modulators may be detected through the use of cell-free protein translation systems, such as E. coli extract, rabbit reticulocyte lysate and wheat germ extract, see Spirin, A.S., Cell-free protein synthesis bioreactor (1991), herein incorporated by reference in its entirety, by comparing translation of proteins in the presence and absence of the modulators disclosed herein.
  • Modulator effects on protein translation may also be monitored using protein gel electrophoretic or immune complex analysis to determine qualitative and quantitative differences after addition of the modulators.
  • labeled TTR or RBP may be injected into a subject, wherein a candidate modulator added before, during or after the injection of the labeled TTR or RBP.
  • the subject may be a mammal, for example a human; however other mammals, such as primates, horse, dog, sheep, goat, rabbit, mice or rats may also be used.
  • a biological sample is then removed from the subject and the label detected to determine TTR or RBP availability.
  • a biological sample may comprise, but is not limited to, plasma, blood, urine, feces, mucus, tissue, tears or saliva. Detection of the labeled reagents disclosed herein may take place using any of the conventional means known to those of ordinary skill in the art, depending upon the nature of the label. Examples of monitoring devices for
  • the present invention also provide a serum RBP monitoring kit for determining a real time and accurate dosage for a patient who takes the invention fenretinide powder in a suitable formulation.
  • the kit determines the therapeutically effective amount of fenretinide to reduce the serum RBP level by at least about 60% or reduce the rate of geographic atrophy lesion size increase in an eye comprise a RBP assay unit.
  • the RBP assay unit may be any assay units known in the art.
  • the RBP assay unit in some instances is an electronic basis assay unit that comprises reagents useful to detect serum RBP levels. PREPARATION OF THE FENRETINIDE POWDER
  • fenretinide is synthesized using standard synthetic techniques known to those of skill in the art or using methods known in the art in combination with methods described herein. See, e.g., U.S. Patent Application Publication 2004/0102650; Um, S. J., et al., Chem. Pharm. Bull., 52:501-506 (2004).
  • fenretinide is optionally purchased from various commercial suppliers.
  • the invention fenretinide powder is produced from a suspension comprising corn oil and/or polysorbate at suitable conditions, or by milled down (e.g., micronized) fenretinide solid (said solid can be produced by any means, including precipitation and crystallization) to a desired size that is about or less than 10 micron (e.g., 6 micron).
  • the invention fenretinide powder is prepared by precipitation, e.g., from ethanol water solution, in which the solubility of the fenretinide powder is limited.
  • the diameter of at least 80% of the particles in the fenretinide powder is about or less than 10 micron (e.g. about 6 micron).
  • the invention fenretinide powder is prepared by particle size reduction (e.g. milling or micronization) of a fenretinide solid (prepared by any known methods including, but not limited to, a synthetic method before crystallization procedure; a
  • crystallization process from any solvent systems such as isopropanol water, ethanol water or other solvent systems; a precipitation procedure from any solvent systems that produce a supernatant comprising fenretinide) where the micronization reduces the diameter of most of (e.g. at least 50% , at least 60% or at least 70% or at least 80%) the particles in the fenretinide solid to about or less than 10 micron, or to about or less than 9 micron, or to about or less than 8 micron, or to about or less than 7 micron, or to about or less than 6 micron, or to about or less than 5 micron, or to about or less than 4 micron, or to about or less than 2 micron, or to about or less than 1 micron.
  • An ordinary skill in the art would readily recognize other suitable methods to reduce the particle size of fenretinide solid to the desired diameter in accordance with the practice of the present invention.
  • the present invention provides a polymorphic form of fenretinide characterized by a powder x-ray diffraction pattern wherein said x-ray diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1.
  • the present invention provides a polymorphic form of fenretinide characterized by a predominant endotherm at 164°C as measured by DSC.
  • the present invention provides a polymorphic form of fenretinide characterized by: (i) a powder x-ray diffraction pattern wherein said x-ray diffraction pattern having 2 theta values at 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1 and (ii) a predominant endotherm at 164°C as measured by DSC.
  • the invention fenretinide powder is further formulated in accordance with the practice of the present invention.
  • the fenretinide powder is formulated to be a solid dosage form or a suspension comprising corn oil and a surfactant.
  • a solid dosage form of the invention fenretinide polymorphic form, wherein the polymorphic form of fenretinide is present in at least about 50% to about 99.99% of the total weight.
  • Example 1 Preparation of fenretinide powder lot No. 1 in capsule
  • Fenretinide was prepared in accordance with the known methods. The solid was then re-dissolved in ethanol-water solution either at room temperature or at other suitable temperature to produce a precipitate. The precipitate was filtered and dried under high vacuum to afford fenretinide powder. The powder was formulated in corn oil and polysorbate in capsule for clinical trial.
  • the diameter of the fenretinide powder is about 5.4 micron (before formulation, FIG 1C).
  • the powder was formulated in a suspension comprising corn oil and polysorbate in a capsule.
  • the diameter of the fenretinide powder after retrieved from the suspension was about 5.5 micron (FIG. IB and 1 A).
  • the after retrieved powder was tested by x- ray diffraction and differential scanning calorimetry (DSC) (See FIGS 2 and 3A).
  • the x-ray diffraction of the solid shows peaks at angles 2 theta of 7.0 ⁇ 0.1, 17.2 ⁇ 0.1, and 20.2 ⁇ 0.1.
  • the DSC plot shows a predominant endothermic peak at about 164°C. Lot No. 1 in capsule was used for subjects ("Early Enroller") in the clinical trial as in Example 4.
  • Example 2 Preparation of fenretinide powder lot No. 2 and lot No. 3.
  • Fenretinide was prepared in accordance with the known methods (e.g. crystallization from isopropyl alcohol water). The solid was further milled down to about 7.2 micron (D50) as lot No. 2 and about 2.4 micron (D50) as lot No. 3.
  • Example 3 Preparation of fenretinide lot No. 2 in capsule
  • Lot No. 2 was further formulated to a suspension comprising corn oil and
  • FIG 1 A and IB show the diameter of the particle in lot No. 2 retrieved from capsule formation is about 12 micron (D50) due to agglomeration.
  • the distribution diagram of particle size in FIG IB shows that more particles in lot No. 2 have more than 10 micron in diameter (about 50%).
  • particle size distribution of lot No. 1 retrieved from capsule formulation has similar distribution as before.
  • Example 4 Standard preparation of suspension versus pre-mixing
  • fenretinide particles agglomerate after 4-6 days in suspension, increasing the particle size of the fenretinide in the pharmaceutical composition.
  • Lot No. 3 particle size of 2.4 micron
  • FIG. 4C and 4D See FIG. 4C and 4D.
  • Fenretinide and placebo softgel Drug Fenretinide
  • Placebo softgel capsules Placebo Drug: Fenretinide
  • a secondary analysis of lesion growth as a function of reduction in RBP was also performed. In these correlation analyses, only patients with RBP and lesion data at baseline and 24 months were considered. Accordingly, only patients with reductions in RBP at 24 months could be considered.
  • the y-axis in each plot shows the change (increase) in lesion size (FIG. 7A-7C) or in percentage (FIG. 7D-F), relative to baseline and the x-axis shows reduction in RBP relative to baseline. Symbols in each plot represent individual patients.
  • the horizontal dashed line in each plot shows the median growth of lesions in the placebo group (-50% increase from baseline, ⁇ 2mm 2 /year).
  • VA Visual acuity
  • Luminance threshold can be measured for the resolution of a visual-acuity object.

Abstract

La présente invention a pour objet des compositions de nouvelles poudres de fenrétinide qui réduisent les niveaux de RBP sérique et/ou de rétinol-RBP sérique et qui sont utilisées pour traiter des affections ophtalmiques telles que l'atrophie géographique (GA), une forme avancée de la dégénérescence maculaire sèche liée à l'âge et pour la prévention de la néovascularisation choroïdienne (CNV, ou DMLA humide).
EP11847841.1A 2010-12-06 2011-12-05 Compositions et méthodes pour le traitement d'affections ophtalmiques Withdrawn EP2648711A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US42021110P 2010-12-06 2010-12-06
PCT/US2011/063338 WO2012078525A2 (fr) 2010-12-06 2011-12-05 Compositions et méthodes pour le traitement d'affections ophtalmiques

Publications (1)

Publication Number Publication Date
EP2648711A2 true EP2648711A2 (fr) 2013-10-16

Family

ID=46207669

Family Applications (1)

Application Number Title Priority Date Filing Date
EP11847841.1A Withdrawn EP2648711A2 (fr) 2010-12-06 2011-12-05 Compositions et méthodes pour le traitement d'affections ophtalmiques

Country Status (2)

Country Link
EP (1) EP2648711A2 (fr)
WO (1) WO2012078525A2 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10406127B2 (en) 2014-07-25 2019-09-10 Laurent Pharmaceuticals Solid oral formulation of fenretinide
AU2015292212B2 (en) 2014-07-25 2019-03-14 Laurent Pharmaceuticals Solid oral formulation of fenretinide

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005079774A2 (fr) * 2004-02-17 2005-09-01 President And Fellows Of Harvard College Gestion de troubles ophtalmologiques, notamment la degenerescence maculaire
UA81382C2 (en) * 2005-07-11 2007-12-25 Composition for treating retinol-related diseases by modulation of retinol binding
US7973079B2 (en) * 2007-09-27 2011-07-05 Revision Therapeutics, Inc. Methods and compounds for treating retinol-related diseases

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2012078525A3 *

Also Published As

Publication number Publication date
WO2012078525A2 (fr) 2012-06-14
WO2012078525A3 (fr) 2012-09-27

Similar Documents

Publication Publication Date Title
Khan et al. Chitosan coated PLGA nanoparticles amplify the ocular hypotensive effect of forskolin: Statistical design, characterization and in vivo studies
JP6480420B2 (ja) コルヒチンの徐放性製剤およびその使用方法
CN101252924B (zh) 通过血清视黄醇、血清视黄醇结合蛋白(rbp)和/或血清视黄醇-rbp调节治疗眼部疾病的方法和组合物
KR102475256B1 (ko) 미토콘드리아 및 대사 장애를 치료하는 방법
US7973079B2 (en) Methods and compounds for treating retinol-related diseases
US11730725B2 (en) Niraparib formulations
KR102475825B1 (ko) 디아미노페노티아진 (diaminophenothiazines)의 투여 및 투여량 (dosage)
KR20200118117A (ko) 소아용 니라파립 제제 및 소아용 치료 방법
JP6895752B2 (ja) ニコチン酸および/またはニコチンアミドを含む、腸内微生物叢を改変することにより血中脂質レベルに有益に影響を及ぼすための医薬組成物
KR20170092719A (ko) 안티센스 조성물 및 그의 제조 및 사용 방법
JP2010540541A (ja) レチノール関連疾患を治療するための方法及び化合物
WO2015173786A1 (fr) Compositions et méthodes pour le traitement et le diagnostic d'affections oculaires
JP2022106927A (ja) フェニル酪酸ナトリウムを含む口当たりの良い組成物及びその使用
JP2022523672A (ja) がんを治療するための金属キレート化剤併用療法
KR102438395B1 (ko) 발프로산을 포함하는 지연 방출 약제학적 제제, 및 그의 용도
Haupt et al. Lipid-based intravesical drug delivery systems with controlled release of trospium chloride for the urinary bladder
JP2019501199A (ja) プロリポソームテストステロンウンデカノエート製剤
JP6725515B2 (ja) 小分子を用いた鉄欠乏生物における生理機能の回復
EP3233808B1 (fr) Applications thérapeutiques comprenant des procédés de mesure de l'inhibition de la kinase à terminaison n c-jun dans la peau
CN105916522A (zh) 用于治疗神经变性疾病的新方法
EP2648711A2 (fr) Compositions et méthodes pour le traitement d'affections ophtalmiques
CA3085330A1 (fr) Traitement d'une maladie du tractus gastro-intestinal avec un modulateur de s1p
JP2022538898A (ja) 抗癌化合物e7766の治療コンプライアンスを向上させるシステム
US9283243B2 (en) CD36 inhibition to control obesity and insulin sensitivity
CN111954524A (zh) 秋水仙碱抑制肿瘤生长和转移的用途

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130704

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ACUCELA, INC.

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20140701