EP4096630A1 - Topical roflumilast formulation having improved delivery and plasma half-life - Google Patents

Topical roflumilast formulation having improved delivery and plasma half-life

Info

Publication number
EP4096630A1
EP4096630A1 EP21708433.4A EP21708433A EP4096630A1 EP 4096630 A1 EP4096630 A1 EP 4096630A1 EP 21708433 A EP21708433 A EP 21708433A EP 4096630 A1 EP4096630 A1 EP 4096630A1
Authority
EP
European Patent Office
Prior art keywords
roflumilast
phosphate
formulation
ceteth
topical
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
EP21708433.4A
Other languages
German (de)
English (en)
French (fr)
Inventor
David W. Osborne
Bhaskar Chaudhuri
Archie W. Thurston
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.)
Arcutis Biotherapeutics Inc
Original Assignee
Arcutis Biotherapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US16/778,845 external-priority patent/US11129818B2/en
Application filed by Arcutis Biotherapeutics Inc filed Critical Arcutis Biotherapeutics Inc
Publication of EP4096630A1 publication Critical patent/EP4096630A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the invention pertains to a method for improving the elimination kinetics for topically administered roflumilast. More particularly, the invention pertains to a pharmaceutically acceptable emulsion, suspension, gel, foam or solution formulated to have improved delivery and a longer half-life after topical administration.
  • psoriasis treatment adherence is negatively impacted if the prescribed treatment is difficult to follow because it interferes with daily routines or requires more frequent dosing than once daily.
  • adherence rates for a once daily regimen was 82% compared to only a 44% adherence rate for twice daily dosing (Zaghloul, 2004).
  • adherence with a treatment decreases if the duration of a treatment is long (Rosenstock, 1985). This means that noticeable improvement of the psoriatic plaque (defined as reduction in scale, thickness or inflammation) within the first week of topical application improves treatment adherence compared to a therapy that requires three or four weeks before noticeable clearing of disease.
  • Adherence linked with treatment implementation is the level of agreement in the patient’s actual dosing regimen and the prescribed dosing regimen between initiation and discontinuation of topical therapy.
  • adherence imperfection is most often random missed doses which can be denoted as nonconsecutive missed doses or by chance two consecutive missed doses. Three or more consecutive missed doses may be defined as a drug holiday to denote this most severe adherence imperfection.
  • the circumstance of how sensitive therapeutic success is under imperfect adherence is driven by the property known as forgiveness (Urquhart, 1997).
  • a forgiving drug would be one in which therapeutic outcomes are robust to common patterns of imperfect adherence.
  • Forgiveness is a function of the duration of action of the drug substance administered from a specific formulation and the dose interval of the drug product. When the duration of action greatly exceeds the dose interval, then the drug is considered forgiving.
  • the forgiveness index is the number of sequentially missed doses that can occur with a minimal loss of drug effect.
  • the duration of drug effect relates to the pharmacokinetic (PK) and pharmacodynamic (PD) properties inherent to the drug substance and extrinsic PK properties of the drug delivery system.
  • the plasma half-life One factor responsible for the duration of drug effect is the plasma half-life.
  • the plasma concentration of a drug is halved after one half-life. In each succeeding half-life, the plasma concentration is decreased so less drug is eliminated.
  • 50% of the drug which was absorbed remains in the body; after 2 half-lives, 25% of the drug remains in the body and after 4 half-lives, 6.25% of the drug remains in the body, which is unlikely to have a significant therapeutic effect.
  • the half-life of a drug is critical in determining an appropriate dosing interval. Small improvements in half-life for a short half-life drug can significantly lower the administered dosage. Short half-lives result in high peak to trough ratios and require more frequent dosing. More frequent dosing can result in a poor patient outcome due to a lessening of patient compliance (imperfect adherence).
  • Roflumilast is known to be suitable as a bronchial therapeutic agent as well as for the treatment of inflammatory disorders.
  • Compositions containing roflumilast are used in human and veterinary medicine and have been proposed for the treatment and prophylaxis of diseases including but not limited to: inflammatory and allergen-induced airway disorders (e.g. bronchitis, asthma, COPD); dermatoses (e.g. proliferative, inflammatory and allergen induced skin disorders), and generalized inflammations in the gastrointestinal region (Crohn's disease and ulcerative colitis).
  • Topical application of potent pharmacological agents like roflumilast for treating skin diseases has been found to provide superior delivery, lower systemic exposure and greater ease of use for patients.
  • the molecular structure of the compound ultimately dictates the ability of the drug to cross the epithelium of the tissue to which the product is applied.
  • selection of the components of the formulation dictates the maximum skin permeation that the formulator can achieve.
  • Creams, lotions, gels, ointments and foams are just a few of the more familiar forms of topical products that contain active pharmaceutical ingredients (API) for application to the skin.
  • API active pharmaceutical ingredients
  • the ability of a dissolved active ingredient to permeate the barrier of the skin is determined by its molecular structure.
  • a well -known relationship between molecular structure and skin penetration is that increasing molecular weight decreases the rate that an active crosses the skin (JD Bos, MM Meinardi, Exp Dermatol. 2000 Jun;9(3): 165-9).
  • Another well -understood relationship is that increasing the octanol- water partition coefficient of a hydrophilic active initially increases the rate that an active permeates the skin, but then decreases skin permeation once the active becomes too lipophilic to partition out of the stratum corneum and into the lower layers of the epidermis (D.W. Osborne and W.J.
  • the optimal octanol-water partition coefficient is usually at log P values of 2-3.
  • the rate that an active ingredient crosses into the viable epidermis can be further modified based on the composition of the topical product.
  • Final pH of the formulation may be critical, because dissolved ionized active ingredients typically do not permeate the skin as effectively as active ingredients that do not carry a charge (N. Li, X. Wu, W. Jia, M.C. Zhang, F. Tan, and J Zhang. Drug Dev Indust Pharm 38(8)985-994).
  • Functional ingredients such as skin penetration enhancers (D.W. Osborne and J.J.
  • this metastable topical product will equilibrate and active ingredient particles will form. If a topical product contains a volatile solvent such as ethanol, then evaporation of the solvent upon storage could result in precipitation of the active ingredient.
  • a less soluble polymorph may nucleate in the topical product and form active ingredient particles that will not re dissolve.
  • Other products may be formulated too close to the saturation limit of the active ingredient with the result that minor shifts in storage temperatures will cause precipitation. It should be noted that the dramatic temperature shifts that can occur during shipping are expected to cause the reversible precipitation of the active ingredient.
  • a suspended active ingredient properties in addition to molecular structure influence skin permeation.
  • the ratio of dissolved to suspended active ingredient can have a significant influence on the amount of active delivered after topical application. It has been shown that optimal drug delivery can be achieved for particular drugs and particular diseases by utilizing a topical composition that includes a dissolved active ingredient that has the capacity to permeate the stratum corneum layer of the epidermis and become available systemically, along with an active ingredient in a microparticulate state that does not readily cross the stratum corneum of the epidermis (US 5,863,560 hereby incorporated by reference). Another property of a suspended active ingredient that affects its delivery is the distribution of suspended particle size.
  • a topical product composition to modify the skin permeation is similar for suspended active ingredients and dissolved active ingredients. Because skin permeability is dependent upon additional properties of the suspended active ingredients, consistent delivery from topical products containing suspended actives is more difficult to maintain than for topical products containing only dissolved active ingredients.
  • Consistent delivery of a suspended active ingredient from a topical product is assured by formulation into a product in which the suspended particles do not significantly change in size or amount over the shelf life of the product.
  • Change over time in the ratio of dissolved active ingredient to particulate active ingredient can dramatically change the skin permeation of the active ingredient.
  • the same mechanisms described above (supersaturation, temperature changes, evaporation, polymorphic transformation) that can cause precipitation of dissolved active ingredients can alter the dissolved-to-particulate ratio for suspended active ingredients.
  • Change over time in the particle size or particle size distribution of the dispersed active ingredient can also dramatically change the skin permeation of the active ingredient. Sometimes this change in particle size or particle size distribution can be explained by Ostwald ripening of the particles.
  • Ostwald ripening occurs when small particles in the topical product dissolve and redeposit onto larger particles suspended in the same container of topical product. Over time this phenomenon shifts the particle size distribution toward larger particles at the expense of the smaller particles. Ostwald ripening and precipitation of a less soluble polymorph are two major problems in developing topical products containing suspended actives.
  • successful treatment also depends on the elimination kinetics of the active ingredient. After a drug is absorbed into a patient’s body, elimination begins thereby reducing the concentration over time. The half-life determines the length of time that the drug will be effective. When administering multiple 0.375 mg doses of immediate release oral roflumilast (Huang, 2018), the mean plasma half-life of the drug was found to be about 1 day (mean value of 12-subjects was 25.6 hours with a standard deviation of 8.5 hours).
  • mean plasma concentration at the time of the first missed dose (1 day after the most recent dose) will be approximately 5.7 ng/mL (half the 11.4 ng/mL maximum plasma concentration). If two consecutive doses are missed, then the roflumilast plasma concentration will be about 2.8 ng/mL two days after the most recent dose and about 1.4 ng/mL just prior to a return to treatment adherence, i.e. the patient does not miss a third consecutive dose, and taking a tablet 72 hours after the most recent oral dose of roflumilast.
  • roflumilast could be increased so that systemic plasma levels of roflumilast did not decrease to half each day that a dose is missed, then the forgiveness index for the drug delivery system would be increased.
  • use of a sustained release oral drug delivery system instead of the immediate release tablet studied by Huang would be an example of extrinsic PK property modification to increase the adherence imperfection forgiveness of the product that would benefit the patient that missed one or more doses of roflumilast.
  • a topical roflumilast formulation with a longer half-life and consistent delivery would be advantageous. It would be advantageous to develop and provide a pharmaceutical formulation containing roflumilast that after multiple topical applications could maintain consistent delivery and therapeutically effective dose levels despite adherence imperfections such as one, two or more consecutive days of missed dosing.
  • hexylene glycol inhibits crystalline growth of suspended or precipitated roflumilast particles in formulations containing pharmaceutically acceptable solvents and thus results in consistent delivery with topically applied formulations due to better skin permeability.
  • a solvent and phosphate ester surfactant blend of dicetyl phosphate and ceteth-10 phosphate in a topical formulation containing roflumilast increases the plasma half-life and the duration of effect. The increased half-life and duration of effect removes the concern that adherence imperfection will decrease therapeutic success and increases the adherence imperfection forgiveness of topically applied roflumilast.
  • Figure 1 shows Sample 19-2 “dry” roflumilast crystals from ferrer-lnterquim S.A. Batch A14367P, the drug substance used in all the examples in this specification.
  • the roflumilast crystals are 0.01mm -0.02 mm in length.
  • Figure 2 shows Sample 20-3 roflumilast crystals suspended in equimolar hexylene glycol:water solution after storage for six weeks at room temperature under 10X power.
  • the roflumilast crystals are 0.01mm -0.02 mm in length.
  • Figure 3 shows Sample 20-2 roflumilast crystals suspended in equimolar diethylene glycol monoethyl ethenwater solution after storage for six weeks at room temperature.
  • the roflumilast crystals are 0.04mm -0.20 mm in length and 0.01mm -0.02 mm in width.
  • Figure 4 shows Sample 20-3 roflumilast crystals suspended in equimolar hexylene glycol:water solution after storage for six weeks at room temperature under 4X power.
  • the roflumilast crystals are 0.01mm -0.02 mm in length.
  • Figure 5 shows Sample 21-2 roflumilast crystals suspended in equimolar ethanol:water solution after storage for six weeks at room temperature.
  • the roflumilast crystals are 0.05mm -0.25 mm in length and 0.02 mm in width.
  • Figure 6 shows Sample 21-3 roflumilast crystals suspended in equimolar PEG 400:water solution after storage for six weeks at room temperature.
  • the roflumilast crystals are 0.05mm -0.07 mm in length and 0.02 mm in width.
  • Figure 7 shows Sample 21-4 roflumilast crystals suspended in equimolar DMSO:water solution after storage for six weeks at room temperature.
  • the roflumilast crystals are 0.10mm -0.67 mm in length and 0.02mm -0.10 mm in width.
  • Figure 8 shows Sample 21-5 roflumilast crystals suspended in equimolar propylene glycol:water solution after storage for six weeks at room temperature.
  • the roflumilast crystals are 0.20mm - 1.60 mm in length and 0.02 mm in width.
  • Figure 9 shows Sample 20-1 roflumilast crystals suspended in equimolar NMP:water solution after storage for six weeks at room temperature.
  • the roflumilast crystals are 0.10mm - 1.55 mm in length and 0.02mm -0.13 mm in width.
  • the roflumilast crystals are 0.02 mm - 0.04 mm in length and 0.02 mm in width.
  • Figures 11A and 11 B show roflumilast particles precipitated in a cream composition after one freeze thaw cycle.
  • Figure 11a shows Sample 36-1 roflumilast particles precipitated in a cream composition with diethylene glycol monoethyl ether (DEGEE) and without hexylene glycol. The three largest roflumilast particles were measured (0.07 mm X 0.09 mm; 0.06 mm X 0.06 mm; and 0.10 mm X 0.05 mm) and found to have a mean surface area of 5,000 square microns.
  • Figure 11 b shows Sample 36-2 roflumilast particles precipitated in a cream composition with both diethylene glycol monoethyl ether (DEGEE) and hexylene glycol.
  • DEGEE diethylene glycol monoethyl ether
  • the three largest roflumilast particles were measured (0.05 mm X 0.03 mm; 0.05 mm X 0.03 mm and 0.05 mm X 0.03 mm) and found to have a mean surface area of 1 ,500 square microns.
  • Roflumilast is a compound of the formula (I) (I) wherein R1 is difluoromethoxy, R2 is cyclopropylmethoxy and R3 is 3,5-dichloropyrid-4- yi.
  • This compound has the chemical name N-(3,5-dichloropyrid-4-yl)-3- cyclopropylmethoxy-4-difluoromethoxybenzamid- e (INN: roflumilast).
  • Hexylene glycol (PharmaGrade. USP/NF) is 2-methyl-2,4-pentanediol of the formula (II).
  • the emulsifier blend of cetearyl alcohol (CAS 67762300), dicetyl phosphate (CAS 2197639) and ceteth-10 phosphate (CAS 50643-20-4) is manufactured by Croda under the tradename CRODAFOSTM CES.
  • This commercially available emulsifier blend is a self-emulsifying wax that is predominately the waxy material cetearyl alcohol (which is a mixture cetyl alcohol (C16H34O) and stearyl alcohol (C18H38O)) combined with 10- 20% dicetyl phosphate and 10-20% ceteth-10 phosphate.
  • Self-emulsifying waxes form an emulsion when blended with water.
  • CRODAFOSTM CES When CRODAFOSTM CES is added to water it spontaneously forms an emulsion having a pH of about 3. Sodium hydroxide solution is added to increase the pH to the desired value.
  • Ceteth-10 Phosphate The present invention is directed to the addition of hexylene glycol, a solvent and/or a phosphate ester surfactant blend of dicetyl phosphate and ceteth-10 phosphate to a roflumilast-containing pharmaceutical composition that contains a pharmaceutically acceptable solvent, including water, to inhibit growth of roflumilast crystals in the composition and/or to extend the half-life of rolfumilast in a patient’s body after administration.
  • a pharmaceutically acceptable solvent including water
  • hexylene glycol For topical products designed to contain suspended roflumilast particles or crystals, the addition of hexylene glycol to a composition containing roflumilast, will inhibit (i.e. , prevent or substantially reduce in comparison to compositions that do not contain a hexylene glycol) changes in particle size distribution over the shelf life of the product and assure consistent bioavailability.
  • hexylene glycol inhibits the growth of precipitated roflumilast particles.
  • Typical storage conditions for a topical pharmaceutical cream are: Store at room temperature: 60°F/15°C-80°F/26°C. Do not freeze. It is understood by product development scientists and regulatory agency reviewers that a topical product will not always be stored over this temperature range. Therefore, the FDA requires that all topical products undergo freeze-thaw cycling and temperature excursion studies. The active is neither required nor expected to remain in solution when the product is exposed to temperatures of -20°C, dramatically below 15°C (60°F) of the labeled storage condition.
  • Hexylene glycol can be added between 0.1% and 20% on a weight/weight basis, preferably between 0.25% and 8% on a weight/weight basis and most preferably between 0.5% and 2% on a weight/weight basis.
  • a self-emulsifying wax blend of dicetyl phosphate and ceteth-10 phosphate and a solvent is included in the formulation.
  • the plasma half-life of roflumilast after intravenous administration in humans is about 15 hours; after oral administration the plasma half-life is about 17-30 hours (see Bethke et al. , High Absolute Bioavailability of the New Oral Phosphodiesterase-4 Inhibitor Roflumilast, International Journal of Clinical Pharmacology and Therapeutics, vol. 49, No.1, 2011, pp.51-57).
  • the plasma half-life of roflumilast after topical administration of a formulation comprising a self-emulsifying wax blend of dicetyl phosphate and ceteth-10 phosphate and a solvent is 3.4-3.7 days (about 81-89 hours).
  • the topical roflumilast product formulations that benefit from the addition of hexylene glycol, a self-emulsifying wax blend of dicetyl phosphate and ceteth-10 phosphate and/or a solvent include but are not limited to aerosols, foams, sprays, emulsions (which can also be called creams, lotions, or ointments), gels (two phase or single phase), liquids, ointments, pastes, shampoos, suspensions, and systems. These are the tier two terms within compendia taxonomy for dosage forms containing pharmaceutical active ingredients (US Pharmacopeia ⁇ 1151 >).
  • the roflumilast formulations can be prepared by methods known in the art (e.g. see the ’298 patent and U.S. Appln No. 14/075,035).
  • compositions containing 0.005 - 2.0% roflumilast that may be in one of the following forms:
  • the product may be a formulation in which hexylene glycol, a self-emulsifying wax blend of dicetyl phosphate and ceteth-10 phosphate, and/or a solvent is added to an emulsion comprising a discrete phase of a hydrophobic component and a continuous aqueous phase that includes water and optionally one or more polar hydrophilic excipients as well as additional solvents, co-solvents, salts, surfactants, emulsifiers, and other components.
  • These emulsions may include water- soluble or water-swellable polymers that help to stabilize the emulsion.
  • the emulsifier is a self-emulsifying wax blend of dicetyl phosphate and ceteth-10 phosphate.
  • Thickened Aqueous gels These systems include an aqueous phase which has been thickened by suitable natural, modified natural, or synthetic thickeners such as described below. Alternatively, the thickened aqueous gels can be thickened using suitable polyethoxylate alky chain surfactants or other nonionic, cationic, or anionic systems.
  • Thickened Hydroalcoholic gels These systems include a blend of water and alcohol as the polar phase which has been thickened by suitable natural, modified natural, or synthetic polymers such as described below.
  • the thickened hydroalcoholic gels can be thickened using suitable polyethoxylate alky chain surfactants or other nonionic, cationic, or anionic systems.
  • the alcohol can be ethanol, isopropyl alcohol or other pharmaceutically acceptable alcohol.
  • Hydrophilic gels These are systems in which the continuous phase includes at least one water soluble or water dispersible hydrophilic component other than water.
  • the formulations may optionally also contain water up to 60% by weight. Higher levels may be suitable in some compositions.
  • Suitable hydrophilic components include one or more glycols such as polyols such as glycerin, propylene glycol, butylene glycols, polyethylene glycols (PEG), random or block copolymers of ethylene oxide, propylene oxide, and/or butylene oxide, polyalkoxylated surfactants having one or more hydrophobic moieties per molecule, silicone copolyols, blend of ceteareth-6 and stearyl alcohol as well as combinations thereof, and the like.
  • glycols such as polyols such as glycerin, propylene glycol, butylene glycols, polyethylene glycols (PEG), random or block copolymers of ethylene oxide, propylene oxide, and/or butylene
  • a water-in-oil emulsion The compositions may be formulations in which roflumilast is incorporated into an emulsion that includes a continuous phase of a hydrophobic component and an aqueous phase that includes water and optionally one or more polar hydrophilic carrier(s) as well as salts or other components.
  • These emulsions may include oil-soluble or oil-swellable polymers as well as one or more emulsifier(s) that help to stabilize the emulsion.
  • the emulsifier is a self- emulsifying wax blend of dicetyl phosphate and ceteth-10 phosphate.
  • a hydrophilic or hydrophobic ointment The compositions are formulated with a hydrophobic base (e.g. petrolatum, thickened or gelled water insoluble oils, and the like) and optionally having a minor amount of a water soluble phase. Hydrophilic ointments generally contain one or more surfactants or wetting agents
  • Compositions according to the present invention may include one or more solvents or co-solvents to obtain the desired level of active ingredient solubility in the topical product.
  • the solvent may also modify skin permeation or the activity of other excipients contained in the formulation.
  • Solvents include but are not limited to acetone, ethanol, benzyl alcohol, butyl alcohol, diethyl sebacate, diethylene glycol monoethyl ether, diisopropyl adipate, dimethyl sulfoxide, ethyl acetate, isopropyl alcohol, isopropyl isostearate, isopropyl myristate, N-methyl pyrrolidinone, polyethylene glycol, glycerol, propylene glycol and SD alcohol.
  • compositions according to the present invention may include a moisturizer to increase the level of hydration.
  • the moisturizer can be a hydrophilic material including humectants or it can be a hydrophobic material including emollients.
  • Suitable moisturizers include but are not limited to:1,2,6-hexanetriol, 2-ethyl-1,6-hexanediol, butylene glycol, glycerin, polyethylene glycol 200-8000, butyl stearate, cetostearyl alcohol, cetyl alcohol, cetyl esters wax, cetyl palmitate, cocoa butter, coconut oil, cyclomethicone, dimethicone, docosanol, ethylhexyl hydroxystearate, fatty acids, glyceryl isostearate, glyceryl laurate, glyceryl monostearate, glyceryl oleate, glyceryl palmitate, glycol distearate
  • compositions according to the present invention optionally can include one or more surfactants to emulsify the composition and to help wet the surface of the actives or excipients.
  • surfactant means an amphiphile (a molecule possessing both polar and nonpolar regions which are covalently bound) capable of reducing the surface tension of water and/or the interfacial tension between water and an immisicible liquid.
  • Surfactants include but are not limited to alkyl aryl sodium sulfonate, Amerchol-CAB, ammonium lauryl sulfate, apricot kernel oil PEG-6 esters, Arlacel, benzalkonium chloride, Ceteareth-6, Ceteareth-12, Ceteareth-15, Ceteareth-30, cetearyl alcohol/ceteareth-20, cetearyl ethylhexanoate, ceteth-10, ceteth-2, ceteth-20, ceteth-23, choleth-24, cocamide ether sulfate, cocamine oxide, coco betaine, coco diethanolamide, coco monoethanolamide, coco-caprylate/caprate, disodium cocoamphodiacetate, disodium laureth sulfosuccinate, disodium lauryl sulfoacetate, disodium lauryl sulfosuccinate, disodium oleamido monoethanolamine sulfos
  • soluble, swellable, or insoluble organic polymeric thickeners such as natural and synthetic polymers or inorganic thickeners such as acrylates copolymer, carbomer 1382, carbomer copolymer type B, carbomer homopolymer type A, carbomer homopolymer type B, carbomer homopolymer type C, carboxy vinyl copolymer, carboxymethylcellulose, carboxypolymethylene, carrageenan, guar gum, hydroxyethyl cellulose, hydroxypropyl cellulose, microcrystalline wax, and methylcellulose,
  • soluble, swellable, or insoluble organic polymeric thickeners such as natural and synthetic polymers or inorganic thickeners such as acrylates copolymer, carbomer 1382, carbomer copolymer type B, carbomer homopolymer type A, carbomer homopolymer type B, carbomer homopolymer type C, carboxy vinyl copolymer, carboxymethylcellulose, carboxypolymethylene, carrage
  • compositions according to the present invention may be formulated with additional components such as fillers, carriers and excipients conventionally found in cosmetic and pharmaceutical topical products.
  • Additional components including but not limited to antifoaming agents, preservatives (e.g. p-hydroxybenzoic esters, benzyl alcohol, phenylmercury salts, chlorocresol), antioxidants, sequestering agents, stabilizers, buffers, pH adjusting solutions, skin penetration enhancers, film formers, dyes, pigments, diluents, bulking agents, fragrances and other excipients to improve the stability or aesthetics, may be added to the composition.
  • preservatives e.g. p-hydroxybenzoic esters, benzyl alcohol, phenylmercury salts, chlorocresol
  • antioxidants e.g. p-hydroxybenzoic esters, benzyl alcohol, phenylmercury salts, chlorocresol
  • sequestering agents e.g
  • compositions according to the present invention may be formulated with additional active agents depending on the condition being treated.
  • additional active agents include but are not limited to NSAIDs (e.g. Aspirin, Ibuprofen, Ketoprofen, Naproxen), Apremilast, JAK inhibitors (e.g. Tofacitinib, Ruxolitinib, Oclacit), leukotriene inhibitors (e.g. Zileuton, Zafirlukast, Montelukast), mast cell stabilizers (e.g. Nedocromil, Cromolyn sodium, Ketotifen, Pemirolast), Anthralin (dithranol), Azathioprine,
  • NSAIDs e.g. Aspirin, Ibuprofen, Ketoprofen, Naproxen
  • JAK inhibitors e.g. Tofacitinib, Ruxolitinib, Oclacit
  • leukotriene inhibitors e
  • Tacrolimus Coal tar, Methotrexate, Methoxsalen, Salicylic acid, Ammonium lactate, Urea, Hydroxyurea, 5-fluorouracil, Propylthouracil, 6-thioguanine, Sulfasalazine, Mycophenolate mofetil, Fumaric acid esters, Corticosteroids (e.g.
  • calcipotriene calcitriol
  • Acitretin Tazarotene
  • Cyclosporine e.g. adenosine
  • Resorcinol e.g. adenosine
  • Colchicine e.g. adenosine
  • antibiotics e.g. erythromycin, ciprofloxacin, metronidazole
  • compositions according to the present invention can be administered by any suitable administration route including but not limited to oral, rectal, parenteral (e.g. intradermal, subcutaneous, intramuscular, intravenous, intramedullary, intra arterial, intrathecal, epidural), ocular, inhalation, nebulization, cutaneously (topically), transdermally, and mucosally (e.g. sublingual, buccal, nasally).
  • parenteral e.g. intradermal, subcutaneous, intramuscular, intravenous, intramedullary, intra arterial, intrathecal, epidural
  • ocular, inhalation nebulization
  • cutaneously topically
  • transdermally e.g. sublingual, buccal, nasally
  • mucosally e.g. sublingual, buccal, nasally
  • Suitable pharmaceutical dosage forms include but are not limited to emulsions, suspensions, sprays, oils, ointments, fatty ointments, creams, pastes, gels, foams transdermal patches and solutions (e.g. injectable, oral).
  • the composition preferably contains roflumilast, salts of roflumilast, the N-oxide of roflumilast or salts thereof in an amount of 0.005 - 2 % w/w, more preferably 0.05 - 1% w/w, and most preferably 0.1 - 0.5% w/w per dosage unit.
  • the composition preferably contains hexylene glycol in an amount of between 0.1% and 20% w/w, more preferably between 0.25% and 8% w/w and most preferably between 0.5% and 2% w/w.
  • the composition preferably contains a phosphate ester surfactant in the formulation which is in an amount sufficient to produce a stable emulsion having uniform globule size.
  • concentration of the phosphate ester surfactant generally may be any concentration between 1.0% to 25% w/w.
  • the preferred concentration can be different for different administration forms.
  • the concentration of the phosphate ester surfactant is between 2.5% and 20%, with a more preferred concentration range between 5% and 15%, and a most preferred concentration being about 10% w/w.
  • the concentration is preferably between 1.0%-10%, more preferably between 1.0% - 10%, and most preferably 2%.
  • the phosphate ester surfactant is provided in a self-emulsifying wax blend of dicetyl phosphate and ceteth-10 phosphate.
  • the composition preferably contains a solvent in an amount sufficient to obtain the desired level of active ingredient solubility in the formulation.
  • the solvent is preferably is in an amount of 10-30% (w/w).
  • the ratio of solvent to water is preferably from 1:10 to 20:1.
  • the solvent is diethylene glycol monoethyl ether (DEGEE).
  • the topical formulation containing roflumilast is applied to the skin in an amount that is sufficient to obtain the desired pharmacologic effect, which typically is to ameliorate the signs and/or symptoms of a medical disorder.
  • the amount of the formulation that is applied may vary depending on the amount of roflumilast that is contained within the formulation, the concentration of the roflumilast within the formulation, and the frequency in which the formulation is intended to be applied.
  • the formulation is applied with a frequency between weekly to several times daily, preferably between every other day to three times daily, and most preferably one or two times daily.
  • the composition can be used in veterinary and in human medicine for the treatment and prevention of all diseases regarded as treatable or preventable by using roflumilast, including but not limited to acute and chronic airway disorders such as bronchitis, allergic bronchitis, asthma, and COPD; proliferative, inflammatory and allergic dermatoses such as psoriasis, scalp psoriasis, or inverse psoriasis, irritant and allergic contact eczema, hand eczema, atopic dermatitis, seborrheic dermatitis, lichen simplex, sunburn, aphthous ulcers, lichen planus, vitiligo, pruritus in the genital or anal regions, alopecia areata, hypertrophic scars, discoid lupus erythematosus, follicular and extensive pyodermas, endogenous and exogenous acne, acne rosacea, disorders which are
  • the composition is used to treat proliferative, inflammatory and allergic dermatoses such as psoriasis (vulgaris), eczema, acne, Lichen simplex, sunburn, pruritus, alopecia areata, hypertrophic scars, discoid lupus erythematosus, and pyodermias.
  • the composition can include additional active agents suitable for treating the patient’s condition.
  • the composition may additionally include Anthralin (dithranol), Azathioprine, Tacrolimus, Coal tar, Methotrexate, Methoxsalen, Salicylic acid, Ammonium lactate, Urea, Hydroxyurea, 5-fluorouracil, Propylthouracil, 6-thioguanine, Sulfasalazine, Mycophenolate mofetil, Fumaric acid esters, Corticosteroids (e.g.
  • Triamcinolone Fluocinolone, Fluocinonide, Flurandrenolide, Diflorasone, Desonide, Desoximetasone, Dexamethasone, Halcinonide, Halobetasol, Hydrocortisone, Methylprednisolone
  • Colchicine Adalimumab, Ustekinumab, Infliximab, and/or antibiotics.
  • the formulation for topical application containing roflumilast may be prepared by processes typically used in the field of manufacture of pharmaceutical formulations for topical application.
  • a single-phase formulation such as a liquid
  • the constituents of the formulation may be combined and mixed until a homogenous solution or suspension of the active ingredient is obtained.
  • a multiphase formulation such as an emulsion
  • the components of the aqueous phase and of the oil phase may be separately combined and mixed until homogenous solutions are obtained and then the aqueous solution and the oil solution may be combined and mixed, such as by shear mixing, to form the formulation.
  • the one or more drug actives may be dissolved (molecularly dispersed), complexed, or associated with an excipient or other active, or may be particulate (amorphous or crystalline).
  • the oil phase may be added to the water phase, or the water phase may be added to the oil phase.
  • the phases may be combined and mixed, such as at elevated temperatures of 50-90°C or at room temperature, that is between 20-30°C, or at a temperature between room temperature and the elevated temperatures.
  • roflumilast API (Batch A14367P from Interquim S.A.) dry powder was tapped onto a microscope slide, a coverslip was moved into place and crystal habit and particle size of the API was examined using polarized light microscopy using a 10X objective (figure 1, microscope sample 19-2).
  • EXAMPLE 2 0.0092 grams of roflumilast (Batch A14367P from Interquim S.A.) was weighed into a liquid scintillation vial. An equimolar blend of hexylene glycol (lot 1AC0818, Spectrum) and distilled water was added dropwise with mixing to the vial containing roflumilast to produce a suspension of roflumilast in excess of the solubility limit. An equimolar blend is 86.7% hexylene glycol and 13.3% water on a weight/weight percent basis. After mixing each addition of hexylene glycol:water blend, the tightly capped vial was returned to a water bath set at 25°C.
  • roflumilast 0.0064 grams was added to this sample (labeled 12-3) to form a finely dispersed suspension at 25°C and the vial was then stored undisturbed at about 15- 18°C, protected from the light for six weeks.
  • a sample of the roflumilast crystals was removed from the vial, placed on a microscope slide (with coverslip) and then examined using polarized light microscopy using a 4X objective (figure 4, microscope sample 20- 3).
  • roflumilast Batch A14367P from Interquim S.A.
  • Polyethylene glycol 400 (lot 1 DE0880, Spectrum) was added dropwise with mixing to the vial containing roflumilast to produce a suspension of roflumilast in excess of the solubility limit.
  • the tightly capped vial was returned to a water bath set at 25°C. It required 0.5486 grams of propylene glycol 400 to completely dissolve the 0.0180 grams of roflumilast and give a 3.18% roflumilast in polyethylene glycol 400 solution.
  • This sample (labeled as “PEG 400” page 1) was a solution at 25°C and was then stored undisturbed at about 15-18°C, protected from the light for six weeks. Roflumilast crystals precipitated due to the cooler storage temperature. A sample of the roflumilast crystals was removed from the vial, placed on a microscope slide (with coverslip) and then examined using polarized light microscopy using a 4X objective (figure 6, microscope sample 21-3).
  • a formulation of the invention hereafter referred to as Formulation 3, was made by combining roflumilast at a concentration of 0.3 % w/w with Crodafos CES (the phosphate ester surfactant blend of dicetyl phosphate and ceteth-10 phosphate combined with cetostearyl alcohol) and adding diethylene glycol monoethyl ether, as well as other ingredients to create a complete marketable formulation.
  • This formulation was buffered with NaOH to obtain a pH of 5.5.and water.
  • Formulation 3 is the same as formulation 2 except that formulation 3 contains 0.3% roflumilast and formulation 2 contains 0.5% roflumilast.
  • Comparative Formulation 4 A formulation that is not of the invention, hereafter referred to as Comparative Formulation 4, was made by combining roflumilast at a concentration of 0.3 % w/w with Crodafos CES (the phosphate ester surfactant blend of dicetyl phosphate and ceteth-10 phosphate combined with cetostearyl alcohol) and water. The formulation was buffered with NaOH to obtain a pH of 5.5. Comparative Formulation 4 does not contain diethylene glycol monoethyl ether or hexylene glycol.
  • Crodafos CES the phosphate ester surfactant blend of dicetyl phosphate and ceteth-10 phosphate combined with cetostearyl alcohol
  • Comparative Formulation 5 A formulation that is not of the invention, hereafter referred to as Comparative Formulation 5, was made by combining roflumilast at a concentration of 0.2%. This formulation is that of the closest prior art known to the inventors and is disclosed in Example 3 of Bolle et al, U.S. Patent Application No. US 2006/0084684.
  • Comparative Formulation 6 A formulation that is not of the invention, hereafter referred to as Comparative Formulation 6, was made by combining roflumilast at a concentration of 0.3%.
  • This formulation contains the phosphate ester potassium cetyl phosphate (Crodafos MCK) as the emulsifier but does not contain dicetyl phosphate or ceteth-10 phosphate which are the phosphate ester emulsifiers contained in the self-emulsifying wax Crodafos CES.
  • compositions of these formulations are shown below in Table 1.
  • Crodafos CES is consistent between batches of product but is not publicly disclosed by the manufacturer (Croda).
  • the safety data sheet for Crodafos CES states that this emulsifier is composed of 60-80% cetostearyl alcohol, 10-20% dicetyl phosphate and 10-20% cetheth-10 phosphate.
  • Formulation 5 phosphate-ester surfactant blend
  • Formulation 6 nonionic surfactant blend
  • Glyceryl Stearate/P EG-100 Stearate is the nomenclature used by the US Food and Drug Administration to describe the nonionic emulsifier blend sold using the tradename Arlacel ® 165 and Tego Care ® 165.
  • Medium-Chain Triglycerides is the nomenclature used by the US Food and Drug Administration to describe the cosmetic ingredient Capryli/Capric Triglyceride which is sold using tradenames including Miglyol ® 812 and Crodamol ® GTCC.
  • Example 6 Elimination Kinetics after 14-Day Dosing of Formulations of Example 5
  • Male and female swine (Gottingen Minipig® breed) are ordered to weigh 8 to 12 kg at arrival.
  • the hair is clipped from the back of each animal.
  • the pigs are sedated for the shaving procedure. Care is taken to avoid abrading the skin.
  • the formulation containing the phosphate ester surfactant Crodafos CES, hexylene glycol and the solvent diethylene glycol monoethyl ether showed significant increase in the plasma half-life and duration of effect of roflumilast after topical application.
  • Comparative Formulation 5 The closest prior art formulation (Comparative Formulation 5) and Comparative Formulation 6, a cream formulation that replaced Crodafos CES (the phosphate ester surfactant blend of dicetyl phosphate and ceteth-10 phosphate combined with cetostearyl alcohol) with Crodafos MKC (the phosphate ester surfactant potassium cetyl phosphate combined with cetostearyl alcohol) had decreased plasma half-lives compared to Formulation 3 that contained Crodafos CES.
  • Crodafos CES the phosphate ester surfactant blend of dicetyl phosphate and ceteth-10 phosphate combined with cetostearyl alcohol
  • Crodafos MKC the phosphate ester surfactant potassium cetyl phosphate combined with cetostearyl alcohol
  • Comparative Formulation 4 that did not combine hexylene glycol and diethylene glycol monoethyl ether with the phosphate ester surfactant blend of dicetyl phosphate and ceteth-10 phosphate combined with cetostearyl alcohol (Crodafos CES) did not have an increased half-life and duration of roflumilast effect after topical application in pigs.
  • a roflumilast cream was prepared according to the following formulation.
  • Subjects were adult (>18 y/o) males or females with chronic plaque psoriasis. Subjects had an Investigator’s Global Assessment of disease severity (IGA) of at least Mild (‘2’) at Baseline. Subjects with an IGA of ‘Mild’ (2) were limited to 20% of total enrollment. Subjects with an IGA of ‘Severe’
  • BSA Body Surface Area
  • IGA intertriginous area lesion IGA score
  • Concentration values that are reported BLQ were reported as below limit of quantification (BLQ) in the concentration data listings and considered non-numerical. Nonnumerical values were ignored in the concentration by nominal time summary statistics. Summary statistics (i.e. N, arithmetic mean, SD, CV%, minimum, median, maximum, geometric mean, geometric SD, 95% confidence intervals for both the arithmetic mean and geometric mean) were calculated for plasma concentrations for each analyte, nominal day and dose strength. All concentration and descriptive statistic values were reported to three significant figures.
  • Mean N-oxide pre-dose concentrations were 11.2 and 9.18 ng/mL following topical administration of ARQ-151 0.3% on Day 29 and Day 85, respectively. Normalizing the BSA treated, the mean concentration values were 2.66 and 2.10 ng/mL, respectively. Mean N-oxide pre-dose concentrations were 6.53 and 4.63 ng/mL following topical administration of ARQ-151 0.15% on Day 29 and Day 85, respectively. Normalizing the BSA treated, the mean concentration values were 1.68 and 1.28 ng/mL, respectively. Overall, mean roflumilast and the N-oxide pre-dose plasma concentration values were within 2-fold of each other on Day 29 and Day 85.
  • the arithmetic mean (AM) Day 15 pre-dose concentrations following topical administration of ARQ-151 0.15% in patients with atopic dermatitis with a mean BSA treated area of 6.5% (n 6) was 1.99 ng/mL (Study ARQ-151 -102), dose normalized of 0.306 ng/mL.
  • an extrapolated AUC value for the ARQ-151-102 study would be 47.8 or 21.0 h*ng/ml_ - which is in good alignment with the measured values. From this study, using the Day 29 pre-dose concentrations, the extrapolated AM and GM AUC values would be 26.9 and 18.6 h*ng/ml_
  • Roflumilast plasma pre-dose concentration values increase about 1.6- and 1.7- fold on Day 29 and Day 85, respectively between the 2-fold increase in dose strength.
  • the roflumilast N-oxide (N-oxide) plasma pre-dose concentration values increase about 1.7- and 2.0-fold on Day 29 and Day 85, respectively between the 2-fold increase in dose strength.
  • the first BLQ value was set equal to 0.05 ng/mL
  • the plasma half-life of roflumilast after intravenous administration is about 15 hours; after oral administration the plasma half-life is about 17-30 hours.
  • the plasma half-life of roflumilast after topical administration of a formulation comprising hexylene glycol, diethylene glycol monoethyl ether (Transcutol P) and a self-emulsifying wax blend of dicetyl phosphate and ceteth-10 phosphate is 3.4-3.7 days (about 81-89 hours).
  • the increased half-life and duration of effect removes the concern that adherence imperfection will decrease therapeutic success and increases the adherence imperfection forgiveness of topically applied roflumilast.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Dermatology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Dispersion Chemistry (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
EP21708433.4A 2020-01-31 2021-01-29 Topical roflumilast formulation having improved delivery and plasma half-life Pending EP4096630A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US16/778,845 US11129818B2 (en) 2017-06-07 2020-01-31 Topical roflumilast formulation having improved delivery and plasma half life
PCT/US2021/015740 WO2021155173A1 (en) 2020-01-31 2021-01-29 Topical roflumilast formulation having improved delivery and plasma half-life

Publications (1)

Publication Number Publication Date
EP4096630A1 true EP4096630A1 (en) 2022-12-07

Family

ID=74759470

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21708433.4A Pending EP4096630A1 (en) 2020-01-31 2021-01-29 Topical roflumilast formulation having improved delivery and plasma half-life

Country Status (10)

Country Link
EP (1) EP4096630A1 (ko)
JP (1) JP2023513092A (ko)
KR (1) KR20220134617A (ko)
CN (1) CN115551478A (ko)
AU (1) AU2021214399A1 (ko)
BR (1) BR112022015104A2 (ko)
CA (1) CA3166300A1 (ko)
IL (1) IL295172A (ko)
MX (1) MX2022009399A (ko)
WO (1) WO2021155173A1 (ko)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024058848A1 (en) * 2022-09-15 2024-03-21 Arcutis Biotherapeutics, Inc. Pharmaceutical compositions of roflumilast and solvents capable of dissolving high amounts of the drug

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT706513E (pt) 1993-07-02 2002-10-31 Byk Gulden Lomberg Chem Fab Benzimidazois substituidos por fluoralcoxi e sua utilizacao como inibidores de fosfodiesterase nucleotidica ciclica
US5863560A (en) 1996-09-11 1999-01-26 Virotex Corporation Compositions and methods for topical application of therapeutic agents
ME00565A (en) 2002-05-28 2011-12-20 Topically applicable pharmaceutical preparation
DE102004046235A1 (de) 2004-09-22 2006-03-30 Altana Pharma Ag Arzneimittelzubereitung
US9895359B1 (en) 2017-06-07 2018-02-20 Arcutis, Inc. Inhibition of crystal growth of roflumilast
US11534493B2 (en) * 2017-09-22 2022-12-27 Arcutis Biotherapeutics, Inc. Pharmaceutical compositions of roflumilast in aqueous blends of water-miscible, pharmaceutically acceptable solvents
KR20210044191A (ko) * 2018-06-04 2021-04-22 아큐티스, 인크. 로플루밀라스트 피부 침투 지연 시간의 개선 방법 및 제형
US20200155524A1 (en) * 2018-11-16 2020-05-21 Arcutis, Inc. Method for reducing side effects from administration of phosphodiesterase-4 inhibitors

Also Published As

Publication number Publication date
BR112022015104A2 (pt) 2022-09-27
KR20220134617A (ko) 2022-10-05
AU2021214399A1 (en) 2022-08-25
JP2023513092A (ja) 2023-03-30
MX2022009399A (es) 2022-09-19
WO2021155173A1 (en) 2021-08-05
CA3166300A1 (en) 2021-08-05
IL295172A (en) 2022-09-01
CN115551478A (zh) 2022-12-30

Similar Documents

Publication Publication Date Title
US11793796B2 (en) Inhibition of crystal growth of roflumilast
US12005052B2 (en) Topical roflumilast formulation having improved delivery and plasma half-life
JP7402301B2 (ja) 水混和性の薬学的に許容可能な溶媒の水性ブレンド中のロフルミラストの医薬組成物
US20210161870A1 (en) Roflumilast formulations with an improved pharmacokinetic profile
WO2021155173A1 (en) Topical roflumilast formulation having improved delivery and plasma half-life
US12011437B1 (en) Roflumilast formulations with an improved pharmacokinetic profile
US12016848B2 (en) Roflumilast formulations with an improved pharmacokinetic profile
WO2022169615A1 (en) Roflumilast formulations with an improved pharmacokinetic profile

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

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

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20220802

AK Designated contracting states

Kind code of ref document: A1

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

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230514

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40084898

Country of ref document: HK