WO2019087158A2 - Formulation comprising oxazolidin-2-one-pyrimidine derivative - Google Patents

Formulation comprising oxazolidin-2-one-pyrimidine derivative Download PDF

Info

Publication number
WO2019087158A2
WO2019087158A2 PCT/IB2018/058677 IB2018058677W WO2019087158A2 WO 2019087158 A2 WO2019087158 A2 WO 2019087158A2 IB 2018058677 W IB2018058677 W IB 2018058677W WO 2019087158 A2 WO2019087158 A2 WO 2019087158A2
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical composition
composition
topical pharmaceutical
total weight
formula
Prior art date
Application number
PCT/IB2018/058677
Other languages
French (fr)
Other versions
WO2019087158A3 (en
Inventor
Eric Aubin
Paolo DE MARCO
Rajesh A. KUMAR
Andreas Lerchner
Isabelle Rault
Bernd Ulrich Riebesehl
Frank Hans Seiler
Karin SVEDBERG
Henricus Lambertus Gerardus Maria Tiemessen
Original Assignee
Novartis Ag
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 Novartis Ag filed Critical Novartis Ag
Publication of WO2019087158A2 publication Critical patent/WO2019087158A2/en
Publication of WO2019087158A3 publication Critical patent/WO2019087158A3/en

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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • 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
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to novel topical pharmaceutical compositi
  • compositions as well as uses of such compositions.
  • the present invention also relates to a novel crystalline form of the compound of formula (I). BACKGROUND OF THE INVENTION
  • the compound of formula (I) is useful for the treatment or prevention of a disease or disorder selected from non-melanoma skin cancers such as basal cell carcinoma and squamous cell carcinoma (SCC); their pre-malignant stages such as squamous cell carcinoma in situ (SCCis), actinic keratosis (AK), solar keratosis and chronically sun damaged skin; and other hyperproliferative skin disorders caused by dysregulation of skin fibroblasts such as skin fibrosis, scleroderma, hypertrophic scars and keloids.
  • compositions that may improve efficacy, bioavailability, stability, and/or acceptance by the patient and that may reduce potential side effects.
  • compositions as described herein by providing the composition for use in diseases, particularly for the treatment of dermatological diseases as described herein, and by providing a process to produce the composition as described herein, as well as by providing the compound of formula (I) in crystalline form.
  • Figure 1 x-ray powder diffraction (XRPD) spectrum of the compound of formula (I) in crystalline form.
  • Figure 2 Differential scanning calorimetry (DSC) curve of the compound of formula (I) in crystalline form.
  • TGA Thermogravimetric analysis
  • Figure 4 FT-IR spectrum of the compound of formula (I) in crystalline form.
  • Figure 5 Scanning electron microscope (SEM) picture of the compound of formula (I) in crystalline form.
  • the amorphous compound of formula (I) showed some tendency to light induced degradation; 8% degradation was observed in the photostability assay described in WO2014072956.
  • the amorphous compound of formula (I) showed a weak positive signal in the in vitro phototoxicity assay (3T3) with a photoirritation factor (PIF) of 7, indicating an increased phototoxicity potential.
  • the compound of formula (I) can be obtained in crystalline form, which shows an enhanced photostability compared to the compound of formula (I) in amorphous form known from WO2014072956.
  • active agent refers to a compound of formula (I)
  • Active agent is also intended to represent amorphous and crystalline forms such as polymorphs.
  • Active agent is also intended to represent a solvate thereof, a pharmaceutical acceptable salt thereof and its mixtures.
  • Active agent is also intended to represent material exhibiting specific solid state properties such as specific crystal shapes and/or milled forms of the “Active agent”, e.g. in micronized form.
  • R represents a Ci- 6 alkyl group and wherein said Ci- 6 alkyl group is a straight or branched hydrocarbon chain consisting solely of carbon and hydrogen atoms, containing no unsaturation, and having from one to six carbon atoms.
  • Ci-6alkyl include, but are not limited to, methyl, ethyl, n-propyl, 1-methylethyl (/so-propyl), n-butyl, n-pentyl and 1 , 1- dimethylethyl (f-butyl).
  • non-solvated refers to a crystalline form of a molecule, atom, and/or ions that does not further comprise molecules of a solvent or solvents incorporated into the crystalline lattice structure.
  • pharmaceutically acceptable salts refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which typically are not biologically or otherwise undesirable.
  • the compounds of the invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
  • excipient refers to a substance that may be an ingredient of the topical pharmaceutical composition of the invention and which is not the active agent.
  • the term “subject” refers to primates (e.g., humans, male or female), dogs, rabbits, guinea pigs, pigs, rats and mice. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human.
  • the term “inhibit”, “inhibition” or “inhibiting” refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
  • treat refers to alleviating or ameliorating the disease or disorder (i.e., slowing or arresting the development of the disease or at least one of the clinical symptoms thereof); or alleviating or ameliorating at least one physical parameter or biomarker associated with the disease or disorder, including those which may not be discernible to the patient.
  • the term “prevent”, “preventing” or “prevention” of any disease or disorder refers to the prophylactic treatment of the disease or disorder; or delaying the onset or progression of the disease or disorder.
  • a subject is "in need of” a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.
  • the term “twice daily” refers to once in the morning and once in the evening.
  • the term “a”, “an”, “the” and similar terms used in the context of the invention are to be construed to cover both the singular and plural unless otherwise indicated herein or clearly contradicted by the context.
  • the invention provides a topical pharmaceutical composition which is an oil-in-water cream comprising up to 0.35 wt.%, based on the total weight of the
  • composition of a compound of formula (I), an emulsifier, and:
  • an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol, and phenylethyl alcohol; or
  • antimicrobial preservative selected from the group of parabens.
  • the invention provides a topical pharmaceutical composition which is an oil-in-water cream comprising up to 0.35 wt.%, based on the total weight of the composition, of a compound of formula (I), an emulsifier, and from 0.8 to 2.3 wt.%, based on the total weight of the composition, of an antimicrobial preservative, selected from the group of benzyl alcohol, phenoxyethanol and phenylethyl alcohol.
  • an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol and phenylethyl alcohol.
  • topical pharmaceutical compositions according to the invention allow the active agent to be formulated into stable compositions and allow for a suitable penetration and permeation profile, favoring the penetration of the active agent into the different skin layers, while limiting permeation through the skin into the systemic circulation.
  • the active agent may be obtained according to the methods described in
  • WO2014072956 Particularly suitable for the inventive compositions as active agent is the compound of formula (I) in crystalline form.
  • the amount of active agent in the inventive composition may vary over a certain range and may be present in an amount by weight of up to 0.35% by weight of the composition of the invention. Suitable amounts for the active agent may be determined by the skilled person in routine experiments; typically they are in the range from 0.05 to 0.35 wt.%, or 0.05 to 0.30 wt.%, preferably from 0.10 to 0.27 wt.%, such as 0.10, 0.15, 0.20 or 0.25 wt.%, more preferably from 0.20 to 0.27 wt.%, such 0.20 or 0.25 wt.%.
  • cream refers to a semisolid dosage form.
  • oil-in-water cream refers to a cream in form of an emulsion, wherein the oil phase is the dispersed phase and the aqueous phase is the continuous phase.
  • Typical excipients in an oil-in-water cream according to the invention are solvents and one or more emulsifier. Additional excipients such one or more excipients may be added. Such excipients may for example be selected from the group of thickening agents, buffers and preservatives.
  • liquid crystal cream refers to a cream that has the potential to form a liquid crystal network where multiple layers of surfactant and water surround an oil droplet.
  • Suitable compositions of the solvent in the context of the invention may be determined by the skilled person.
  • the solvent may be selected from one or more solvents selected from the group of:
  • Fatty acid esters such as fatty acid glycerides; preferred are medium chain
  • the topical pharmaceutical composition of the invention comprises from 10 to 20 wt.% of fatty acid glycerides. In another preferred embodiment, the topical pharmaceutical composition of the invention comprises from 10 to 20 wt.% of medium chain triglycerides, most preferably 14 to 16 wt.% of medium chain triglycerides.
  • Preferred medium chain triglycerides are Miglyol ® 812, Miglyol ® 810 and Crodamol ® GTCC, most preferred is Miglyol ® 812.
  • the topical pharmaceutical composition of the invention contains from 2 to 30 wt.% of solvents with penetration enhancing activity.
  • the topical pharmaceutical composition of the invention comprises from 2 to 30 wt.% of diethylene glycol monoethylether, most preferably 8 to 12 wt.% of diethylene glycol monoethylether.
  • the topical pharmaceutical composition of the invention contains from 2 to 15 wt.% of glycols. In another preferred embodiment, the topical pharmaceutical composition of the invention contains from 2 to 15 wt.% of propylene glycol, most preferably 4 to 6 wt.% of propylene glycol.
  • the topical pharmaceutical composition of the invention contains from 30 to 70 wt.% of water, most preferably 40 to 60 wt.% of water.
  • the solvent of the topical pharmaceutical composition of the invention consists of a mixture of medium chain triglycerides, diethylene glycol
  • the solvent of the topical pharmaceutical composition of the invention consists of a mixture of medium chain triglycerides, diethylene glycol monoethylether, propylene glycol and water wherein the amount of triglycerides is 14 to 16 wt.% based on the total weight of the composition, the amount of diethylene glycol monoethylether is 8 to 12 wt.% based on the total weight of the composition, the amount of propylene glycol is 4 to 6 wt.% based on the total weight of the composition, and water is 40 to 60 wt.% based on the total weight of the composition.
  • An emulsifier is an agent that prevents coalescence of the emulsion. Suitable emulsifiers in the context of the invention may be determined by the skilled person in routine
  • the emulsifier is from 0.05 to 10 wt.%, preferably from 0.2 to 6.0 wt.% based on the total weight of the composition.
  • an emulsifier may be selected from one or more agents selected from the group of:
  • Cs to C20 alkyl ethers such as macrogol 21 stearyl ether (Brij ® S721), macrogol 2 stearyl ether (Brij ® S2), macrogol 20 stearyl ether (Brij ® S20), polyethylene glycol hexadecyl ether (Cetomacrogol ® 1000) and PEG-100 stearyl ether (Myrj ® S100); preferred is macrogol 21 stearyl ether.
  • Brij ® S721 macrogol 21 stearyl ether
  • Brij ® S2 macrogol 2 stearyl ether
  • Brij ® S20 macrogol 20 stearyl ether
  • Myrj ® S100 PEG-100 stearyl ether
  • the topical pharmaceutical composition of the invention contains from 0.2 to 4.0 wt.% of polyoxyl stearyl ethers. In another preferred embodiment, the topical pharmaceutical composition of the invention contains from 0.2 to 4.0 wt.% of macrogol 21 stearyl ether, most preferably 0.7 to 1.5 wt.% of macrogol 21 stearyl ether. b) Glycerol monostearate 40-55. In a preferred embodiment, the topical pharmaceutical composition of the invention contains from 0.8 to 5 wt.% of glycerol monostearate 40- 55, most preferably 1.5 to 2.5 wt.% of glycerol monostearate 40-55.
  • emulsifiers such as anionic surfactants such as sodium alkyl sulfates e.g. sodium cetostearyl sulfate and sodium laurylsulfate, docusate sodium, lauric acid and phospholipids; macrogolglycerol hydroxystearate (Kolliphor ® RH40); apricot kernel oil PEG-6 esters (Labrafil ® M1944CS); polyethylene glycol sorbitan monooleate (Tween ® 80); polyethylene glycol sorbitan monostearate (Tween ® 60); sorbitan stearate (Span ® 60) and lecithin.
  • anionic surfactants such as sodium alkyl sulfates e.g. sodium cetostearyl sulfate and sodium laurylsulfate, docusate sodium, lauric acid and phospholipids; macrogolglycerol hydroxystearate (Kolliphor ® RH40); apricot
  • the topical pharmaceutical composition of the invention contains an emulsifier.
  • the topical pharmaceutical composition of the invention contains an emulsifier, wherein the emulsifier is a mixture of macrogol 21 stearyl ether and glycerol monostearate 40-55.
  • the topical pharmaceutical composition of the invention contains an emulsifier, wherein the emulsifier is a mixture of macrogol 21 stearyl ether and glycerol monostearate 40-55, wherein the amount of macrogol 21 stearyl ether is 0.7 to 1.5 wt.% based on the total weight of the composition, and the amount of glycerol monostearate 40-55 is 1.5 to 2.5 wt.% based on the total weight of the composition.
  • the emulsifier is a mixture of macrogol 21 stearyl ether and glycerol monostearate 40-55, wherein the amount of macrogol 21 stearyl ether is 0.7 to 1.5 wt.% based on the total weight of the composition, and the amount of glycerol monostearate 40-55 is 1.5 to 2.5 wt.% based on the total weight of the composition.
  • a thickening agent is an excipient that adjusts the viscosity of the emulsion.
  • Suitable thickening agents in the context of the invention may be determined by the skilled person in routine experiments; in the context of the invention, the thickening agent is from 4 to 12 wt.%, preferably from 6.0 to 10.0 wt.% based on the total weight of the composition.
  • a thickening agent may be selected from one or more agents selected from the group of: a) Thickening agents which also act as co-emulsifiers, such as fatty alcohols; preferred are (Cio to C25) fatty alcohols.
  • the topical pharmaceutical composition of the invention contains from 4 to 12 wt.% of (C10 to C25) fatty alcohols. In another preferred embodiment, the topical pharmaceutical composition of the invention contains from 6.0 to 10.0 wt.% (C10 to C25) fatty alcohols, more preferably 6.0 to 10.0 wt.% of a mixture of cetyl alcohol and stearyl alcohol, even more preferably, the topical pharmaceutical composition of the invention contains from 3.0 to 5.0 wt.% of stearyl alcohol and from 3.0 to 5.0 wt.% of cetyl alcohol.
  • HPC hydroxypropyl methyl cellulose
  • HPMC hyperromellose
  • HEC hydroxyethyl cellulose
  • carboxymethyl cellulose cellulose gum, CMC
  • carbomers e.g. carbopol ® 910, carbopol ® 934, carbopol ® 934P, carbopol ® 940, carbopol ® 941 and carbopol ® 974; Noveon ® polycarbophil
  • polyvinylalcohol for example Mowiol ® 26-88
  • polyvinylpyrrolidone povidone, PVP
  • polyvinylpolypyrrolidone crospovidone, PVPP
  • xanthan gum sodium hyaluronate and hyaluronic acid (hyaluronan, HA).
  • the topical pharmaceutical composition of the invention contains a thickening agent.
  • the topical pharmaceutical composition of the invention contains a thickening agent, wherein the thickening agent is a mixture of stearyl alcohol and cetyl alcohol.
  • the topical pharmaceutical composition of the invention contains a thickening agent, wherein the thickening agent is a mixture of stearyl alcohol and cetyl alcohol, wherein the amount of stearyl alcohol is 3.0 to 5.0 wt.% based on the total weight of the composition, and the amount of cetyl alcohol is 3.0 to 5.0 wt.% based on the total weight of the composition.
  • a buffering agent serves to maintain the pH of the emulsion within a certain narrow range and comprise mixtures of appropriate amounts of an acid and a base.
  • Suitable buffering agents in the context of the invention may be determined by the skilled person in routine experiments; in the context of the invention, the buffering agent is chosen so that the final pH value of the emulsion is from pH 5.0 to pH 5.8, preferably from pH 5.2 to pH 5.6.
  • a buffering agent may for example be selected from one or more agents selected from the group of citric acid, lactic acid and/or appropriate salts thereof.
  • the topical pharmaceutical composition of the invention contains a buffering agent.
  • the topical pharmaceutical composition of the invention contains a buffering agent, wherein the buffering agent is a citric acid / citrate buffer.
  • the topical pharmaceutical composition of the invention contains a buffering agent, wherein the buffering agent is a citric acid / citrate buffer, wherein the amount of citric acid / citrate is the amount necessary to ensure that the final pH value of the emulsion is maintained from pH 5.0 to pH 5.8, preferably from pH 5.2 to pH 5.6, to avoid the increased formation of degradation products.
  • An antimicrobial preservative serves to prevent bacterial growth, mold growth, fermentation, oxidation, or any combination thereof of the emulsion.
  • the topical pharmaceutical composition of the invention contains a suitable antimicrobial preservative.
  • suitable antimicrobial preservatives in the context of the present invention are antimicrobial preservatives that lead to formulations which are stable in terms of chemical and physical stability and are suitable for use with sunlight exposure for example benzyl alcohol, phenoxyethanol and phenylethyl alcohol.
  • the amount of preservative that is suitable may depend on the nature of the antimicrobial preservative.
  • the topical pharmaceutical composition of the invention contains from 0.8 to 2.3 wt.% of an antimicrobial preservative, selected from the group of benzyl alcohol, phenoxyethanol and phenylethyl alcohol. In a more preferred embodiment, the topical pharmaceutical composition of the invention contains from 0.8 to 2.3 wt.% of benzyl alcohol as an antimicrobial preservative, in an even more preferred embodiment, the topical pharmaceutical composition of the invention contains from 0.8 to 1.0 wt.% of benzyl alcohol as an antimicrobial preservative, in a most preferred embodiment, the topical pharmaceutical composition of the invention contains 1.0 wt.% of benzyl alcohol as a preservative.
  • topical pharmaceutical composition of the invention consists of:
  • an emulsifier and:
  • an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol, and phenylethyl alcohol; or
  • antimicrobial preservative selected from the group of parabens.
  • a topical pharmaceutical composition according to embodiment A1 comprising from 0.05 to 0.35 wt.% of a compound of formula (I).
  • an antimicrobial preservative selected from the group of parabens.
  • a topical pharmaceutical composition according to embodiment A1 comprising from
  • an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol and phenylethyl alcohol.
  • a topical pharmaceutical composition according to embodiment A2 comprising from 0.8 to 2.3 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol and phenylethyl alcohol.
  • a topical pharmaceutical composition according to any one of embodiments A8 to A10, wherein the medium chain triglycerides are selected from Miglyol ® 812, Miglyol ® 810 and Crodamol ® GTCC.
  • A12 A topical pharmaceutical composition according to embodiment A1 1 , wherein the medium chain triglycerides are Miglyol ® 812.
  • a topical pharmaceutical composition according to embodiment A14 wherein the content of solvents with penetration enhancing activity are from 8 to 12 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A15 wherein the content of solvents with penetration enhancing activity are 10 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A18 wherein the content of glycols are from 2 to 15 wt.% based on the total weight of the composition.
  • A20 A topical pharmaceutical composition according to embodiment A19, wherein the content of glycols are from 4 to 6 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A20 wherein the content of glycols are 5 wt.% based on the total weight of the composition.
  • A22) A topical pharmaceutical composition according to any one of embodiments A18 to A21 , wherein the glycols are propylene glycol.
  • A23) A topical pharmaceutical composition according to any one of embodiments A1 to A22, wherein the content of water is from 30 to 70 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A22 wherein the content of water is from 40 to 60 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to any one of embodiments A1 to A25, wherein the content of the emulsifier is from 0.05 to 10 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A26 wherein the content of the emulsifier is from 0.2 to 6.0 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A28 wherein the polyoxyethylene (Cs to C20) alkyl ethers are selected from one or more members of the group consisting of macrogol 21 stearyl ether, macrogol 2 stearyl ether, macrogol 20 stearyl ether, polyethylene glycol hexadecyl ether and PEG- 100 stearyl ether.
  • the polyoxyethylene (Cs to C20) alkyl ethers are selected from one or more members of the group consisting of macrogol 21 stearyl ether, macrogol 2 stearyl ether, macrogol 20 stearyl ether, polyethylene glycol hexadecyl ether and PEG- 100 stearyl ether.
  • A30) A topical pharmaceutical composition according to embodiment A29, wherein the polyoxyethylene (Cs to C20) alkyl ethers is macrogol 21 stearyl ether.
  • a topical pharmaceutical composition according to any one of embodiments A28 to A30, wherein the emulsifier is a mixture of macrogol 21 stearyl ether and glycerol monostearate 40-55.
  • A32 A topical pharmaceutical composition according to embodiment A31 , wherein the content of macrogol 21 stearyl ether is from 0.7 to 1.5 wt.% based on the total weight of the composition, and the content of glycerol monostearate 40-55 is from 1.5 to 2.5 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A32 wherein the content of macrogol 21 stearyl ether is from 0.9 to 1.3 wt.% based on the total weight of the composition, and the content of glycerol monostearate 40-55 is 2.0 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A34 comprising a thickening agent which also act as co-emulsifier.
  • A37 A topical pharmaceutical composition according to embodiment A36, wherein the content of (C10 to C25) fatty alcohols is 4 to 12 wt.% based on the total weight of the composition.
  • A38 A topical pharmaceutical composition according to embodiment A37, wherein the content of (C1 0 to C25) fatty alcohols is 6.0 to 10.0 wt.% based on the total weight of the composition.
  • A39) A topical pharmaceutical composition according to any one of embodiments A36 to A38, wherein the (C1 0 to C25) fatty alcohols are a mixture of cetyl alcohol and stearyl alcohol.
  • A40) A topical pharmaceutical composition according to embodiment A39, wherein the content of cetyl alcohol is from 3.0 to 5.0 wt.%, based on the total weight of the composition, and the content of stearyl alcohol is from 3.0 to 5.0 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A40 wherein the content of cetyl alcohol is 4.0 wt.%, based on the total weight of the composition, and the content of stearyl alcohol is 4.0 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to any one of embodiments A1 to A41 comprising a buffering agent.
  • a topical pharmaceutical composition according to any one of embodiments A42 to A45, wherein the buffering agent is a citric acid / citrate buffer.
  • a topical pharmaceutical composition according to any one of embodiments A1 to A46, wherein the content of the antimicrobial preservative is from 0.8 to 1.0 wt.% based on the total weight of the composition.
  • A49) A topical pharmaceutical composition according to embodiment A48, wherein the content of benzyl alcohol is 1.0 wt.% based on the total weight of the composition.
  • A50) A topical pharmaceutical composition according to any one of embodiments A1 to A49, wherein the content of compound of formula (I) is from up to 0.30 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A50 wherein the content of compound of formula (I) is from 0.05 to 0.30 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A51 wherein the content of compound of formula (I) is from 0.10 to 0.27 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A52 wherein the content of compound of formula (I) is from 0.20 to 0.27 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition according to embodiment A53, wherein the content of compound of formula (I) is 0.25 wt.% based on the total weight of the composition.
  • a topical pharmaceutical composition consisting of:
  • a topical pharmaceutical composition consisting of Medium chain triglycerides (Miglyol ® 15 812)
  • a topical pharmaceutical composition consisting of:
  • a topical pharmaceutical composition consisting of [wt.%]
  • a topical pharmaceutical composition consisting of
  • Purified water 56.99 A60 A process to prepare the topical pharmaceutical composition according to any one of embodiments A1 to A59, comprising the steps of
  • step (c) Adding the solution/suspension of step (a) to the melt of step (b) to obtain a melt containing the compound of formula (I);
  • step (e) Mixing the melt of step (c) with the aqueous phase of step (d) and cooling the mixture down to obtain the oil-in-water cream.
  • step (a) A process according to embodiment A60, wherein in step (a) the compound of formula (I) is dissolved/suspended in solvents with penetration enhancing activity.
  • step (b) A process according to embodiments A60 or A61 , wherein in step (b) the melt is prepared from fatty acid glycerides, glycols, an emulsifier, an antimicrobial preservative and a thickening agent.
  • step (d) A process according to any one of embodiments A60 to A62, wherein in step (d) the aqueous phase is an aqueous buffer solution of pH 5.2 - 5.6.
  • A64 A process according to any one of embodiments A60 to A63, wherein the compound of formula (I) used in step (a) is in crystalline form according to embodiments B1 to B6.
  • compositions according to any one of embodiments A1 to A59 or A65 in the treatment or prevention of a disease or disorder selected from any one of Lists A to K.
  • composition according to any one of embodiments A1 to A59 or A65 in the manufacture of a medicament for the treatment or prevention of a disease or disorder selected from any one of Lists A to K.
  • A69 A method of treating a disease or disorder selected from any one of Lists A to K in a subject, comprising the step of administering to the subject a therapeutically effective amount of a composition according to any one of embodiments A1 to A59 or A65.
  • A70 A method of preventing a disease or disorder selected from any one of Lists A to K in a subject, comprising the step of administering to the subject a therapeutically effective amount of a composition according to any one of embodiments A1 to A59 or A65.
  • composition according to any one of embodiments A1 to A59 or A65 in the treatment of cutaneous squamous cell carcinoma in situ (cSCCis).
  • composition according to any one of embodiments A1 to A59 or A65 in the manufacture of a medicament for the treatment of cutaneous squamous cell carcinoma in situ (cSCCis).
  • A74 A method of treating cutaneous squamous cell carcinoma in situ (cSCCis) in a subject, comprising the step of administering to the subject a therapeutically effective amount of a composition according to any one of embodiments A1 to A59 or A65.
  • a composition for use according to embodiment A71 wherein the application of the composition is twice daily.
  • A77 Use according to embodiment A73, wherein the medicament is applied twice daily.
  • A78 A method according to embodiment A74, wherein the composition is applied twice daily.
  • a combination comprising a therapeutically effective amount of a crystalline form according to any one of embodiments B1 to B6, and one or more therapeutically active agents.
  • a method of treating a disease or disorder selected from any one of Lists A to K in a subject comprising the step of administering to the subject a therapeutically effective amount of a form according to any one of embodiments B1 to B6.
  • a method of preventing a disease or disorder selected from any one of Lists A to K in a subject comprising the step of administering to the subject a therapeutically effective amount of a form according to any one of embodiments B1 to B6.
  • B16 Use of crystalline form according to any one of embodiments B1 to B6 in the manufacture of a medicament for the treatment of cutaneous squamous cell carcinoma in situ (cSCCis).
  • B17 A method of treating cutaneous squamous cell carcinoma in situ (cSCCis) in a subject, comprising the step of administering to the subject a therapeutically effective amount of a form according to any one of embodiments B1 to B6.
  • Non-melanoma skin cancers such as basal cell carcinoma and squamous cell carcinoma (SCC); their pre-malignant stages such as squamous cell carcinoma in situ (SCCis), actinic keratosis (AK), solar keratosis and chronically sun damaged skin; and other hyperproliferative skin disorders caused by dysregulation of skin fibroblasts such as skin fibrosis, scleroderma, hypertrophic scars and keloids.
  • List F Hyperproliferative skin disorders caused by dysregulation of skin fibroblasts; other than non-melanoma skin cancers and their pre-malignant stages.
  • Step time > 40 s for Lynxeye, > 100 s for Vantec detector
  • Mass spectrometer Triple Quad 5500 Applied Biosystems (TS-013-819) MS conditions Turbo Spray, positive ion mode.
  • Citric acid anhydrous and sodium citrate dehydrate were added to the water and heated to 70°C under stirring, until a clear solution was obtained.
  • Table 1 Pharmaceutical compositions, oil-in-water cream
  • Xanthan gum was dispersed in propylene glycol to form xanthan gum premix. This xanthan gum premix was then transferred into water and carbopol 974P (which was pre- stirred at 25°C) was added.
  • step 2 The mixture from step 1 was stirred at 75°C until a homogenous gel was formed.
  • step 4 The mixture of step 4 was then stirred at 75°C until clear solution was formed.
  • step 5 was then added to the gel obtained in step 2, under stirring and homogenization at 75°C.
  • step 6 The homogenized mixture of step 6 was allowed to cool to 40°C
  • the compound of formula (I) showed some tendency to degradation when exposed to excipients providing oxidative potential such as PEG and oleylalcohol.
  • Medium chain triglycerides demonstrated exceptionally good compatibility with the compound of formula (I).
  • the standard preclinical in vitro assay for phototoxicity assessment is the "in vitro 3T3 neutral red uptake (NRU) phototoxicity test” (OECD (2004), in vitro 3T3 NRU phototoxicity test.
  • the compound of formula (I) showed a photoirritation factor (PI F) of 7 in this test.
  • the local lymph node assay assesses the photo-sensitization potential of compounds and compositions.
  • the compositions were repeatedly dosed to mice which were then exposed to UV-Vis radiation with a light dose of at least 10 J/cm 2 . Lymph node weight, ear weight and lymph node cell count were then analysed.
  • compositions as prepared above, in 10 g aluminum tubes, under inverse storage were tested for stability. Testing was performed at 5 °C / ambient RH, 25 °C / 60 %RH, 40 °C / 75 %RH and other conditions as indicated.
  • Impurities at RRT 0.66, RRT 1 .08 and RRT 1 .37 are by products from drug substance, not included in total impurities.
  • composition Var B showed good stability data at 5°C, at long term condition of
  • the pharmaceutical composition Var B clearly shows a better stability in terms of
  • compositions of the present invention were tested for penetration into, as well as permeation through dermatomized healthy human skin in the static Franz cells for the compound of formula (I).
  • the integrity of skin was determined in each Franz Cell by evaluating the permeation of tritiated water (HOT) through the skin; after pre-equilibration, 500 of HOT (approx. 0.1 MBq/mL) were applied on the surface of the skin fixed on the cells. After 30 min of equilibration, the HOT was removed from the skin with cotton tips; 3.5 mL of the receptor phase (composition of the receptor fluid is described below) were sampled in order to measure the fraction of HOT, which permeated across the skin. Radioactivity in the receptor phase was measured using the Liquid Scintillation Systems Tri-Carb 3110 (Perkin Elmer Instr. Co., Meriden, CT, USA). For quench correction an external standard method was used. Quench correction curves were established by means of sealed standards (Perkin Elmer Instruments).
  • the skin was used as a membrane separating the donor and receptor chambers of the static Franz diffusion cells ( ⁇ 7 mL volume, -1.5 cm diameter) assembled in the Logan ® system.
  • the receptor fluid contained 100 U/mL of a 1 % penicillin/streptomycin mixture (v/v) to prevent microbiological contamination.
  • the effective skin area for diffusion was 1.77 cm 2 and the volumes of receiver compartment were in the range of 6.33 to 7.00 ml_, respectively.
  • the temperature of the cells was kept constant using a circulating water bath at 32 ⁇ 1 °C. Magnetic stirrer bars were constantly used during the entire experiment to ensure receptor uniformity.
  • test compositions nominal weight of 300 mg were applied as a single dose on the skin samples mounted on the Franz diffusion cells (corresponding to a sampling time of 0 h). The compositions were left on the skin for 24 h or 48 h.
  • the donor compartments of Franz diffusion cells were semi-occluded with parafilm (Parafilm' 8 M) with holes.
  • the permeation rate (flux) of the compound of formula (I) through the skin is indicative for a low systemic exposure in vivo.
  • composition was applied once daily to 10% of the body surface area over 2 weeks and skin was washed 6 h after each application under occlusion.
  • pipetted 50 ⁇ _ of plasma sample Standards, QCs, Blank, Zero and incurred samples.
  • a and B skin biopsies from the application (skin treated with composition containing test item (not the placebo cream)) were collected at necropsy from each animal. Skin samples were placed in FastPrep tubes and immediately frozen on dry ice and stored at -65°C or below until bioanalysis.
  • Skin tissue was put into a Lysing Matrix D tube (for 2 ml_). An adequate volume of acetonitrile was added to obtain a final dilution of 5 (1 g of tissue is considered to be equal to 1 ml_; 1 ml_ of acetonitrile was considered to be equal to 1 g).
  • the skin tissue samples were homogenized at 4.0 M/s for 20s (2 cycles) using Fast Prep homogenizer and then
  • Plasma samples were extracted using protein precipitation and analyzed by LC-MS/MS using ElectroSpray Ionization (ESI) in positive ion mode according LC-MS/MS-method 2. Skin samples were homogenized using Fast Prep with acetonitrile and analyzed by LC- MS/MS using ElectroSpray Ionization (ESI) in positive ion mode according LC-MS/MS- method 3.
  • ESI ElectroSpray Ionization
  • the crystalline form is not solvated, slightly hygroscopic and characterized by a melting at 184°C with an enthalpy of 66.5 J/g, and a glass transition temperature at around 89-97°C.
  • the 1 H NMR conformed to the one of the compound of formula (I) in amorphous form.
  • LCMS (method 4): 507.1 [M-H], 509.1 [M+H].
  • Table 7.1 List of most significant 2-Theta peaks from X-ray Powder Diffraction Pattern with tolerances ⁇ 0.5 of compound of formula (I) in crystalline form (including low/weak peaks for information). 2-Theta in degree Intensity [%]
  • Thermodynamic solubility was determined by the shake flask method. Compound of formula (I) was weighed precisely into a glass vial. Medium was added and the sample was kept stirred using a magnetic stirrer for 24h at ambient conditions. After 24h excess compound of formula (I) was removed by filtration using 0.4micron PVDC filter tube. The content of the solution was quantified by HPLC (HPLC-method 1).
  • Thermodynamic solubility was determined by the shake flask method. Compound of formula (I) was weighed precisely into a glass vial. Medium was added and the sample was kept stirred using a magnetic stirrer for 24h at ambient conditions. After 24h excess compound of formula (I) was removed by filtration using 0.4micron PVDC filter tube. The content of the solution was quantified by HPLC (HPLC-method 1).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Dermatology (AREA)
  • Dispersion Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to novel topical pharmaceutical compositions comprising an oxazolidin-2-one-pyrimidine compound of formula (I), a method of manufacturing such compositions, as well as uses of such compositions. The present invention also relates to a novel crystalline form of the compound of formula (I).

Description

Formulation Comprising Oxazolidin-2-one-Pyrimidine Derivative
FIELD OF THE INVENTION
The present invention relates to novel topical pharmaceutical compositi
comprising an oxazolidin-2-one- rimidine compound of formula (I)
Figure imgf000002_0001
a method of manufacturing such compositions as well as uses of such compositions.
The present invention also relates to a novel crystalline form of the compound of formula (I). BACKGROUND OF THE INVENTION
The compound of formula (I) is useful for the treatment or prevention of a disease or disorder selected from non-melanoma skin cancers such as basal cell carcinoma and squamous cell carcinoma (SCC); their pre-malignant stages such as squamous cell carcinoma in situ (SCCis), actinic keratosis (AK), solar keratosis and chronically sun damaged skin; and other hyperproliferative skin disorders caused by dysregulation of skin fibroblasts such as skin fibrosis, scleroderma, hypertrophic scars and keloids.
The compound of formula (I) is described in international patent application
WO2014072956.
It is desirable to identify compositions, and uses of these compositions that may improve efficacy, bioavailability, stability, and/or acceptance by the patient and that may reduce potential side effects.
It is furthermore desirable to identify novel, more stable forms of the compound of formula (I).
These objectives are achieved by providing a composition as described herein; by providing the composition for use in diseases, particularly for the treatment of dermatological diseases as described herein, and by providing a process to produce the composition as described herein, as well as by providing the compound of formula (I) in crystalline form. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 : x-ray powder diffraction (XRPD) spectrum of the compound of formula (I) in crystalline form. Figure 2: Differential scanning calorimetry (DSC) curve of the compound of formula (I) in crystalline form.
Figure 3: Thermogravimetric analysis (TGA) diagram of the compound of formula (I) in crystalline form.
Figure 4: FT-IR spectrum of the compound of formula (I) in crystalline form.
Figure 5: Scanning electron microscope (SEM) picture of the compound of formula (I) in crystalline form.
DETAILED DESCRIPTION OF THE INVENTION
Due to its limited solubility, the compound of formula (I) presents specific difficulties in relation to topical galenic compositions, since solubility is an important factor influencing drug loading.
In addition, other challenges, in particular with respect to photo-stability and stability towards oxidation in contact with excipients used for topical compositions, have to be addressed. For example, the amorphous compound of formula (I) showed some tendency to light induced degradation; 8% degradation was observed in the photostability assay described in WO2014072956.
In addition, the amorphous compound of formula (I) showed a weak positive signal in the in vitro phototoxicity assay (3T3) with a photoirritation factor (PIF) of 7, indicating an increased phototoxicity potential.
In accordance with the invention it has been found that a stable pharmaceutical composition comprising the compound of formula (I) with a suitable drug loading can be obtained and which is suitable for use with sunlight exposure. Consequently, the risks of undesirable photosensitizing side effects are diminished by a composition according to the invention.
Several other types of topical compositions have been investigated, some of which are included in the example section as reference examples for comparison.
In accordance with the invention it has also been found that the compound of formula (I) can be obtained in crystalline form, which shows an enhanced photostability compared to the compound of formula (I) in amorphous form known from WO2014072956.
Terms used in the specification have the following meanings:
As used herein "active agent" refers to a compound of formula (I)
Figure imgf000004_0001
"Active agent" is also intended to represent amorphous and crystalline forms such as polymorphs. "Active agent" is also intended to represent a solvate thereof, a pharmaceutical acceptable salt thereof and its mixtures. "Active agent" is also intended to represent material exhibiting specific solid state properties such as specific crystal shapes and/or milled forms of the "Active agent", e.g. in micronized form.
As used herein "parabens" refers to compounds of the following formula
Figure imgf000004_0002
wherein R represents a Ci-6alkyl group and wherein said Ci-6alkyl group is a straight or branched hydrocarbon chain consisting solely of carbon and hydrogen atoms, containing no unsaturation, and having from one to six carbon atoms. Examples of Ci-6alkyl include, but are not limited to, methyl, ethyl, n-propyl, 1-methylethyl (/so-propyl), n-butyl, n-pentyl and 1 , 1- dimethylethyl (f-butyl).
As used herein "non-solvated" refers to a crystalline form of a molecule, atom, and/or ions that does not further comprise molecules of a solvent or solvents incorporated into the crystalline lattice structure.
As used herein, "pharmaceutically acceptable salts" refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which typically are not biologically or otherwise undesirable. In many cases, the compounds of the invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
As used herein, "excipient" refers to a substance that may be an ingredient of the topical pharmaceutical composition of the invention and which is not the active agent.
As used herein, the term "subject" refers to primates (e.g., humans, male or female), dogs, rabbits, guinea pigs, pigs, rats and mice. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human. As used herein, the term "inhibit", "inhibition" or "inhibiting" refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
As used herein, the term "treat", "treating" or "treatment" of any disease or disorder refers to alleviating or ameliorating the disease or disorder (i.e., slowing or arresting the development of the disease or at least one of the clinical symptoms thereof); or alleviating or ameliorating at least one physical parameter or biomarker associated with the disease or disorder, including those which may not be discernible to the patient.
As used herein, the term "prevent", "preventing" or "prevention" of any disease or disorder refers to the prophylactic treatment of the disease or disorder; or delaying the onset or progression of the disease or disorder.
As used herein, a subject is "in need of" a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.
As used herein, the term "twice daily" refers to once in the morning and once in the evening. As used herein, the term "a", "an", "the" and similar terms used in the context of the invention (especially in the context of the claims) are to be construed to cover both the singular and plural unless otherwise indicated herein or clearly contradicted by the context.
Throughout this specification and in the claims that follow, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising" as well as the word "contain", or variations such as "contains" or "containing", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
It is further understood that the various embodiments, preferences and ranges of this invention, as provided / disclosed in the specification and claims may be combined with other specified features to provide further embodiments.
In a first aspect, the invention provides a topical pharmaceutical composition which is an oil-in-water cream comprising up to 0.35 wt.%, based on the total weight of the
composition, of a compound of formula (I), an emulsifier, and:
from 0.4 to 2.9 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol, and phenylethyl alcohol; or
from 0.01 to 0.6 wt.%, based on the total weight of the composition, of an
antimicrobial preservative selected from the group of parabens.
In one embodiment of the first aspect, the invention provides a topical pharmaceutical composition which is an oil-in-water cream comprising up to 0.35 wt.%, based on the total weight of the composition, of a compound of formula (I), an emulsifier, and from 0.8 to 2.3 wt.%, based on the total weight of the composition, of an antimicrobial preservative, selected from the group of benzyl alcohol, phenoxyethanol and phenylethyl alcohol.
It was found that topical pharmaceutical compositions according to the invention allow the active agent to be formulated into stable compositions and allow for a suitable penetration and permeation profile, favoring the penetration of the active agent into the different skin layers, while limiting permeation through the skin into the systemic circulation.
The active agent may be obtained according to the methods described in
WO2014072956. Particularly suitable for the inventive compositions as active agent is the compound of formula (I) in crystalline form. The amount of active agent in the inventive composition may vary over a certain range and may be present in an amount by weight of up to 0.35% by weight of the composition of the invention. Suitable amounts for the active agent may be determined by the skilled person in routine experiments; typically they are in the range from 0.05 to 0.35 wt.%, or 0.05 to 0.30 wt.%, preferably from 0.10 to 0.27 wt.%, such as 0.10, 0.15, 0.20 or 0.25 wt.%, more preferably from 0.20 to 0.27 wt.%, such 0.20 or 0.25 wt.%.
As used herein "cream" refers to a semisolid dosage form.
As used herein, "oil-in-water cream" refers to a cream in form of an emulsion, wherein the oil phase is the dispersed phase and the aqueous phase is the continuous phase.
Typical excipients in an oil-in-water cream according to the invention are solvents and one or more emulsifier. Additional excipients such one or more excipients may be added. Such excipients may for example be selected from the group of thickening agents, buffers and preservatives.
As used herein, "liquid crystal cream" refers to a cream that has the potential to form a liquid crystal network where multiple layers of surfactant and water surround an oil droplet.
Suitable compositions of the solvent in the context of the invention may be determined by the skilled person. The solvent may be selected from one or more solvents selected from the group of:
a) Fatty acid esters such as fatty acid glycerides; preferred are medium chain
triglycerides. In a preferred embodiment, the topical pharmaceutical composition of the invention comprises from 10 to 20 wt.% of fatty acid glycerides. In another preferred embodiment, the topical pharmaceutical composition of the invention comprises from 10 to 20 wt.% of medium chain triglycerides, most preferably 14 to 16 wt.% of medium chain triglycerides. Preferred medium chain triglycerides are Miglyol® 812, Miglyol® 810 and Crodamol® GTCC, most preferred is Miglyol® 812.
b) Solvents with penetration enhancing activity, such as diethylene glycol
monoethylether (Transcutol®) or oleyl alcohol. In a preferred embodiment, the topical pharmaceutical composition of the invention contains from 2 to 30 wt.% of solvents with penetration enhancing activity. In a preferred embodiment, the topical pharmaceutical composition of the invention comprises from 2 to 30 wt.% of diethylene glycol monoethylether, most preferably 8 to 12 wt.% of diethylene glycol monoethylether.
c) Glycols or triols, such as propylene glycol, dipropylene glycol, butylene glycol or glycerol or alcohols such as ethanol or isopropanol; preferred is propylene glycol. In a preferred embodiment, the topical pharmaceutical composition of the invention contains from 2 to 15 wt.% of glycols. In another preferred embodiment, the topical pharmaceutical composition of the invention contains from 2 to 15 wt.% of propylene glycol, most preferably 4 to 6 wt.% of propylene glycol.
d) Water, preferably purified or distilled water. In a preferred embodiment, the topical pharmaceutical composition of the invention contains from 30 to 70 wt.% of water, most preferably 40 to 60 wt.% of water.
In a preferred embodiment, the solvent of the topical pharmaceutical composition of the invention consists of a mixture of medium chain triglycerides, diethylene glycol
monoethylether, propylene glycol and water. In a more preferred embodiment, the solvent of the topical pharmaceutical composition of the invention consists of a mixture of medium chain triglycerides, diethylene glycol monoethylether, propylene glycol and water wherein the amount of triglycerides is 14 to 16 wt.% based on the total weight of the composition, the amount of diethylene glycol monoethylether is 8 to 12 wt.% based on the total weight of the composition, the amount of propylene glycol is 4 to 6 wt.% based on the total weight of the composition, and water is 40 to 60 wt.% based on the total weight of the composition.
An emulsifier is an agent that prevents coalescence of the emulsion. Suitable emulsifiers in the context of the invention may be determined by the skilled person in routine
experiments; in the context of the invention, the emulsifier is from 0.05 to 10 wt.%, preferably from 0.2 to 6.0 wt.% based on the total weight of the composition. As used herein an emulsifier may be selected from one or more agents selected from the group of:
a) Polyoxyethylene (Cs to C20) alkyl ethers such as macrogol 21 stearyl ether (Brij® S721), macrogol 2 stearyl ether (Brij® S2), macrogol 20 stearyl ether (Brij® S20), polyethylene glycol hexadecyl ether (Cetomacrogol® 1000) and PEG-100 stearyl ether (Myrj® S100); preferred is macrogol 21 stearyl ether. In a preferred
embodiment, the topical pharmaceutical composition of the invention contains from 0.2 to 4.0 wt.% of polyoxyl stearyl ethers. In another preferred embodiment, the topical pharmaceutical composition of the invention contains from 0.2 to 4.0 wt.% of macrogol 21 stearyl ether, most preferably 0.7 to 1.5 wt.% of macrogol 21 stearyl ether. b) Glycerol monostearate 40-55. In a preferred embodiment, the topical pharmaceutical composition of the invention contains from 0.8 to 5 wt.% of glycerol monostearate 40- 55, most preferably 1.5 to 2.5 wt.% of glycerol monostearate 40-55.
c) Other emulsifiers, such as anionic surfactants such as sodium alkyl sulfates e.g. sodium cetostearyl sulfate and sodium laurylsulfate, docusate sodium, lauric acid and phospholipids; macrogolglycerol hydroxystearate (Kolliphor® RH40); apricot kernel oil PEG-6 esters (Labrafil® M1944CS); polyethylene glycol sorbitan monooleate (Tween® 80); polyethylene glycol sorbitan monostearate (Tween® 60); sorbitan stearate (Span® 60) and lecithin.
In a preferred embodiment, the topical pharmaceutical composition of the invention contains an emulsifier. In another preferred embodiment, the topical pharmaceutical composition of the invention contains an emulsifier, wherein the emulsifier is a mixture of macrogol 21 stearyl ether and glycerol monostearate 40-55. In a more preferred embodiment, the topical pharmaceutical composition of the invention contains an emulsifier, wherein the emulsifier is a mixture of macrogol 21 stearyl ether and glycerol monostearate 40-55, wherein the amount of macrogol 21 stearyl ether is 0.7 to 1.5 wt.% based on the total weight of the composition, and the amount of glycerol monostearate 40-55 is 1.5 to 2.5 wt.% based on the total weight of the composition.
A thickening agent is an excipient that adjusts the viscosity of the emulsion. Suitable thickening agents in the context of the invention may be determined by the skilled person in routine experiments; in the context of the invention, the thickening agent is from 4 to 12 wt.%, preferably from 6.0 to 10.0 wt.% based on the total weight of the composition. As used herein a thickening agent may be selected from one or more agents selected from the group of: a) Thickening agents which also act as co-emulsifiers, such as fatty alcohols; preferred are (Cio to C25) fatty alcohols. In a preferred embodiment, the topical pharmaceutical composition of the invention contains from 4 to 12 wt.% of (C10 to C25) fatty alcohols. In another preferred embodiment, the topical pharmaceutical composition of the invention contains from 6.0 to 10.0 wt.% (C10 to C25) fatty alcohols, more preferably 6.0 to 10.0 wt.% of a mixture of cetyl alcohol and stearyl alcohol, even more preferably, the topical pharmaceutical composition of the invention contains from 3.0 to 5.0 wt.% of stearyl alcohol and from 3.0 to 5.0 wt.% of cetyl alcohol.
b) Other thickening agents, such as cellulose for example hydroxypropyl cellulose
(HPC), hydroxypropyl methyl cellulose (hypromellose, HPMC), hydroxyethyl cellulose (HEC) or carboxymethyl cellulose (cellulose gum, CMC); carbomers e.g. carbopol® 910, carbopol® 934, carbopol® 934P, carbopol® 940, carbopol® 941 and carbopol® 974; Noveon® polycarbophil; polyvinylalcohol for example Mowiol® 26-88; polyvinylpyrrolidone (povidone, PVP); polyvinylpolypyrrolidone (crospovidone, PVPP); xanthan gum; sodium hyaluronate and hyaluronic acid (hyaluronan, HA).
In a preferred embodiment, the topical pharmaceutical composition of the invention contains a thickening agent. In another preferred embodiment, the topical pharmaceutical composition of the invention contains a thickening agent, wherein the thickening agent is a mixture of stearyl alcohol and cetyl alcohol. In a more preferred embodiment, the topical pharmaceutical composition of the invention contains a thickening agent, wherein the thickening agent is a mixture of stearyl alcohol and cetyl alcohol, wherein the amount of stearyl alcohol is 3.0 to 5.0 wt.% based on the total weight of the composition, and the amount of cetyl alcohol is 3.0 to 5.0 wt.% based on the total weight of the composition.
A buffering agent serves to maintain the pH of the emulsion within a certain narrow range and comprise mixtures of appropriate amounts of an acid and a base. Suitable buffering agents in the context of the invention may be determined by the skilled person in routine experiments; in the context of the invention, the buffering agent is chosen so that the final pH value of the emulsion is from pH 5.0 to pH 5.8, preferably from pH 5.2 to pH 5.6. As used herein a buffering agent may for example be selected from one or more agents selected from the group of citric acid, lactic acid and/or appropriate salts thereof.
In a preferred embodiment, the topical pharmaceutical composition of the invention contains a buffering agent. In another preferred embodiment, the topical pharmaceutical composition of the invention contains a buffering agent, wherein the buffering agent is a citric acid / citrate buffer. In a more preferred embodiment, the topical pharmaceutical composition of the invention contains a buffering agent, wherein the buffering agent is a citric acid / citrate buffer, wherein the amount of citric acid / citrate is the amount necessary to ensure that the final pH value of the emulsion is maintained from pH 5.0 to pH 5.8, preferably from pH 5.2 to pH 5.6, to avoid the increased formation of degradation products.
An antimicrobial preservative serves to prevent bacterial growth, mold growth, fermentation, oxidation, or any combination thereof of the emulsion.
The topical pharmaceutical composition of the invention contains a suitable antimicrobial preservative. Suitable antimicrobial preservatives in the context of the present invention are antimicrobial preservatives that lead to formulations which are stable in terms of chemical and physical stability and are suitable for use with sunlight exposure for example benzyl alcohol, phenoxyethanol and phenylethyl alcohol. The amount of preservative that is suitable may depend on the nature of the antimicrobial preservative.
In a one embodiment, the topical pharmaceutical composition of the invention contains from 0.8 to 2.3 wt.% of an antimicrobial preservative, selected from the group of benzyl alcohol, phenoxyethanol and phenylethyl alcohol. In a more preferred embodiment, the topical pharmaceutical composition of the invention contains from 0.8 to 2.3 wt.% of benzyl alcohol as an antimicrobial preservative, in an even more preferred embodiment, the topical pharmaceutical composition of the invention contains from 0.8 to 1.0 wt.% of benzyl alcohol as an antimicrobial preservative, in a most preferred embodiment, the topical pharmaceutical composition of the invention contains 1.0 wt.% of benzyl alcohol as a preservative.
In a preferred embodiment, the topical pharmaceutical composition of the invention consists of:
Figure imgf000010_0001
The following enumerated embodiments of the invention are described herein. It will be recognized that features specified in each enumerated embodiment may be combined with other specified features to provide further embodiments of the invention.
A1) A topical pharmaceutical composition in the form of an oil-in-water cream
comprising up to 0.35 wt.%, based on the total weight of the composition, of a compound of formula (I)
Figure imgf000011_0001
an emulsifier, and:
from 0.4 to 2.9 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol, and phenylethyl alcohol; or
from 0.01 to 0.6 wt.%, based on the total weight of the composition, of an
antimicrobial preservative selected from the group of parabens.
A2) A topical pharmaceutical composition according to embodiment A1 , comprising from 0.05 to 0.35 wt.% of a compound of formula (I).
A3) A topical pharmaceutical composition according to embodiment A1 or A2, comprising from 0.8 to 2.3 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol, and phenylethyl alcohol; or
from 0.05 to 0.3 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of parabens.
A4) A topical pharmaceutical composition according to embodiment A1 , comprising from
0.8 to 2.3 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol and phenylethyl alcohol.
A5) A topical pharmaceutical composition according to embodiment A2, comprising from 0.8 to 2.3 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol and phenylethyl alcohol.
A6) A topical pharmaceutical composition according to any one of embodiments A1 to A5, comprising fatty acid glycerides.
A7) A topical pharmaceutical composition according to embodiment A6, wherein the content of fatty acid glycerides is from 10 to 20 wt.% based on the total weight of the composition.
A8) A topical pharmaceutical composition according to embodiment A6 or A7, wherein the fatty acid glycerides are medium chain triglycerides. A9) A topical pharmaceutical composition according to embodiment A8, wherein the content of medium chain triglycerides is from 14 to 16 wt.% based on the total weight of the composition.
A10) A topical pharmaceutical composition according to embodiment A9, wherein the content of medium chain triglycerides is 15 wt.% based on the total weight of the
composition.
A1 1) A topical pharmaceutical composition according to any one of embodiments A8 to A10, wherein the medium chain triglycerides are selected from Miglyol® 812, Miglyol® 810 and Crodamol® GTCC.
A12) A topical pharmaceutical composition according to embodiment A1 1 , wherein the medium chain triglycerides are Miglyol® 812.
A13) A topical pharmaceutical composition according to any one of embodiments A1 to A12, comprising solvents with penetration enhancing activity.
A14) A topical pharmaceutical composition according to embodiment A13, wherein the content of solvents with penetration enhancing activity are from 2 to 30 wt.% based on the total weight of the composition.
A15) A topical pharmaceutical composition according to embodiment A14, wherein the content of solvents with penetration enhancing activity are from 8 to 12 wt.% based on the total weight of the composition.
A16) A topical pharmaceutical composition according to embodiment A15, wherein the content of solvents with penetration enhancing activity are 10 wt.% based on the total weight of the composition.
A17) A topical pharmaceutical composition according to any one of embodiments A13 to A16, wherein the solvents with penetration enhancing activity is diethylene glycol monoethylether.
A18) A topical pharmaceutical composition according to any one of embodiments A1 to A17, comprising glycols.
A19) A topical pharmaceutical composition according to embodiment A18, wherein the content of glycols are from 2 to 15 wt.% based on the total weight of the composition.
A20) A topical pharmaceutical composition according to embodiment A19, wherein the content of glycols are from 4 to 6 wt.% based on the total weight of the composition.
A21) A topical pharmaceutical composition according to embodiment A20, wherein the content of glycols are 5 wt.% based on the total weight of the composition.
A22) A topical pharmaceutical composition according to any one of embodiments A18 to A21 , wherein the glycols are propylene glycol. A23) A topical pharmaceutical composition according to any one of embodiments A1 to A22, wherein the content of water is from 30 to 70 wt.% based on the total weight of the composition.
A24) A topical pharmaceutical composition according to embodiment A22, wherein the content of water is from 40 to 60 wt.% based on the total weight of the composition.
A25) A topical pharmaceutical composition according to embodiments A23 or A24, wherein the water is distilled or purified water.
A26) A topical pharmaceutical composition according to any one of embodiments A1 to A25, wherein the content of the emulsifier is from 0.05 to 10 wt.% based on the total weight of the composition.
A27) A topical pharmaceutical composition according to embodiment A26, wherein the content of the emulsifier is from 0.2 to 6.0 wt.% based on the total weight of the composition. A28) A topical pharmaceutical composition according to embodiments A26 or A27, wherein the emulsifier is selected from one or more members of the group consisting of
polyoxyethylene (Cs to C20) alkyl ethers and glycerol monostearate 40-55.
A29) A topical pharmaceutical composition according to embodiment A28, wherein the polyoxyethylene (Cs to C20) alkyl ethers are selected from one or more members of the group consisting of macrogol 21 stearyl ether, macrogol 2 stearyl ether, macrogol 20 stearyl ether, polyethylene glycol hexadecyl ether and PEG- 100 stearyl ether.
A30) A topical pharmaceutical composition according to embodiment A29, wherein the polyoxyethylene (Cs to C20) alkyl ethers is macrogol 21 stearyl ether.
A31) A topical pharmaceutical composition according to any one of embodiments A28 to A30, wherein the emulsifier is a mixture of macrogol 21 stearyl ether and glycerol monostearate 40-55.
A32) A topical pharmaceutical composition according to embodiment A31 , wherein the content of macrogol 21 stearyl ether is from 0.7 to 1.5 wt.% based on the total weight of the composition, and the content of glycerol monostearate 40-55 is from 1.5 to 2.5 wt.% based on the total weight of the composition.
A33) A topical pharmaceutical composition according to embodiment A32, wherein the content of macrogol 21 stearyl ether is from 0.9 to 1.3 wt.% based on the total weight of the composition, and the content of glycerol monostearate 40-55 is 2.0 wt.% based on the total weight of the composition.
A34) A topical pharmaceutical composition according to any one of embodiments A1 to A33, comprising a thickening agent.
A35) A topical pharmaceutical composition according to embodiment A34, comprising a thickening agent which also act as co-emulsifier. A36) A topical pharmaceutical composition according to embodiment A35, wherein the thickening agent which also act as co-emulsifier is selected from one or more members of the group of (C10 to C25) fatty alcohols.
A37) A topical pharmaceutical composition according to embodiment A36, wherein the content of (C10 to C25) fatty alcohols is 4 to 12 wt.% based on the total weight of the composition.
A38) A topical pharmaceutical composition according to embodiment A37, wherein the content of (C10 to C25) fatty alcohols is 6.0 to 10.0 wt.% based on the total weight of the composition.
A39) A topical pharmaceutical composition according to any one of embodiments A36 to A38, wherein the (C10 to C25) fatty alcohols are a mixture of cetyl alcohol and stearyl alcohol. A40) A topical pharmaceutical composition according to embodiment A39, wherein the content of cetyl alcohol is from 3.0 to 5.0 wt.%, based on the total weight of the composition, and the content of stearyl alcohol is from 3.0 to 5.0 wt.% based on the total weight of the composition.
A41) A topical pharmaceutical composition according to embodiment A40, wherein the content of cetyl alcohol is 4.0 wt.%, based on the total weight of the composition, and the content of stearyl alcohol is 4.0 wt.% based on the total weight of the composition.
A42) A topical pharmaceutical composition according to any one of embodiments A1 to A41 , comprising a buffering agent.
A43) A topical pharmaceutical composition according to embodiment A42, wherein the buffering agent is chosen so that the final pH value of the composition is from pH 5.0 and pH 5.8.
A44) A topical pharmaceutical composition according to embodiment A43, wherein the buffering agent is chosen so that the final pH value of the composition is from pH 5.2 and pH 5.6.
A45) A topical pharmaceutical composition according to embodiments A43 or A44, wherein the buffering agent is chosen so that the final pH value of the composition is pH 5.4.
A46) A topical pharmaceutical composition according to any one of embodiments A42 to A45, wherein the buffering agent is a citric acid / citrate buffer.
A47) A topical pharmaceutical composition according to any one of embodiments A1 to A46, wherein the content of the antimicrobial preservative is from 0.8 to 1.0 wt.% based on the total weight of the composition.
A48) A topical pharmaceutical composition according to embodiment A47, wherein the antimicrobial preservative is benzyl alcohol.
A49) A topical pharmaceutical composition according to embodiment A48, wherein the content of benzyl alcohol is 1.0 wt.% based on the total weight of the composition. A50) A topical pharmaceutical composition according to any one of embodiments A1 to A49, wherein the content of compound of formula (I) is from up to 0.30 wt.% based on the total weight of the composition.
A51) A topical pharmaceutical composition according to embodiment A50, wherein the content of compound of formula (I) is from 0.05 to 0.30 wt.% based on the total weight of the composition.
A52) A topical pharmaceutical composition according to embodiment A51 , wherein the content of compound of formula (I) is from 0.10 to 0.27 wt.% based on the total weight of the composition.
A53) A topical pharmaceutical composition according to embodiment A52, wherein the content of compound of formula (I) is from 0.20 to 0.27 wt.% based on the total weight of the composition.
A54) A topical pharmaceutical composition according to embodiment A53, wherein the content of compound of formula (I) is 0.25 wt.% based on the total weight of the composition. A55) A topical pharmaceutical composition consisting of:
Figure imgf000015_0002
A topical pharmaceutical composition consisting of
Figure imgf000015_0001
Medium chain triglycerides (Miglyol® 15 812)
Stearyl alcohol 4.0
Cetyl alcohol 4.0
Glycerol monostearate
2.0 40-55 (type II)
Diethylene glycol mono
10 ethyl ether (Transcutol®)
Propylene glycol 5.0
Benzyl alcohol 1 .0
Macrogol 21 stearyl ether
1 .3 (Brij® S721)
Citric acid anhydrous 0.12
Sodium citrate dihydrate 0.29
Purified water 56.99
A topical pharmaceutical composition consisting of:
Figure imgf000016_0001
A topical pharmaceutical composition consisting of [wt.%]
Compound of formula (1) 0.30
Medium chain
triglycerides (Miglyol® 15 812)
Stearyl alcohol 4.0
Cetyl alcohol 4.0
Glycerol monostearate
2.0 40-55 (type II)
Diethylene glycol mono
10 ethyl ether (Transcutol®)
Propylene glycol 5.0
Benzyl alcohol 2.0
Macrogol 21 stearyl ether
0.9 (Brij® S721)
Citric acid anhydrous 0.12
Sodium citrate di hydrate 0.29
Purified water 56.39
A topical pharmaceutical composition consisting of
[wt.%]
Compound of formula (I) 0.30
Medium chain
triglycerides (Miglyol® 15 812)
Stearyl alcohol 4.0
Cetyl alcohol 4.0
Glycerol monostearate
2.0 40-55 (type II)
Diethylene glycol mono
10 ethyl ether (Transcutol®)
Propylene glycol 5.0
Phenoxyethanol 1 .0
Macrogol 21 stearyl ether
1 .3 (Brij® S721)
Citric acid anhydrous 0.12
Sodium citrate di hydrate 0.29
Purified water 56.99 A60) A process to prepare the topical pharmaceutical composition according to any one of embodiments A1 to A59, comprising the steps of
(a) Preparing a solution/suspension of the compound of formula (I) in a suitable
solvent;
(b) Preparing a melt of the remaining solvent components of the pharmaceutical composition not including water, the emulsifier and the antimicrobial preservative and optionally the thickening agent;
(c) Adding the solution/suspension of step (a) to the melt of step (b) to obtain a melt containing the compound of formula (I);
(d) Preparing the aqueous phase;
(e) Mixing the melt of step (c) with the aqueous phase of step (d) and cooling the mixture down to obtain the oil-in-water cream.
A61) A process according to embodiment A60, wherein in step (a) the compound of formula (I) is dissolved/suspended in solvents with penetration enhancing activity.
A62) A process according to embodiments A60 or A61 , wherein in step (b) the melt is prepared from fatty acid glycerides, glycols, an emulsifier, an antimicrobial preservative and a thickening agent.
A63) A process according to any one of embodiments A60 to A62, wherein in step (d) the aqueous phase is an aqueous buffer solution of pH 5.2 - 5.6.
A64) A process according to any one of embodiments A60 to A63, wherein the compound of formula (I) used in step (a) is in crystalline form according to embodiments B1 to B6.
A65) A composition according to any one of embodiments A1 to A59, wherein the composition is prepared from the compound of formula (I) in crystalline form according to embodiments B1 to B6.
A66) A composition according to any one of embodiments A1 to A59 or A65 for use in the treatment or prevention of a disease or disorder selected from any one of Lists A to K.
A67) Use of a composition according to any one of embodiments A1 to A59 or A65 in the treatment or prevention of a disease or disorder selected from any one of Lists A to K.
A68) Use of a composition according to any one of embodiments A1 to A59 or A65 in the manufacture of a medicament for the treatment or prevention of a disease or disorder selected from any one of Lists A to K.
A69) A method of treating a disease or disorder selected from any one of Lists A to K in a subject, comprising the step of administering to the subject a therapeutically effective amount of a composition according to any one of embodiments A1 to A59 or A65. A70) A method of preventing a disease or disorder selected from any one of Lists A to K in a subject, comprising the step of administering to the subject a therapeutically effective amount of a composition according to any one of embodiments A1 to A59 or A65.
A71) A composition according to any one of embodiments A1 to A59 or A65 for use in the treatment of cutaneous squamous cell carcinoma in situ (cSCCis).
A72) Use of a composition according to any one of embodiments A1 to A59 or A65 in the treatment of cutaneous squamous cell carcinoma in situ (cSCCis).
A73) Use of a composition according to any one of embodiments A1 to A59 or A65 in the manufacture of a medicament for the treatment of cutaneous squamous cell carcinoma in situ (cSCCis).
A74) A method of treating cutaneous squamous cell carcinoma in situ (cSCCis) in a subject, comprising the step of administering to the subject a therapeutically effective amount of a composition according to any one of embodiments A1 to A59 or A65.
A75) A composition for use according to embodiment A71 , wherein the application of the composition is twice daily.
A76) Use according to embodiment A72, wherein the application of the composition is twice daily.
A77) Use according to embodiment A73, wherein the medicament is applied twice daily. A78) A method according to embodiment A74, wherein the composition is applied twice daily.
A79) A composition for use according to embodiments A71 or A75, wherein the total daily dose of the compound of formula (I) is 0.21 mg.
A80) Use according to embodiments A72 or A76, wherein the total daily dose of the compound of formula (I) is 0.21 mg.
A81) Use according to embodiments A73 or A77, wherein the total daily dose of the compound of formula (I) is 0.21 mg.
A82) A method according to embodiments A74 or A78, wherein the total daily dose of the compound of formula (I) is 0.21 mg.
B1) A crystalline form of a compound of formula (I)
Figure imgf000019_0001
B2) The crystalline form according to embodiment B1 , which is non-solvated. B3) The crystalline form according to embodiments B1 or B2, characterized by an X-Ray powder diffraction pattern comprising the following peaks given at degrees 2-Theta ±0.5 degrees: 3.97, 7.96, 19.22, 14.40.
B4) The crystalline form according to embodiments B1 or B2, having a X-ray diffraction spectrum substantially the same as the X-ray powder diffraction spectrum shown in Figure 1. B5) The crystalline form according to embodiments B1 or B2, having a differential scanning calorimetry curve substantially the same as that shown in Figure 2.
B6) The crystalline form according to embodiments B1 or B2, having a thermogravimetric analysis diagram substantially the same as that shown in Figure 3.
B7) The crystalline form according to any one of embodiments B1 to B6, for use as a pharmaceutical.
B8) A combination comprising a therapeutically effective amount of a crystalline form according to any one of embodiments B1 to B6, and one or more therapeutically active agents.
B9) A crystalline form according to any one of embodiments B1 to B6 for use in the treatment or prevention of a disease or disorder selected from any one of Lists A to K.
B10) Use of a crystalline form according to any one of embodiments B1 to B6 in the treatment or prevention of a disease or disorder selected from any one of Lists A to K.
B1 1) Use of a crystalline form according to any one of embodiments B1 to B6 in the manufacture of a medicament for the treatment or prevention of a disease or disorder selected from any one of Lists A to K.
B12) A method of treating a disease or disorder selected from any one of Lists A to K in a subject, comprising the step of administering to the subject a therapeutically effective amount of a form according to any one of embodiments B1 to B6.
B13) A method of preventing a disease or disorder selected from any one of Lists A to K in a subject, comprising the step of administering to the subject a therapeutically effective amount of a form according to any one of embodiments B1 to B6.
B14) A crystalline form according to any one of embodiments B1 to B6 for use in the treatment of cutaneous squamous cell carcinoma in situ (cSCCis).
B15) Use of a crystalline form according to any one of embodiments B1 to B6 in the treatment of cutaneous squamous cell carcinoma in situ (cSCCis).
B16) Use of crystalline form according to any one of embodiments B1 to B6 in the manufacture of a medicament for the treatment of cutaneous squamous cell carcinoma in situ (cSCCis). B17) A method of treating cutaneous squamous cell carcinoma in situ (cSCCis) in a subject, comprising the step of administering to the subject a therapeutically effective amount of a form according to any one of embodiments B1 to B6. Lists of diseases and disorders:
List A: Non-melanoma skin cancers such as basal cell carcinoma and squamous cell carcinoma (SCC); their pre-malignant stages such as squamous cell carcinoma in situ (SCCis), actinic keratosis (AK), solar keratosis and chronically sun damaged skin; and other hyperproliferative skin disorders caused by dysregulation of skin fibroblasts such as skin fibrosis, scleroderma, hypertrophic scars and keloids.
List B: Non-melanoma skin cancers.
List C: Basal cell carcinoma and squamous cell carcinoma (SCC).
List D: Pre-malignant stages of Non-melanoma skin cancers.
List E: Squamous cell carcinoma in situ (SCCis), actinic keratosis (AK), solar keratosis and chronically sun damaged skin.
List F: Hyperproliferative skin disorders caused by dysregulation of skin fibroblasts; other than non-melanoma skin cancers and their pre-malignant stages.
List G: Skin fibrosis, scleroderma, hypertrophic scars and keloids.
List H: Squamous cell carcinoma in situ (SCCis).
List J: Cutaneous squamous cell carcinoma in situ (cSCCis).
List K: Keloids.
The invention is illustrated by the following Examples.
Abbreviations:
AUC Area Under the plasma concentration-time Curve
BLQ Below the Lower Limit of Quantification
°C degree(s) Celsius
DAD Diode Array Detector
DSC Differential scanning calorimetry
DVS Dynamic vapor sorption
FBS Fetal Bovine Serum
h hour(s)
HOT tritiated water
IR Infra-Red spectroscopy
LC Liquid chromatography
LC-MS/MS Liquid chromatography - Tandem mass spectrometry LLNA local lymph node assay
LLOQ lower limit of quantification
LSC Liquid Scintillation Counting
m month(s)
MBq mega Becquerel
MS mass spectrometry
PBS Phosphate-buffered saline
PEG Polyethylene glycol
RH relative humidity
rpm revolutions per minute
RRT Relative retention time
RT room temperature
SEM Scanning electron microscopy
T Temperature
TGA Thermogravimetric analysis
TGA-FTIR Thermogravimetric analysis in line with IR detector
ULOQ upper limit of quantification
v/v volume per volume
wt.% or % by weight. weight percent
XRPD X-ray powder diffraction
Methods:
TG-method
Instrument Mettler 850
Temperature range 30-300°C
Scan rate 20K/min
Nitrogen flow 45.0ml_/min
DSC-method
Instrument Perkin Elmer Diamond and TA instrument Q2000
Temperature range 30.00°C - 300.00°C
Scan rate 20.00°C/min
Nitrogen flow 20.0m L/min
XRPD-method
Instrument Bruker D8 Advance
Irradiation CuKa (30 kV, 40 mA)
Divergence slice 2.5 mm (Soller slit) and 6 mm (antiscatter slit)
Measuring slice V12 (variable) Scan type Continuous scan
Step time > 40 s for Lynxeye, > 100 s for Vantec detector
Scan range 2° - 40° (2 theta value)
IR-method
Instrument Bruker Vertex SYS-IR-00002
Mode ATR
Scan range 4000 cm-1 - 600 cm-1
Technique Neat sample compressed on the ATR cell
Microscopy
Optical microscopy Olympus BX51
SEM instrument Zeiss SUPRA 40
HPLC-method 1
Instrument Agilent 1200
Column Phenomenex Gemini C18, 150*3.0 mm, 3.0 μηι
Column Temperature 40 °C
Flow rate 0.7 mL/min
Injection 3 μΙ_
Detection 248 nm/DAD
Mobile phase composition:
Mobile phase A 20mM (NH4)2HP04 in water
Mobile phase B Acetonitrile
Gradient:
Time
A [%] B[%]
(min)
0.00 50 50 Start of acquisition
16 15 85
19 15 85 End of acquisition
19.1 50 50
25 50 50 Next injection
HPLC-method 2
Principle RP HPLC with UV detection
Apparatus HP 1290 (Agilent)
Column Acquity UPLC BEH C18
Length 50 mm, internal diameter 2.1 mm, particle size 1.7μηι, or equivalent
Flow rate 0.4 ml/min Detection 248 nm
Column temperature 40°C
Auto-sampler temperature 25°C
Needle wash Ethanol
Injection volume 2 μΙ of the test and reference solutions
Mobile phase composition:
Mobile phase A 10mM Ammoniumacetate buffer : Acetonitrile (95/5 v/v) Mobile phase B Water / Acetonitrile (5/95 v/v)
Gradient:
Time
A [%] B[%]
(min)
0 95 5
0.5 80 20
17 72 28
21 15 85
23 15 85
23.1 95 5
25 95 5
LC-MS/MS-method 1
LC condition
Analytical column Ascentis Express C18 50 x 2.1 mm, 2.7 μηι from Supeico Injection volume 2 μΙ_ (20 μΙ_ loop)
Column oven 50 °C
Mobile phases:
A: 0.1 % formic acid in water
B: Acetonitrile
Flow rate: 0.7 mL/min
Gradient:
Time Divert
Valve A [%] B[%]
(min)
Position
0.00 waste 95 5
0.50 MS
1.50 MS 5 95
2.20 MS 5 95
2.30 MS 95 5 2.50 waste 95 5
3.00 waste 95 5
LC system Pump(s) Agilent Binary pumps 1200 and 1100
Autosampler PAL HTS-xt, AB Sciex.
Mass spectrometer API6500, AB Sciex.
MS conditions Turbo Spray, positive ion mode.
LC-MS/MS-method 2
LC condition
Analytical column Ascentis Express C18 50 x 2.1 mm, 2.7 μηι from Supeico Injection volume 15 μί (20 μί ΐοορ)
Column oven 50 °C
Mobile phases:
A: 0.1 % formic acid in water
B: Acetonitrile (UHPLC-MS grade)
Flow rate: 0.7 mL/min
Gradient:
Time Divert
Valve A [%] B[%;
(min)
Position
0.00 waste 95 5
0.50 MS
1.50 MS 5 95
2.20 MS 5 95
2.30 MS 95 5
2.50 waste 95 5
3.00 stop run 95 5
Mass spectrometer TQ 6500 from Applied Biosystems
Mobile phase MS
cleaning Acetonitrile (UHPLC-MS grade) with a constant flow of 300 μΙ_/ηιίη
Auto-sampler washing
solutions Autosampler wash 1 : Acetonitrile HPLC
Autosampler wash 2: Methanol-2 Propanol-Tetrahydrofurane
(1-1-1 , v-v-v) Column pressure Around 3200 psi
Ionization source and
conditions ESI positive ionization mode
Source temperature 600°C
Collision Activated Dissociation in position 10 (with N2 gas)
Curtain gas (N2) flow rate: 20 psi
Gas 1 (Air): 40 psi and Gas 2 (Air): 40 psi
IS Voltage: 5500 V
Entrance Potential: 10V
MS/MS mode Centroid, MRM (MS/MS)
LC-MS/MS-method 3
LC condition
Analytical column Ascentis Express C18 50 x 2.1 mm, 2.7 μηι from Supeico
Injection volume 3 μΙ_ (20 μΙ_ loop)
Column oven 50 °C
Mobile phases:
A: 0.1 % formic acid in water
B: Acetonitrile (UHPLC-MS grade)
Flow rate: 0.7 mL/min
Gradient:
Time Divert
Valve A [%] B[%;
(min)
Position
0.00 waste 95 5
0.80 MS
1.50 MS 5 95
2.00 waste
2.20 waste 5 95
2.30 waste 95 5
3.00 stop run 95 5
LC system Pump(s) Agilent Binary pumps 1200 and 1100
Mass spectrometer Triple Quad 5500, Applied Biosystems (TS-013-819) MS conditions Turbo Spray, positive ion mode.
Mobile phase MS
cleaning Acetonitrile (UHPLC-MS grade) with a constant flow of 300 μΙ_/ηιίη
Auto-sampler washing
solutions Autosampler wash 1 : Acetonitrile HPLC
Autosampler wash 2: Methanol-2 Propanol-Tetrahydrofurane
(1-1-1 , v-v-v)
Column pressure Around 3200 psi
Ionization source and
conditions ESI positive ionization mode
Source temperature 600°C
Collision Activated Dissociation in position 7 (with N2 gas)
Curtain gas (N2) flow rate: 20 psi
Gas 1 (Air): 40 psi and Gas 2 (Air): 40 psi
IS Voltage: 5500 V
Entrance Potential: 10V
MS/MS mode Centroid, MRM (MS/MS)
LCMS-method 4
Instrument Agilent 1200 LC combined 6130 Quadrupole MS
Column Agilent Poroshell 120 SB-C18 75*3.0 mm, 2.7μηι
Flow rate 0.8 ml/min
Temperature 40 °C
Mobile phases:
A: 0.1 % formic acid in water
B: Acetonitrile
Gradient:
Time
A[%] B [%]
(min)
0.0 95 5
0.5 95 5
8.0 5 95
12.0 5 95
12.1 95 5
15.0 95 5 EXAMPLES
1. Pharmaceutical Compositions
a) oil-in-water cream compositions and preparation
The following steps were used to prepare the oil-in-water cream compositions:
1. Citric acid anhydrous and sodium citrate dehydrate were added to the water and heated to 70°C under stirring, until a clear solution was obtained.
2. In another vessel, medium chain triglycerides, stearyl alcohol, cetyl alcohol, glycerol monostearate 40-55, macrogol 21 stearyl ether, the antimicrobial preservative and propylene glycol were mixed at 70°C under stirring. To the melt, a solution/suspension of the compound of formula (I) in diethylene glycol monoethylether was added.
3. The mixture from step 2 was transferred into the mixture from step 1 while stirring and homogenizing under low to moderate vacuum and homogenization was continued under stirring.
4. The cream was cooled down to 25°C ± 5°C under stirring.
Table 1 : Pharmaceutical compositions, oil-in-water cream
Var A Var B Var C Var D Var E Ref Var Y
Composition [wt.%] [wt.%] [wt.%] [wt.%] [wt.%] [wt.%]
Compound of formula
0.30 0.25 0.1 0.30 0.30 0.30 (I)
Medium chain
triglycerides (Miglyol® 15 15 15 15 15 15 812)
Stearyl alcohol 4.0 4.0 4.0 4.0 4.0 4.0
Cetyl alcohol 4.0 4.0 4.0 4.0 4.0 4.0
Glycerol monostearate
2.0 2.0 2.0 2.0 2.0 2.0 40-55 (type II)
Diethylene glycol
monoethylether 10 10 10 10 10 10
(Transcutol®)
Propylene glycol 5.0 5.0 5.0 5.0 5.0 5.0
Benzyl alcohol 1 .0 1 .0 1 .0 2.0 - 3.0
Phenoxyethanol - - - - 1 .0 -
Macrogol 21 stearyl
1 .3 1 .3 0.9 0.9 1 .3 1 .3 ether (Brij® S721)
Citric acid anhydrous 0.12 0.12 0.12 0.12 0.12 0.12 Sodium citrate
0.29 0.29 0.29 0.29 0.29 0.29 dihydrate
Purified water 56.99 57.04 57.59 56.39 56.99 54.99 b) Reference liquid crystal cream compositions and preparation
The following steps were used to prepare the liquid crystal cream compositions:
1. Xanthan gum was dispersed in propylene glycol to form xanthan gum premix. This xanthan gum premix was then transferred into water and carbopol 974P (which was pre- stirred at 25°C) was added.
2. The mixture from step 1 was stirred at 75°C until a homogenous gel was formed.
3. Compound of formula (I) was suspended in one aliquot of diethylene glycol
monoethylether.
4. In another vessel, the following components were mixed: the polyoxyl stearyl ethers, corn oil, cetyl alcohol, capryl caproyi polyoxyl glyceride, benzyl alcohol and compound of formula (I) suspension from step 3.
5. The mixture of step 4 was then stirred at 75°C until clear solution was formed.
6. The solution of step 5 was then added to the gel obtained in step 2, under stirring and homogenization at 75°C.
7. The homogenized mixture of step 6 was allowed to cool to 40°C
8. 2 N NaOH solution was then added to the material of step 7, whereby stirring and homogenization was continued.
9. The mixture was allowed to cool to RT with stirring.
Table 2: Pharmaceutical compositions, liquid crystal cream
Figure imgf000029_0001
Xanthan gum 0.20 0.20
NaOH (2N solution) 1.8 0.25
Purified water 47.7 49.25
2. Excipient Compatibility of the compound of formula (I)
A saturated solution of the compound of formula (I) in the respective medium was prepared and kept at 50°C for 1 month. Degradation products were analyzed by HPLC (HPLC-method 1)
Table 3: Excipient Compatibility of the compound of formula (I)
Figure imgf000030_0001
The compound of formula (I) showed some tendency to degradation when exposed to excipients providing oxidative potential such as PEG and oleylalcohol. Medium chain triglycerides demonstrated exceptionally good compatibility with the compound of formula (I).
3. In Vitro 3T3 NRU Phototoxicity Assay of the compound of formula (I)
The standard preclinical in vitro assay for phototoxicity assessment is the "in vitro 3T3 neutral red uptake (NRU) phototoxicity test" (OECD (2004), in vitro 3T3 NRU phototoxicity test.
Guideline for testing of chemicals no. 432.; see also J. Schuemann et.al.; Toxicological Sciences 139(1), (2014), p.245-256)
The compound of formula (I) showed a photoirritation factor (PI F) of 7 in this test.
4. UV-Local lymph node assay (LLNA) in mouse with the pharmaceutical compositions
The local lymph node assay (G. F. Gerberick et.al.; Methods 41 (2007) p. 54-60) assesses the photo-sensitization potential of compounds and compositions. The compositions were repeatedly dosed to mice which were then exposed to UV-Vis radiation with a light dose of at least 10 J/cm2. Lymph node weight, ear weight and lymph node cell count were then analysed.
Table 4.1 : LLNA assay, study design untreated positive positive Placebo Placebo Ref Ref Var Y
control control Ref Var Ref Var Y Var (UVA/Vis)
(UVA/Vis) Y (UVA/Vis) Y
Concentration 0 0.5 0.5 0 0 0.3 0.3 [%]
Administration 25 25 25 12.5 12.5 12.5 12.5 Vol per ear
[uL]
Figure imgf000031_0001
Table 4.2: LLNA assay, results
Figure imgf000031_0002
n = 6 n = 6
The results suggest an enhancement of skin irritation and lymph-node activation in the presence of simulated sunlight in this in vivo assay for Ref Var Y composition whereas the pharmaceutical compositions according to the invention with a reduced benzyl alcohol level exemplified by Var A did not show such findings. There were no statistically significant differences in lymph node parameters between irradiated and non-irradiated animals.
5. Stability Tests of the pharmaceutical compositions
Chemical stability:
The pharmaceutical compositions, as prepared above, in 10 g aluminum tubes, under inverse storage were tested for stability. Testing was performed at 5 °C / ambient RH, 25 °C / 60 %RH, 40 °C / 75 %RH and other conditions as indicated.
Table 5.1 : Data for composition Var B:
Figure imgf000032_0001
Impurities at RRT 0.66, RRT 1 .08 and RRT 1 .37 are by products from drug substance, not included in total impurities.
The composition Var B showed good stability data at 5°C, at long term condition of
25°C/60% RH, at accelerated conditions 30°C/75% RH after 12 months storage and at 40°C/75% RH after 6 months storage.
Table 5.2: Data for composition Ref Var Y:
Storage conditions Degradation products (HPLC-method 2) [%] Compound RRT 0.89 RRT 1 .34 RRT 1 .41 Total
of formula
(I) [%]
Initial analysis 100.5 <0.1 <0.1 <0.1 <0.1
5°C/ 1 m 98.3 <0.1 <0.1 <0.1 <0.1 ambient RH 3m 100.6 <0.1 <0.1 <0.1 <0.1
6m 99.1 <0.1 <0.1 <0.1 <0.1
12m 99.7 <0.1 <0.1 <0.1 <0.1
25°C/60% RH 1 m 99.5 <0.1 <0.1 <0.1 <0.1
3m 100.1 <0.1 <0.1 <0.1 <0.1
6m 99.1 <0.1 <0.1 <0.1 <0.1
12m 99.5 0.2 <0.1 0.2 0.3
30°C/75% RH 1 m 98.3 <0.1 <0.1 <0.1 <0.1
3m 99.3 <0.1 <0.1 <0.1 <0.1
6m 99.4 <0.1 <0.1 <0.1 <0.1
12m 95.1 0.1 <0.1 0.3 0.4
40°C/75% RH 1 m 99.3 <0.1 <0.1 <0.1 <0.1
3m 98.2 <0.1 <0.1 <0.1 <0.1
6m 96.2 0.2 0.1 0.3 0.6
Table 5.3: Data for composition Ref Var X:
Storage conditions Compound of Degradation products (HPLC-method 2) [%]
formula (I) [%] RRT RRT RRT RRT RRT Total
0.32 0.91 1 .25 1 .34 1 .41
Initial analysis 101 .3 0.4 <0.1 <0.1 <0.1 <0.1 0.4
5°C/ 1 m 100.1 0.3 <0.1 <0.1 <0.1 <0.1 0.3 ambient RH 3m 100.9 0.4 <0.1 <0.1 <0.1 <0.1 0.4
6m 101 .9 0.4 <0.1 <0.1 <0.1 <0.1 0.4
12m 101 .1 0.4 <0.1 <0.1 <0.1 <0.1 0.4
25°C/60% RH 1 m 99.2 0.4 <0.1 <0.1 <0.1 <0.1 0.4
3m 98.8 0.4 <0.1 <0.1 <0.1 <0.1 0.4
6m 99.3 0.4 0.1 <0.1 <0.1 0.2 0.7
12m 98.8 0.5 0.2 <0.1 <0.1 0.4 1 .0
30°C/75% RH 1 m 100.2 0.4 <0.1 <0.1 <0.1 <0.1 0.4
3m 99.2 0.4 <0.1 <0.1 <0.1 <0.1 0.4
6m 99.7 0.5 0.2 <0.1 <0.1 0.3 1 .0
12m 98.0 0.6 0.2 0.1 <0.1 0.5 1 .4
40°C/75% RH 1 m 99.4 0.4 0.1 <0.1 <0.1 <0.1 0.5
3m 98.0 0.5 0.2 <0.1 <0.1 <0.1 0.8 6m 98.1 0.8 0.2 <0.1 0.1 0.4 1 .5
The pharmaceutical composition Var B clearly shows a better stability in terms of
degradation when compared to the Ref Var X composition. 6. Exposure after dermal administration
a) Testing of the pharmaceutical compositions in the In vitro human skin penetration / permeation assay
The compositions of the present invention were tested for penetration into, as well as permeation through dermatomized healthy human skin in the static Franz cells for the compound of formula (I).
Skin preparation
Frozen punches of 25 mm in diameter of human abdominal plastic surgery skin, already dermatomized to a thickness of -500 μηι, were obtained from Alphenyx (CSO) Technopole Grand Luminy (Pepiniere Biotech), Marseille, France. Before starting the experiment the punches were kept at -20 °C for not more than 6 months after surgery. The thawed punches were assembled between the donor and the receptor chambers of the Franz cells (T. J. Franz; J Invest Derm; 64(3) (1975) p.190-195) of the automated Logan® diffusion cells system (Logan Instruments Corp, Somerset, NJ, USA).
Determination of the skin integrity
The integrity of skin was determined in each Franz Cell by evaluating the permeation of tritiated water (HOT) through the skin; after pre-equilibration, 500 of HOT (approx. 0.1 MBq/mL) were applied on the surface of the skin fixed on the cells. After 30 min of equilibration, the HOT was removed from the skin with cotton tips; 3.5 mL of the receptor phase (composition of the receptor fluid is described below) were sampled in order to measure the fraction of HOT, which permeated across the skin. Radioactivity in the receptor phase was measured using the Liquid Scintillation Systems Tri-Carb 3110 (Perkin Elmer Instr. Co., Meriden, CT, USA). For quench correction an external standard method was used. Quench correction curves were established by means of sealed standards (Perkin Elmer Instruments).
Determination of the in vitro penetration into and permeation across human skin
The skin was used as a membrane separating the donor and receptor chambers of the static Franz diffusion cells (~7 mL volume, -1.5 cm diameter) assembled in the Logan® system. The receptor chambers were filled with the receptor fluid (PBS/FBS, 2/1 , v/v; pH=7.4) to simulate the human physiological conditions and the systemic removal of the drug from skin. In addition, the receptor fluid contained 100 U/mL of a 1 % penicillin/streptomycin mixture (v/v) to prevent microbiological contamination. The effective skin area for diffusion was 1.77 cm2 and the volumes of receiver compartment were in the range of 6.33 to 7.00 ml_, respectively. The temperature of the cells was kept constant using a circulating water bath at 32 ±1 °C. Magnetic stirrer bars were constantly used during the entire experiment to ensure receptor uniformity.
Aliquots of the test compositions (nominal weight of 300 mg) were applied as a single dose on the skin samples mounted on the Franz diffusion cells (corresponding to a sampling time of 0 h). The compositions were left on the skin for 24 h or 48 h.
The donor compartments of Franz diffusion cells were semi-occluded with parafilm (Parafilm'8 M) with holes.
Collection and handling of the samples
For determination of compound of formula (I) that permeated across the skin, aliquots of 0.5 ml_ of the receptor fluid were collected from the receptor compartment at 4, 10, 18, 24, 30, 36, 42, and 48 h after application. For the formulation B with 24 h study duration, no receptor fluid was sampled. The volume of 3.3 ml_ taken from the receptor compartment as a sample and as the volume used to flush the tubes was replaced automatically every time with the same volume of fresh receptor fluid in order to keep the total receptor fluid volume constant during the entire assay. Receptor fluids were frozen and kept at -20°C until bioanalysis for compound quantification.
At the end of the treatment period, the residual formulation on the surface of each skin sample was carefully removed with a cotton tip applicator, and the application area was washed with a cotton tip containing water and gently dried with new cotton applicators. The procedure was repeated three times. The stratum corneum was then separated from the human skin by 20 tape strips using a commercial adhesive tape (Scotch® 550, 3M). All strips were discarded. Biopsies of 12 mm diameter of the treated area of the stripped skin were taken and weighed. The stripped skin biopsy samples were frozen and kept at -20°C until bioanalysis for the quantification of the compound of formula (I).
Analytical methods for determination of:
HOT in receptor fluid LSC: Radioactivity in the receptor phase was measured by
LSC in a Packard TRI-Carb 31 10TR LSC Counter (Perkin Elmer, Zurich, Switzerland). For quench correction an external standard method was used. Quench correction curves were established by means of sealed standards (Packard
Instructions)
Compound of formula (I) LC-MS/MS-method 1 ; LLOQ: 0.250 ng/mL (receptor fluid), 2.50 ng/g (skin samples) Table 6.1 : Testing of pharmaceutical compositions in the In vitro human skin penetration / permeation assay, results
Figure imgf000036_0001
Data represented is the mean of n = 4 replicates
The permeation rate (flux) of the compound of formula (I) through the skin is indicative for a low systemic exposure in vivo.
b) Skin and plasma exposure after multiple topical administration to minipigs
The skin and plasma exposure following topical treatments with the Var B composition was investigated in a minipig toxicity study (Gottingen SPF / female / n=3).
The composition was applied once daily to 10% of the body surface area over 2 weeks and skin was washed 6 h after each application under occlusion.
Plasma:
Blood samples were collected into tubes containing K3EDTA as anticoagulant. Tubes were placed in ice water for max 15 min until centrifugation (10 min, 1500g, +4°C). Plasma was then transferred into uniquely labeled 1.8 mL NUNC 2D coded tubes and frozen at -65°C or below until bioanalysis.
All frozen plasma samples were ultra-sonicated for approximately 5 minutes after thawing and before pipetting.
1. In a 96-well plate, pipetted 50 μΙ_ of plasma sample: Standards, QCs, Blank, Zero and incurred samples.
2. Added 25 μΙ_ of the working solution of [D8] compound of formula (I) at 5.00 ng/mL in acetonitrile-water (1-1 , v-v) into the wells. Except for blank samples where 25 μΙ_ of acetonitrile-water (1-1 , v-v) were added in replacement.
3. Added 150 μΙ_ of 0.1 % formic acid in water in all wells.
4. Sealed the plate and mixed on the 96-DW Eppendorf Mix Mate for approximately 2 minutes at 1600 rpm.
5. Used positive pressure processor for 96-well plate and loaded a volume of 200 μΙ_ sample on isolute SLE+ 96-well plate (200 μΙ_). Applied 30-60 psi pressure to initiate the loading and then waited 5 minutes for complete loading. 6. Eluted twice with 400 μΙ_ ethyl acetate in a new 96-well collection plate and apply vacuum (5-6 psi). Wait approximately 30 sec after each step of elution.
7. Evaporated to dryness under nitrogen stream at 50°C.
8. Reconstituted the dry residue with 150 μΙ_ of acetonitrile / 0.1 % formic acid in water (1-1 , v- v).
9. Sealed the plate and mixed for approximately 5 minutes at 1600 rpm on the 96-DW
Eppendorf Mix Mate. Placed the plate in the cooled stack of the auto-sampler until analysis. Injected 15 μΙ_ into the LC-MS/MS.
Skin
In addition, two skin biopsies (A and B) from the application (skin treated with composition containing test item (not the placebo cream)) were collected at necropsy from each animal. Skin samples were placed in FastPrep tubes and immediately frozen on dry ice and stored at -65°C or below until bioanalysis.
Skin tissue was put into a Lysing Matrix D tube (for 2 ml_). An adequate volume of acetonitrile was added to obtain a final dilution of 5 (1 g of tissue is considered to be equal to 1 ml_; 1 ml_ of acetonitrile was considered to be equal to 1 g). The skin tissue samples were homogenized at 4.0 M/s for 20s (2 cycles) using Fast Prep homogenizer and then
centrifuged for 10 minutes at 10000 rpm (5417R Eppendorf centrifuge) at 4°C.
For unknown, blank and zero samples:
1. Added 200 μΙ_ of acetonitrile / 0.1 % formic acid in water (1-1 , v-v) into a 96 deep well plate.
2. Added 25 μΙ_ of skin homogenate.
3. Added 25 μΙ_ of internal standard working solution [D8]compound of formula (I) working solution in acetonitrile/water (7:3, v/v). For Blank sample, added 25 μΙ_ of acetonitrile/water (7:3, v/v) in replacement.
4. Sealed the plate and mixed for approximately 2 minutes at 1400 rpm on the 96-DW Mix Mate.
5. Placed the plate in the cooled stack of the auto-sampler until analysis. Inject 3 μΙ_ into LC- MS/MS system.
For Standards in acetonitrile:
1. Added 200 μΙ_ of acetonitrile / 0.1 % formic acid in water (1-1 , v-v) into a 96 deep well plate.
2. Added 25 μΙ_ of the corresponding compound of formula (I) working solution in
acetonitrile/water (7:3, v/v).
3. Added 25 μΙ_ of internal standard [D8]compound of formula (I) working solution in acetonitrile/water (7:3, v/v).
4. Sealed the plate and mixed for approximately 2 minutes at 1400 rpm on the 96-DW Mix Mate. 5. Placed the plate in the cooled stack of the auto-sampler until analysis. Injected 3 μΙ_ into LC-MS/MS system.
Analytical methods:
Plasma samples were extracted using protein precipitation and analyzed by LC-MS/MS using ElectroSpray Ionization (ESI) in positive ion mode according LC-MS/MS-method 2. Skin samples were homogenized using Fast Prep with acetonitrile and analyzed by LC- MS/MS using ElectroSpray Ionization (ESI) in positive ion mode according LC-MS/MS- method 3.
LLOQ and ULOQ:
-Plasma: 0.0400 ng/mL and 20.0 ng/mL using 50 of plasma
-Skin: 5.00 ng/g and 5000 ng/g using 25 of skin homogenate
Table 6.2: Testing of pharmaceutical compositions in minipig assay, results
Figure imgf000038_0001
7. Crystallization of compound of formula (I) by heating and cooling in ethanol
The compound of formula (I) in amorphous form was heated in ethanol to reflux for 1 h and a clear solution was formed. This solution was cooled to room temperature. The final suspension was filtered and solid was dried at 50 degree in vacuum to afford compound of formula (I) in crystalline form with a yield of 71 % and an HPLC purity >98% (HPLC - method 1).
The crystalline form is not solvated, slightly hygroscopic and characterized by a melting at 184°C with an enthalpy of 66.5 J/g, and a glass transition temperature at around 89-97°C. The 1 H NMR conformed to the one of the compound of formula (I) in amorphous form. LCMS (method 4): 507.1 [M-H], 509.1 [M+H].
Table 7.1 : List of most significant 2-Theta peaks from X-ray Powder Diffraction Pattern with tolerances ±0.5 of compound of formula (I) in crystalline form (including low/weak peaks for information). 2-Theta in degree Intensity [%]
3.97 92.3
7.96 100.0
9.1 1 16.1
11.90 23.3
13.79 14.3
14.40 33.4
15.1 1 13.4
17.05 12.9
19.22 42.5
19.66 22.6
21.61 17.0
22.71 24.9
23.74 26.3
26.03 12.4
30.89 11.1
Other crystalline forms of the compound of formula (I)
Based on equilibration in different media at 25°C and 50°C, as well as crystallization and precipitation experiments from different media indicate that the crystalline form of the compound of formula (I) according to the invention, is the most stable form so far identified. Although there may be indications of other crystalline forms from these experiments, attempts to isolate and characterize such forms failed.
Comparison of aqueous solubility of the compound of formula (I) in amorphous form with the compound of formula (I) in crystalline form:
Thermodynamic solubility was determined by the shake flask method. Compound of formula (I) was weighed precisely into a glass vial. Medium was added and the sample was kept stirred using a magnetic stirrer for 24h at ambient conditions. After 24h excess compound of formula (I) was removed by filtration using 0.4micron PVDC filter tube. The content of the solution was quantified by HPLC (HPLC-method 1).
Table 7.2: Aqueous solubility of the compound of formula (I)
compound of formula (I), compound of formula (I), amorphous form crystalline form
Solubility [mg/ml] (pH final)
pH 4 <0.01 (4.0) 0.03 (4.6)
pH 7.4 O.01 (7.4) 0.02 (7.4)
Water O.01 (6.8) 0.02 (6.8) The compound of formula (I) in crystalline form shows a slightly better solubility compared to the compound of formula (I) in amorphous form. Aqueous solubility was nevertheless found to be low. Comparison of solubility of the compound of formula (I) in amorphous form with the compound of formula (I) in crystalline form in excipients:
Thermodynamic solubility was determined by the shake flask method. Compound of formula (I) was weighed precisely into a glass vial. Medium was added and the sample was kept stirred using a magnetic stirrer for 24h at ambient conditions. After 24h excess compound of formula (I) was removed by filtration using 0.4micron PVDC filter tube. The content of the solution was quantified by HPLC (HPLC-method 1).
Table 7.3: Excipient solubility of the compound of formula (I)
Figure imgf000040_0001
Decreased solubility was found for the compound of formula (I) in crystalline form compared to the compound of formula (I) in amorphous form. Overall organic solubilities were found to be low to moderate.
Comparison of photostability of the compound of formula (I) in amorphous form with the compound of formula (I) in crystalline form:
For determination of light stability 5-1 Omg of compound of formula (I) were dissolved in ethanol. Solutions (0.5ml) were transferred into polypropylene vials and then exposed to light using a Xenon lamp with a cut-off at 290 nm. Light exposure followed ICH guidelines and are described in the Table 7.4 below. Samples were kept at 15°C during light exposure. After light exposure samples were analyzed by HPLC (HPLC-method 1), and compared to a non- exposed sample held at the same conditions. Table 7.4: Photostability of the compound of formula (I)
Figure imgf000041_0001
when originating from the amorphous form upon exposure to light. Repetition of the experiment using compound of formula (I) in crystalline form reduced the degree of degradation.

Claims

1. A topical pharmaceutical composition in the form of an oil-in-water cream
comprising up to 0.35 wt.%, based on the total weight of the composition, of a compound of formula (I)
Figure imgf000042_0001
an emulsifier, and:
from 0.4 to 2.9 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol, and phenylethyl alcohol; or
from 0.01 to 0.6 wt.%, based on the total weight of the composition, of an
antimicrobial preservative selected from the group of parabens.
2. A topical pharmaceutical composition according to claim 1 , comprising from 0.05 to 0.35 wt.% of a compound of formula (I).
3. A topical pharmaceutical composition according to claim 1 or 2, comprising from 0.8 to 2.3 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol, and phenylethyl alcohol; or from 0.05 to 0.3 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of parabens.
4. A topical pharmaceutical composition according to claim 1 or 2, comprising from 0.8 to 2.3 wt.%, based on the total weight of the composition, of an antimicrobial preservative selected from the group of benzyl alcohol, phenoxyethanol, and phenylethyl alcohol.
5. A topical pharmaceutical composition according to any one of claims 1 to 4, comprising from 10 to 20 wt.%, based on the total weight of the composition, of medium chain triglycerides.
6. A topical pharmaceutical composition according to any one of claims 1 to 5, comprising from 2 to 30 wt.%, based on the total weight of the composition, of diethylene glycol monoethylether.
7. A topical pharmaceutical composition according to any one of claims 1 to 6, comprising from 0.7 to 1.5 wt.%, based on the total weight of the composition, of macrogol 21 stearyl ether and from 1.5 to 2.5 wt.%, based on the total weight of the composition, of glycerol monostearate 40-55.
8. A topical pharmaceutical composition according to any one of claims 1 to 7, comprising from 3.0 to 5.0 wt.%, based on the total weight of the composition, of cetyl alcohol and from 3.0 to 5.0 wt.%, based on the total weight of the composition, of stearyl alcohol.
9. A topical pharmaceutical composition according to any one of claims 1 to 8, comprising a buffering agent chosen so that the final pH value of the composition is from pH 5.2 to pH 5.6.
Figure imgf000043_0001
11. The crystalline form according to claim 10, characterized by an X-Ray powder diffraction pattern comprising the following peaks given at degrees 2-Theta ±0.5 degrees: 3.97, 7.96, 19.22, 14.40.
12. The crystalline form according to claim 10, having a X-ray diffraction spectrum substantially the same as the X-ray powder diffraction spectrum shown in Figure 1.
The crystalline form according to claim 10, having a differential scanning calorimetry substantially the same as that shown in Figure 2.
14. The crystalline form according to any one of claims 10 to 13, for use as a pharmaceutical.
15. A combination comprising a therapeutically effective amount of a crystalline form according to claims 10 to 13, and one or more therapeutically active agents.
16. A process to prepare the topical pharmaceutical composition according to any one of claims 1 to 9, comprising the steps of
(a) Preparing a solution/suspension of the compound of formula (I) in a suitable solvent;
(b) Preparing a melt of the remaining solvent components of the pharmaceutical composition not including water, the emulsifier and the antimicrobial preservative and optionally the thickening agent;
(c) Adding the solution/suspension of step (a) to the melt of step (b) to obtain a melt containing the compound of formula (I);
(d) Preparing the aqueous phase;
(e) Mixing the melt of step (c) with the aqueous phase of step (d) and cooling the mixture down to obtain the oil-in-water cream.
17. A process according to claim 16, wherein the compound of formula (I) used in step (a) is in crystalline form according any one of claims 10 to 13.
18. A composition according to claims 1 to 9, wherein the composition is prepared from the compound of formula (I) in crystalline form according to any one of claims 10 to 13.
19. A composition according to claims 1 to 9 or 18 for use in the treatment or prevention of a disease or disorder selected from non-melanoma skin cancers such as basal cell carcinoma and squamous cell carcinoma (SCC); their pre-malignant stages such as squamous cell carcinoma in situ (SCCis), actinic keratosis (AK), solar keratosis and chronically sun damaged skin; and other hyperproliferative skin disorders caused by dysregulation of skin fibroblasts such as skin fibrosis, scleroderma, hypertrophic scars or keloids.
20. A composition according to claims 1 to 9 or 18 for use in the treatment of cutaneous squamous cell carcinoma in situ (cSCCis).
21. A crystalline form according to claims 10 to 13 for use in the treatment or prevention of a disease or disorder selected from non-melanoma skin cancers such as basal cell carcinoma and squamous cell carcinoma (SCC); their pre-malignant stages such as squamous cell carcinoma in situ (SCCis), actinic keratosis (AK), solar keratosis and chronically sun damaged skin; and other hyperproliferative skin disorders caused by dysregulation of skin fibroblasts such as skin fibrosis, scleroderma, hypertrophic scars or keloids.
22. A crystalline form according to claims 10 to 13 for use in the treatment of cutaneous squamous cell carcinoma in situ (cSCCis).
PCT/IB2018/058677 2017-11-06 2018-11-05 Formulation comprising oxazolidin-2-one-pyrimidine derivative WO2019087158A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN201711039501 2017-11-06
IN201711039501 2017-11-06

Publications (2)

Publication Number Publication Date
WO2019087158A2 true WO2019087158A2 (en) 2019-05-09
WO2019087158A3 WO2019087158A3 (en) 2019-06-20

Family

ID=64316605

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2018/058677 WO2019087158A2 (en) 2017-11-06 2018-11-05 Formulation comprising oxazolidin-2-one-pyrimidine derivative

Country Status (2)

Country Link
TW (1) TW201929861A (en)
WO (1) WO2019087158A2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014072956A1 (en) 2012-11-12 2014-05-15 Novartis Ag Oxazolidin-2-one-pyrimidine derivatives

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014072956A1 (en) 2012-11-12 2014-05-15 Novartis Ag Oxazolidin-2-one-pyrimidine derivatives

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
G. F. GERBERICK, METHODS, vol. 41, 2007, pages 54 - 60
J. SCHUEMANN, TOXICOLOGICAL SCIENCES, vol. 139, no. 1, 2014, pages 245 - 256
T. J. FRANZ, J INVEST DERM, vol. 64, no. 3, 1975, pages 190 - 195

Also Published As

Publication number Publication date
WO2019087158A3 (en) 2019-06-20
TW201929861A (en) 2019-08-01

Similar Documents

Publication Publication Date Title
CA2949614C (en) Topical pharmaceutical formulations comprising crisaborole
US11865113B2 (en) Methods of treating a patient afflicted with non-alcoholic steatohepatitis (NASH)
US20100173965A1 (en) Antifungal composition
US8680054B2 (en) Suspension type topical formulations comprising cyclic depsipeptide
WO2019052133A1 (en) Crystal form of gsk1278863 and preparation method and pharmaceutical use thereof
EA027869B1 (en) Stabilized tacrolimus composition
US8722672B2 (en) Topical pharmaceutical composition including rel-N-[6-[(2R,6S)-2,6-dimethyl-4-morpholinyl]-3-pyridinyl]-2-methyl-4′-(trifluoromethoxY)-[1,1′-biphenyl]-3-carboxamide
US20190105392A1 (en) Caspofungin Acetate Formulations
WO2007117581A2 (en) Stabilized transdermal bupropion preparations
WO2019087158A2 (en) Formulation comprising oxazolidin-2-one-pyrimidine derivative
US20160136148A1 (en) Solid dispersions of amorphous paroxetine mesylate
JP6625208B2 (en) Transdermal formulation
JP2023540664A (en) IRAK4 inhibitors and methods of their topical use
EP4268825A1 (en) Pharmaceutical composition for transdermal administration containing epinastine or salt thereof and containing sulfur-based antioxidant
EA016861B1 (en) Stable formulations of thiadiazole derivative
NZ615551B2 (en) Suspension type topical formulations comprising cyclic depsipeptide

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18803787

Country of ref document: EP

Kind code of ref document: A2