US20140057982A1 - Retinoids and use thereof - Google Patents

Retinoids and use thereof Download PDF

Info

Publication number
US20140057982A1
US20140057982A1 US13/932,294 US201313932294A US2014057982A1 US 20140057982 A1 US20140057982 A1 US 20140057982A1 US 201313932294 A US201313932294 A US 201313932294A US 2014057982 A1 US2014057982 A1 US 2014057982A1
Authority
US
United States
Prior art keywords
substituted
composition
group
branched
straight chain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/932,294
Inventor
Wayne J. Brouillette
Donald D. Muccio
Venkatram Reddy Atigadda
John M. Ruppert
Susan M. Ruppert
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
UAB Research Foundation
Original Assignee
UAB Research Foundation
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 UAB Research Foundation filed Critical UAB Research Foundation
Priority to US13/932,294 priority Critical patent/US20140057982A1/en
Assigned to THE UAB RESEARCH FOUNDATION reassignment THE UAB RESEARCH FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: REDDY, ATIGADDA VENKATRAM, RUPPERT, JOHN, MUCCIO, DONALD D., RUPPERT, SUSAN, BROUILLETTE, WAYNE J.
Publication of US20140057982A1 publication Critical patent/US20140057982A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF ALABAMA AT BIRMINGHAM
Assigned to NATIONAL INSTITUTE OF HEALTH - DIRECTOR DEITR reassignment NATIONAL INSTITUTE OF HEALTH - DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF ALABAMA AT BIRMINGHAM
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/07Retinol compounds, e.g. vitamin A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/202Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids having three or more double bonds, e.g. linolenic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/203Retinoic acids ; Salts thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • 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
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/08Antiseborrheics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/12Keratolytics, e.g. wart or anti-corn preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/16Emollients or protectives, e.g. against radiation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Retinoid receptors and other nuclear receptors that include the steroid, thyroid and vitamin D hormone receptors and other “orphan” receptors without known ligands
  • retinoic acid (RA) and synthetic retinoids act as ligand-dependent transcription factors with different members of nuclear retinoid receptors to control gene transcription responsible for cellular proliferation, differentiation, development and cell death.
  • RARs and RXRs Two classes of nuclear retinoid receptors (RARs and RXRs) have been identified so far, and each has several different subtypes ( ⁇ , ⁇ , ⁇ ).
  • RAR-selective compounds were known to induce skin hyperplasia, an indicator of retinoid activity in the skin
  • RXR-selective compounds were previously shown not to be effective inducers of epidermal hyperplasia, considering them silent partners in the RAR-RXR heterodimer with unlikely clinical utility in non-neoplastic skin conditions. See, for example, Thacher, S.
  • the present invention fulfills a need for new and beneficial uses of retinoid compounds, particularly RXR-selective compounds, for non-neoplastic skin conditions or disorders, as previously not described or realized.
  • the present invention concerns certain RXR-selective retinoid compounds, including analogs, derivatives or isomers of these retinoid compounds, and their novel use in methods for treating non-neoplastic dermal conditions or disorders, including dermal inflammatory conditions.
  • the retinoid compounds can be formulated with pharmaceutically acceptable adjuvants, carriers, excipients, or vehicles to provide pharmaceutical compositions useful in the treatment of dermal conditions or disorders.
  • These pharmaceutical compositions can be in the form of oral dosage forms, parenteral (e.g., injectable) dosage forms, or formulated as topical compositions.
  • UAB30 Preferred retinoid compounds useful in accordance with the subject invention are designated as UAB30, or analogs, derivatives or isomers of those compounds.
  • Alternative processes for making certain retinoid compounds, including UAB30 or its analogs, as well as compositions comprising those compounds and methods of use, are described in US Patent Application, Publ'n No. 2010/0204327, which is also incorporated herein by reference, in its entirety.
  • a method of treating, preventing or ameliorating a dermal condition or disorder such dermatological afflictions associated with excessive skin sebum production (e.g. acne vulgaris) cell differentiation and/or proliferation disorders, (e.g. psoriasis, actinic keratosis, seborrheic dermatitis, warts, hair loss), keratinization disorders (e.g. keratosis, xerosis, icthyosis, lichen, keratoderma, folliculitis), skin pigmentation disorders (e.g.
  • dermatological afflictions associated with excessive skin sebum production e.g. acne vulgaris
  • proliferation disorders e.g. psoriasis, actinic keratosis, seborrheic dermatitis, warts, hair loss
  • keratinization disorders e.g. keratosis, xerosis, icthyosis, lichen, kerato
  • vitiligo melasma, acnitic lentigines
  • skin inflammatory disorders e.g. acne, rosacea, eczema
  • conditions resulting from photodamage e.g. such photo induced or chronological skin aging
  • an effective dose of an RXR-selective retinoid compound e.g., UAB30, or a composition comprising the compound.
  • the method comprises treating an individual in need of treatment for acne vulgaris, psoriasis, actinic keratosis or skin photo-induced damage.
  • the retinoid compounds can be used in combination with one or more other retinoid compounds, or with one or more other non-retinoid compounds, for the treatment.
  • the subject invention concerns a method for treating, preventing or lessening the severity of a non-neoplastic skin condition or disorder in a patient in need thereof, comprising the step of administering to said patient a retinoid compound, or a composition comprising a retinoid compound, having the formula selected from Formula I or Formula II, below:
  • R 1 is H or a C 1 -C 5 or greater branched or straight chain alkyl group
  • R 2 is H, a C 1 -C 15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group, or
  • R 1 and R 2 collectively form a substituted or unsubstituted fused aryl group
  • R 3 is one or more groups comprising, independently, H, CF 3 , a C 1 -C 15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
  • R 6 and R 7 are, independently, H, CF 3 , a C 1 -C 15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R 11 is H, or a C 1 -C 15 branched or straight chain alkyl group.
  • n is from 0 to 3;
  • the method concerns the use of a compound having the formula III, below:
  • R 3 is one or more groups comprising, independently, H, F, diflouro, CF 3 , a C 1 -C 15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
  • R 6 and R 7 are, independently, H, CF 3 , a C 1 -C 15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R 9 is one or more groups comprising, independently, H, F, CF 3 , a C 1 -C 15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R 11 is H, or a C 1 -C 15 branched or straight chain alkyl group
  • n is from 0 to 3;
  • a most preferred embodiment of the subject method comprises the use of a compound of Formula III is the 9(Z) form having the structure shown as Formula IV, below:
  • Non-neoplastic skin conditions or disorders treated, prevented or ameliorated by a retinoid compound of the invention include, but are not limited to acne, psoriasis, actinic keratosis, actinic lentigines, seborrheic dermatitis, rosacea, keratosis pillaris, warts, vitiligo, melasma, folliculitis, eczema, icthyosis, lichen, xerosis, keratoderma, hirsutism or hair loss.
  • the method of the invention can also be useful in treating, preventing or ameliorating a disorder or condition caused by UV damage such as photo-induced skin damage or chronological skin aging.
  • the method can employ the use of a combination of retinoid compound of the invention with a conventional sun-screening agent.
  • compositions useful for treating, preventing or ameliorating a non-neoplastic dermal condition.
  • Compositions according to the invention comprise, as an active pharmaceutical ingredient (API), at least one retinoid compound as described herein and a pharmaceutically acceptable vehicle or carrier.
  • a composition of the invention comprises carriers or vehicles which are pharmaceutically acceptable for oral and/or topical for dermal use or application. More preferably, the composition comprises the compound described herein, designated UAB 30, or an analog, derivative, isomer or salt thereof.
  • compositions of the invention can comprise two or more active ingredients, wherein the active ingredients can be two or more retinoid compounds, or can include one or more retinoid compound in combination with a non-retinoid compound.
  • the active ingredients can be administered sequentially or concomitantly.
  • a composition of the invention comprises an RXR-selective retinoid compound at therapeutically effective doses as can be determined by those of ordinary skill in the art.
  • a retinoid compound of the invention may be provided as a topically applied composition prepared to comprise a concentration of about 0.01 to about 10% of a retinoid compound of the invention as the active pharmaceutical ingredient (API). More preferably, the concentration of retinoid compound in a topically applied composition according to the invention is about 0.025% to about 5%, and most preferably between about 0.1% to about 3% of the composition (w/w in solid and semi-solid dosage forms and w/v in liquid dosage forms).
  • a retinoid compound of the invention may be provided as an orally administered composition prepared to comprise an amount of about 1 mg to about 600 mg of a retinoid compound of the invention as the active pharmaceutical ingredient (API). More preferably, the amount of retinoid compound in an oral composition according to the invention is about 5 mg to about 300 mg, and most preferably between about 20 mg to about 200 mg of the composition.
  • API active pharmaceutical ingredient
  • the present invention is directed to RXR-selective retinoid compounds, e.g., UAB30, as disclosed herein, and its novel use in treating dermal conditions or disorders.
  • retinoid compounds e.g., UAB30 and its analogs or isomers, having the general structures selected from the group of Formula I (a 9(Z) isomer) and Formula II (an all-(E) isomer), as shown below:
  • R 1 is H, CF 3 , or a C1-C5 or greater branched or straight chain alkyl group
  • R 2 is H, CF 3 , a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group, or
  • R 1 and R 2 collectively form a substituted or unsubstituted fused aryl group
  • R 3 is one or more groups comprising, independently, H, F, diflouro, CF 3 , a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
  • R 6 and R 7 are, independently, H, CF 3 , a C1-C15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R 11 is H, or a C1-C15 branched or straight chain alkyl group.
  • n is from 0 to 3;
  • R 1 is an isopropyl or isopentyl alkane, which includes the compound UAB-111.
  • the method concerns the use of a compound wherein R 1 and R 2 in Formulae I or II, above, form a fused aryl group wherein the compound has the formula III, below:
  • R 3 is one or more groups comprising, independently, H, F, diflouro, CF 3 , a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
  • R 6 and R 7 are, independently, H, CF 3 , a C1-C15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R 9 is one or more groups comprising, independently, H, F, CF 3 , a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R 11 is H, or a C1-C15 branched or straight chain alkyl group.
  • n is from 0 to 3;
  • a most preferred embodiment is a compound of Formula III, in the 9(Z) form, having the structure shown as Formula IV, below:
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. In chemical descriptions used herein, “E” may be interchanged synonymously with “trans”, and “Z” may be interchanged synonymously with “cis”.
  • the present invention is directed to a method of treating an individual having a dermal condition or disorder, in which retinoids and its derivatives may be useful.
  • the compound is administered at a dosage range of from about 0.001 mg/cm 2 to about 10 mg/cm 2 of skin surface, and more preferably between about 0.005 mg/cm 2 to about 2 mg/cm 2 of skin surface when applied topically.
  • compounds used in accordance with the subject invention may also be useful when administered orally.
  • Dosage ranges are preferably from about 0.01 mg/kg to about 300 mg/kg of body weight, and more preferably from about 0.1 mg/kg to about 100 mg/kg of body weight, when administered orally.
  • Compounds of Formula III are especially contemplated as applicable for oral dosing in the treatment of non-neoplastic skin conditions or disorders.
  • a method of treating an individual having a dermal condition or disorder by administering to the individual a combination of two or more than two retinoid compounds disclosed herein.
  • the combination may further comprise one or more retinoid compound as described herein, and one or more additional active compounds that are not retinoid compounds as disclosed herein.
  • skin conditions can include dermatological afflictions associated with excessive skin sebum production (e.g. acne vulgaris) cell differentiation and/or proliferation disorders, (e.g. psoriasis, actinic keratosis, seborrheic dermatitis, warts, hair loss) keratinization disorders (e.g. keratosis, xerosis, Icthyosis, lichen, keratoderma, folliculitis), skin pigmentation disorders (e.g.
  • dermatological afflictions associated with excessive skin sebum production e.g. acne vulgaris
  • proliferation disorders e.g. psoriasis, actinic keratosis, seborrheic dermatitis, warts, hair loss
  • keratinization disorders e.g. keratosis, xerosis, Icthyosis, lichen, keratoderma, folliculitis
  • skin pigmentation disorders e
  • vitiligo e.g. acne, rosacea, eczema
  • skin inflammatory disorders e.g. acne, rosacea, eczema
  • conditions resulting from photodamage e.g. such photo induced or chronological skin aging
  • a compound can be combined with at least one pharmaceutically-acceptable carrier to produce a pharmaceutical composition, prepared using techniques known in the art.
  • a composition is prepared by admixing the compound with a pharmaceutically-acceptable carrier. Depending upon the components to be admixed, the components may or may not chemically or physically interact with one another.
  • compositions that include at least one compound of the present invention, together with one or more pharmaceutically acceptable carriers, including excipients, such as diluents, binders and the like, and can further include additives, such as stabilizing agents, preservatives, solubilizing agents, and buffers, as desired.
  • pharmaceutically acceptable carriers including excipients, such as diluents, binders and the like, and can further include additives, such as stabilizing agents, preservatives, solubilizing agents, and buffers, as desired.
  • compositions may include carriers, thickeners, diluents, buffers, preservatives, surface active agents and the like, in addition to the active ingredient.
  • Pharmaceutical compositions may also include more than one active ingredient, such as a retinoid compound according to the invention, and one or more antimicrobial agents, keratolytic agents, benzoyl peroxide, antiinflammatory agents, corticosteroids, or the like.
  • Pharmaceutically-acceptable carriers are known to those skilled in the art. These most typically would be standard carriers for administration to humans, including solutions such as sterile water, saline, and buffered solutions at physiological pH.
  • the pharmaceutical composition may be formulated for administration in a number of ways depending on whether local or systemic treatment is desired, or depending on the area to be treated. Administration can be topical (applied to the surface of the skin), or as an injectable solution or suspension, preferably for subcutaneous injection. Alternatively, administration can be oral, employing a dosage form such as a tablet or capsule, as would be readily understood by persons of ordinary skill in the art.
  • a provided compound of the present invention can be combined as the active ingredient in an admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier used can have different forms depending on the dosage form desired for administration, for example, oral, parenteral, dermal or topical, transdermal, buccal, or the like.
  • a provided compound may be appropriately buffered by means of saline, acetate, phosphate, citrate, acetate or other buffering agents, which may be at any physiologically acceptable pH, generally from about pH 4 to about pH 7.
  • buffering agents may also be employed, such as phosphate buffered saline, a saline and acetate buffer, and the like.
  • a 0.9% saline solution may be employed.
  • a 50 mM solution may be employed.
  • a suitable preservative may be employed, to prevent or limit bacteria and other microbial growth.
  • One such preservative that may be employed is 0.05% benzalkonium chloride.
  • Preparations for administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions.
  • aqueous or non-aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Formulations for topical administration may include ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. It will be expected that the actual preferred amounts of active compound in a specified case will vary according to the specific compound being utilized, the particular compositions formulated, the mode of application, and the particular situs and mammal being treated.
  • typical pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets.
  • oral liquid preparations such as, for example, suspensions, elixirs and solutions
  • carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets.
  • any of a variety of thickening, filler, bulking and carrier additives may be employed, such as starches, sugars, fatty acids and the like.
  • Formulation excipients may include polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol, mannitol, sodium chloride and sodium citrate.
  • any of a variety of measured-release, slow-release or time-release formulations and additives may be employed, so that the dosage may be formulated so as to effect delivery of a provided compound over a period of time, commonly referred to as controlled, delayed, extended, slow, or sustained, release formulations.
  • dosage may be formulated so as to effect delivery of a provided compound over a period of time, commonly referred to as controlled, delayed, extended, slow, or sustained, release formulations.
  • gelatin, sodium carboxymethylcellulose and/or other cellulosic excipients may be included to provide time-release or slower-release formulations, especially for administration by subcutaneous and intramuscular injection.
  • the formulation may be such that an application, administration, or injection is required on a daily, weekly, monthly or other periodic basis, depending on the concentration and amount of a provided compound, the biodegradation rate of a polymer used in the formulation, and other factors known to those of skill in the art.
  • Dosages for a given host can be determined using conventional considerations, e.g. by customary comparison of the differential activities of the subject compounds and of a known agent, e.g., by means of an appropriate conventional pharmacological protocol. Physicians and formulators, skilled in the art of determining doses of pharmaceutical compounds, will have no problems determining dose according to standard recommendations (Physician's Desk Reference, Barnhart Publishing (1999).
  • UV/vis spectra were recorded on an AVIV 14DS spectrophotometer in cyclohexane or methanol solutions (Fisher, Spectrograde). IR spectra were recorded using a Nicolet FT IR spectrometer on thin films. HPLC separations were performed on a Gilson HPLC gradient system using 25-ml pump heads and an ISCO V.sup.4 variable wavelength detector. The column employed was a Whatman Partisil 10 M20/50 (500 ⁇ 22-mm i.d.) with a flow rate of 5 ml/min and monitoring by UV/vis detection at 340 nm.
  • This preparation employed a suspension of NaH (0.050 g, 2.1 mmol) in dry THF (1 ml), a solution of triethyl phosphonoseneciaote (0.11 g, 0.40 mmol) in dry THF (1 ml), HMPA (0.048 g, 0.22 mmol), and a solution of (all-E)-4 (0.065 g, 0.31 mmol) in dry THF (1 ml) to give a 2:1 mixture of esters (all-E) and (13)-5 (0.12 g, 78% yield).
  • This mixture was separated by HPLC on silica gel using 1% Et.sub.2 O, 0.5% THF in hexane.
  • This preparation utilized a solution of KOH (0.258 g, 4.61 mmol) in water (2 ml) and a warm solution of ester (all-E)-5 (0.122 g, 0.378 mmol) in methanol (10 ml) to provide acid (all-E)-UAB30 (0.104 g, 93% yield) as a yellow solid: mp 192-197° C. (cyclohexane).
  • This preparation utilized a solution of KOH (0.15 g, 2.8 mmol) in water (1 ml) and a warm solution of ester (9Z)-5 (0.073 g, 0.23 mmol) in methanol (3 ml) to give acid (9Z)-UAB30 (0.065 g, 98% yield) as a yellow solid: mp 182-185° C. (cyclohexane).
  • This preparation employed a solution of KOH (0.085 g, 1.50 mmol) in water (1 ml) and a warm solution of ester (13Z)-5 (0.040 g, 0.12 mmol) in methanol (4 ml) to give acid (13Z)-UAB30 (0.034 g, 93% yield) as a yellow solid: mp 180-185° C. (cyclohexane).
  • the stimulation the keratinocyte proliferation (reported histologically as epidermal hyperplasia), is believed to be due to increase in the epidermal growth factor receptor activation and is known to be reflective of retinoid effect (Fisher G., Vorhees J., Molecular mechanism of retinoid actions in skin. FASEB J. 1996; 10: 1002-1013).
  • RAR-selective retinoids are associated with significant skin irritation.
  • RXR-selective retinoids have been reported not to efficiently induce epidermal hyperplasia.
  • the RXR-selective compound of the subject invention were identified here as exhibiting keratinocyte proliferation effects comparable to a potent RAR-selective retinoid (tazarotene).
  • UAB30, 4Me-UAB30 and UAB111 at 0.1% and 0.3%, tazarotene at 0.1% and vehicle were applied topically on approximately 8 cm2 (2 ⁇ 4 cm) of the dorsal skin (100 ⁇ L) and on the right ear (20 ⁇ L).
  • the degree of epidermal hyperplasia induced by UAB30 was comparable to the hyperplasia induced by the potent RAR retinoid tazarotene, 0.1%, that reported an average of 2.
  • 4Me-UBA30 0.1% and 0.3% and UAB111 0.1% and 0.3% treatments caused comparable epidermal hyperplasia ranging from 2.2 to 2.4 in the 5 point scale.
  • Utriculus formation and sebaceous gland hyperplasia normally present in this mice strain, was also mildly reduced with all the active treatments.
  • MPO Myeloperoxidase
  • test compounds were UAB30, 4Me-UAB30 and UAB111.
  • Tazarotene (0.1% cream) was purchased by Ricerca Taiwan Ltd.
  • vehicle (70% ethanol/30% propylene glycol) were each administered topically on the back (100 ⁇ L) and on the right ear (20 ⁇ L) once daily for two weeks.
  • mice Female SKH1-E mice weighing 22 ⁇ 2 g, were provided by Charles River, USA. The animals were housed in Individually Ventilated Cages Racks (IVC Racks, 36 Mini Isolator systems) under clean area throughout the experiment. Every 2 or 3 mice were kept in an animal cage (in cm, 26.7 length ⁇ 20.7 width ⁇ 14.0 height) and maintained under controlled temperature (20-24° C.) and humidity (50%-80%) with 12-hour light/dark cycles. The animals were given free access to sterilized standard lab chow [MF-18 (Oriental Yeast Co., Ltd. Japan)] and sterile tap water ad libitum. All aspects of this work, i.e., housing, experimentation and disposal of animals, were performed in general accordance with the Guide for the Care and Use of Laboratory Animals (National Academy Press, Washington, D.C., 2011).
  • mice Female SKH1-E hairless mice (Crl:SKH1-Hrhr) weighing 22 ⁇ 2 g were used. The animals were divided into groups of 5 each. Once a day, 100 ⁇ L of vehicle, tazarotene, and test articles were applied on the back with a micropipette and smeared evenly with gloved finger on approximately 8 cm2 (2 ⁇ 4 cm) of the dorsal skin; 20 ⁇ L was applied to the right ear. The tested animals were treated for two consecutive weeks. Body weight was monitored twice a week.
  • Epidermal hyperplasia on the dorsal skin was used as a marker of retinoid activity.
  • mice were sacrificed on day 15. The dorsal skin was dissected and stretched to the natural size on the filter. Punch biopsies (ED 8 mm) were obtained, fixed in 10% buffered formalin, and embedded in paraffin. Five micro thick sections were cut, stained with H&E, and a representative section of each biopsy was selected for histological evaluation.
  • the thickness of the epidermis and dermis was measured using a microscope with a micrometer in the eyepiece. The details are described in the histopathology report (see d. Histopathology, below).
  • Erythema, desquamation (skin flaking), and skin lesions/abrasion on dorsal skin were assessed visually in individual animals at the end of a 2-week treatment and scored on a scale of 0 to 4+, with 4+ being maximal.
  • the dorsal skin was dissected and stretched to the natural size on the filter.
  • Punch biopsies (ED 8 mm) were obtained, fixed in 10% buffered formalin, and embedded in paraffin. Five micro thick sections were cut, stained with H&E, and a representative section of each biopsy was selected for histological evaluation.
  • Right ear was harvested on day 15 for measurements of myeloperoxidase activity by mouse MPO ELISA kit, a measure of neutrophil infiltrates.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Dermatology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Emergency Medicine (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Toxicology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Birds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention provides new retinoid compounds and uses of the compounds in humans and animals for non-neoplastic dermal or inflammatory conditions or disorders.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This is a continuation of U.S. patent application, Ser. No. 13/594,177, filed Aug. 24, 2012 and issuing as U.S. Pat. No. 8,475,775 on Jul. 2, 2013.
  • ACKNOWLEDGMENTS
  • The research leading to this invention was funded in part by the National Institutes of Health, Grant No. 5 P50 CA 89019. The U.S. Government may have certain rights in this invention.
  • BACKGROUND OF THE INVENTION
  • Retinoid receptors and other nuclear receptors (that include the steroid, thyroid and vitamin D hormone receptors and other “orphan” receptors without known ligands) are targets for drug development. It is thought that retinoic acid (RA) and synthetic retinoids act as ligand-dependent transcription factors with different members of nuclear retinoid receptors to control gene transcription responsible for cellular proliferation, differentiation, development and cell death. Two classes of nuclear retinoid receptors (RARs and RXRs) have been identified so far, and each has several different subtypes (α, γ, γ). Both (E)-RA and (9Z)-RA bind to RARs and activate transcription mediated by RAR/RXR heterodimers, but (9Z)-RA is the only known natural ligand for the RXRs which mediate transcription by forming homodimers or heterodimers.
  • Recent advances in chemoprevention have heightened interest in the use of RXR-selective retinoids in several types of solid organ tumors, and major therapeutic successes have been demonstrated with retinoids in certain lymphomas. Bexarotene (a RXR-selective retinoid) is approved for the treatment of Cutaneous T cell Lymphoma.
  • The wide range of benefits from the uses of these retinoid compounds, including uses in treating, preventing or ameliorating non-neoplastic conditions or disorders, have not been fully identified or explored. Although RAR-selective compounds were known to induce skin hyperplasia, an indicator of retinoid activity in the skin, RXR-selective compounds were previously shown not to be effective inducers of epidermal hyperplasia, considering them silent partners in the RAR-RXR heterodimer with unlikely clinical utility in non-neoplastic skin conditions. See, for example, Thacher, S. M., et al., Receptor Specificity of Retinoid-Induced Epidermal Hyperplasia: Effect of RXR-Selective Agonists and Correlation with Topical Irritation, JPET 282:528-534, 1997. The present invention fulfills a need for new and beneficial uses of retinoid compounds, particularly RXR-selective compounds, for non-neoplastic skin conditions or disorders, as previously not described or realized.
  • SUMMARY OF THE INVENTION
  • The present invention concerns certain RXR-selective retinoid compounds, including analogs, derivatives or isomers of these retinoid compounds, and their novel use in methods for treating non-neoplastic dermal conditions or disorders, including dermal inflammatory conditions. The retinoid compounds can be formulated with pharmaceutically acceptable adjuvants, carriers, excipients, or vehicles to provide pharmaceutical compositions useful in the treatment of dermal conditions or disorders. These pharmaceutical compositions can be in the form of oral dosage forms, parenteral (e.g., injectable) dosage forms, or formulated as topical compositions.
  • Preferred retinoid compounds useful in accordance with the subject invention are designated as UAB30, or analogs, derivatives or isomers of those compounds. The compound, UAB30 and its analogs, as well as a description of how to prepare the compounds, are described in U.S. Pat. No. 6,172,112, which is incorporated herein by reference, in its entirety. Alternative processes for making certain retinoid compounds, including UAB30 or its analogs, as well as compositions comprising those compounds and methods of use, are described in US Patent Application, Publ'n No. 2010/0204327, which is also incorporated herein by reference, in its entirety.
  • In one embodiment of the present invention, there is provided a method of treating, preventing or ameliorating a dermal condition or disorder such dermatological afflictions associated with excessive skin sebum production (e.g. acne vulgaris) cell differentiation and/or proliferation disorders, (e.g. psoriasis, actinic keratosis, seborrheic dermatitis, warts, hair loss), keratinization disorders (e.g. keratosis, xerosis, icthyosis, lichen, keratoderma, folliculitis), skin pigmentation disorders (e.g. vitiligo, melasma, acnitic lentigines), and/or skin inflammatory disorders (e.g. acne, rosacea, eczema), conditions resulting from photodamage (e.g. such photo induced or chronological skin aging), in an individual in need of such treatment by administering to the individual an effective dose of an RXR-selective retinoid compound, e.g., UAB30, or a composition comprising the compound.
  • Preferably, the method comprises treating an individual in need of treatment for acne vulgaris, psoriasis, actinic keratosis or skin photo-induced damage. The retinoid compounds can be used in combination with one or more other retinoid compounds, or with one or more other non-retinoid compounds, for the treatment.
  • Thus, the subject invention concerns a method for treating, preventing or lessening the severity of a non-neoplastic skin condition or disorder in a patient in need thereof, comprising the step of administering to said patient a retinoid compound, or a composition comprising a retinoid compound, having the formula selected from Formula I or Formula II, below:
  • Figure US20140057982A1-20140227-C00001
  • wherein
  • R1 is H or a C1-C5 or greater branched or straight chain alkyl group;
  • R2 is H, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group, or
  • R1 and R2 collectively form a substituted or unsubstituted fused aryl group;
  • R3 is one or more groups comprising, independently, H, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
  • R6 and R7 are, independently, H, CF3, a C1-C15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R11 is H, or a C1-C15 branched or straight chain alkyl group.
  • n is from 0 to 3;
  • wherein one or more carbons in the ring structures of Formula I or II can optionally be replaced with a heteroatom;
    or a pharmaceutically acceptable salt or isomer thereof.
  • More preferably, the method concerns the use of a compound having the formula III, below:
  • Figure US20140057982A1-20140227-C00002
  • wherein,
  • R3 is one or more groups comprising, independently, H, F, diflouro, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
  • R6 and R7 are, independently, H, CF3, a C1-C15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R9 is one or more groups comprising, independently, H, F, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R11 is H, or a C1-C15 branched or straight chain alkyl group;
  • n is from 0 to 3;
  • wherein one or more carbons in the ring structures of Formula I or II can optionally be replaced with a heteroatom;
    or a pharmaceutically acceptable salt or isomer thereof.
  • A most preferred embodiment of the subject method comprises the use of a compound of Formula III is the 9(Z) form having the structure shown as Formula IV, below:
  • Figure US20140057982A1-20140227-C00003
  • Non-neoplastic skin conditions or disorders treated, prevented or ameliorated by a retinoid compound of the invention include, but are not limited to acne, psoriasis, actinic keratosis, actinic lentigines, seborrheic dermatitis, rosacea, keratosis pillaris, warts, vitiligo, melasma, folliculitis, eczema, icthyosis, lichen, xerosis, keratoderma, hirsutism or hair loss.
  • The method of the invention can also be useful in treating, preventing or ameliorating a disorder or condition caused by UV damage such as photo-induced skin damage or chronological skin aging. Thus, the method can employ the use of a combination of retinoid compound of the invention with a conventional sun-screening agent.
  • The subject invention further contemplates pharmaceutical compositions useful for treating, preventing or ameliorating a non-neoplastic dermal condition. Compositions according to the invention comprise, as an active pharmaceutical ingredient (API), at least one retinoid compound as described herein and a pharmaceutically acceptable vehicle or carrier. Preferably, a composition of the invention comprises carriers or vehicles which are pharmaceutically acceptable for oral and/or topical for dermal use or application. More preferably, the composition comprises the compound described herein, designated UAB 30, or an analog, derivative, isomer or salt thereof.
  • Compositions of the invention can comprise two or more active ingredients, wherein the active ingredients can be two or more retinoid compounds, or can include one or more retinoid compound in combination with a non-retinoid compound. The active ingredients can be administered sequentially or concomitantly.
  • A composition of the invention comprises an RXR-selective retinoid compound at therapeutically effective doses as can be determined by those of ordinary skill in the art. A retinoid compound of the invention may be provided as a topically applied composition prepared to comprise a concentration of about 0.01 to about 10% of a retinoid compound of the invention as the active pharmaceutical ingredient (API). More preferably, the concentration of retinoid compound in a topically applied composition according to the invention is about 0.025% to about 5%, and most preferably between about 0.1% to about 3% of the composition (w/w in solid and semi-solid dosage forms and w/v in liquid dosage forms).
  • A retinoid compound of the invention may be provided as an orally administered composition prepared to comprise an amount of about 1 mg to about 600 mg of a retinoid compound of the invention as the active pharmaceutical ingredient (API). More preferably, the amount of retinoid compound in an oral composition according to the invention is about 5 mg to about 300 mg, and most preferably between about 20 mg to about 200 mg of the composition.
  • Other and further aspects, features, and advantages of the present invention will be apparent from the following description of the presently preferred embodiments of the invention given for the purpose of disclosure.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to RXR-selective retinoid compounds, e.g., UAB30, as disclosed herein, and its novel use in treating dermal conditions or disorders.
  • In one embodiment of the present invention, there are provided retinoid compounds, e.g., UAB30 and its analogs or isomers, having the general structures selected from the group of Formula I (a 9(Z) isomer) and Formula II (an all-(E) isomer), as shown below:
  • Figure US20140057982A1-20140227-C00004
  • wherein
  • R1 is H, CF3, or a C1-C5 or greater branched or straight chain alkyl group;
  • R2 is H, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group, or
  • R1 and R2 collectively form a substituted or unsubstituted fused aryl group; and
  • R3 is one or more groups comprising, independently, H, F, diflouro, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
  • R6 and R7 are, independently, H, CF3, a C1-C15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R11 is H, or a C1-C15 branched or straight chain alkyl group.
  • n is from 0 to 3;
  • wherein one or more carbons in the ring structures of Formula I or II can otionally be replaced with a heteroatom;
    or a pharmaceutically acceptable salt or isomer thereof.
  • One preferred compound of Formulae I or II above is where R1 is an isopropyl or isopentyl alkane, which includes the compound UAB-111.
  • More preferably, the method concerns the use of a compound wherein R1 and R2 in Formulae I or II, above, form a fused aryl group wherein the compound has the formula III, below:
  • Figure US20140057982A1-20140227-C00005
  • wherein,
  • R3 is one or more groups comprising, independently, H, F, diflouro, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
  • R6 and R7 are, independently, H, CF3, a C1-C15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R9 is one or more groups comprising, independently, H, F, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, or a substituted or unsubstituted cycloalkyl group;
  • R11 is H, or a C1-C15 branched or straight chain alkyl group.
  • n is from 0 to 3;
  • wherein one or more carbons in the ring structures of Formula I or II can optionally be replaced with a heteroatom;
    or a pharmaceutically acceptable salt or isomer thereof.
  • A most preferred embodiment is a compound of Formula III, in the 9(Z) form, having the structure shown as Formula IV, below:
  • Figure US20140057982A1-20140227-C00006
  • Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. In chemical descriptions used herein, “E” may be interchanged synonymously with “trans”, and “Z” may be interchanged synonymously with “cis”.
  • In another embodiment, the present invention is directed to a method of treating an individual having a dermal condition or disorder, in which retinoids and its derivatives may be useful. Preferably, the compound is administered at a dosage range of from about 0.001 mg/cm2 to about 10 mg/cm2 of skin surface, and more preferably between about 0.005 mg/cm2 to about 2 mg/cm2 of skin surface when applied topically.
  • Advantageously, compounds used in accordance with the subject invention may also be useful when administered orally. Dosage ranges are preferably from about 0.01 mg/kg to about 300 mg/kg of body weight, and more preferably from about 0.1 mg/kg to about 100 mg/kg of body weight, when administered orally. Compounds of Formula III are especially contemplated as applicable for oral dosing in the treatment of non-neoplastic skin conditions or disorders.
  • In yet another embodiment of the present invention, there is provided a method of treating an individual having a dermal condition or disorder by administering to the individual a combination of two or more than two retinoid compounds disclosed herein. The combination may further comprise one or more retinoid compound as described herein, and one or more additional active compounds that are not retinoid compounds as disclosed herein.
  • Representative dermal conditions which can be treated in accordance with the subject invention include, but are not limited to epithelial- or skin-related conditions or disorders. In some embodiments, skin conditions can include dermatological afflictions associated with excessive skin sebum production (e.g. acne vulgaris) cell differentiation and/or proliferation disorders, (e.g. psoriasis, actinic keratosis, seborrheic dermatitis, warts, hair loss) keratinization disorders (e.g. keratosis, xerosis, Icthyosis, lichen, keratoderma, folliculitis), skin pigmentation disorders (e.g. vitiligo, melasma, actinic lentigines), and/or skin inflammatory disorders (e.g. acne, rosacea, eczema) and/or conditions resulting from photodamage (e.g. such photo induced or chronological skin aging).
  • Any of the compounds described herein can be formulated into a pharmaceutical composition. In one aspect, a compound can be combined with at least one pharmaceutically-acceptable carrier to produce a pharmaceutical composition, prepared using techniques known in the art. In one aspect, a composition is prepared by admixing the compound with a pharmaceutically-acceptable carrier. Depending upon the components to be admixed, the components may or may not chemically or physically interact with one another.
  • Compounds provided herein may be formulated into pharmaceutical compositions that include at least one compound of the present invention, together with one or more pharmaceutically acceptable carriers, including excipients, such as diluents, binders and the like, and can further include additives, such as stabilizing agents, preservatives, solubilizing agents, and buffers, as desired.
  • Pharmaceutical compositions may include carriers, thickeners, diluents, buffers, preservatives, surface active agents and the like, in addition to the active ingredient. Pharmaceutical compositions may also include more than one active ingredient, such as a retinoid compound according to the invention, and one or more antimicrobial agents, keratolytic agents, benzoyl peroxide, antiinflammatory agents, corticosteroids, or the like. Pharmaceutically-acceptable carriers are known to those skilled in the art. These most typically would be standard carriers for administration to humans, including solutions such as sterile water, saline, and buffered solutions at physiological pH.
  • The pharmaceutical composition may be formulated for administration in a number of ways depending on whether local or systemic treatment is desired, or depending on the area to be treated. Administration can be topical (applied to the surface of the skin), or as an injectable solution or suspension, preferably for subcutaneous injection. Alternatively, administration can be oral, employing a dosage form such as a tablet or capsule, as would be readily understood by persons of ordinary skill in the art.
  • In practical use, a provided compound of the present invention can be combined as the active ingredient in an admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier used can have different forms depending on the dosage form desired for administration, for example, oral, parenteral, dermal or topical, transdermal, buccal, or the like.
  • If in an aqueous solution, a provided compound may be appropriately buffered by means of saline, acetate, phosphate, citrate, acetate or other buffering agents, which may be at any physiologically acceptable pH, generally from about pH 4 to about pH 7. A combination of buffering agents may also be employed, such as phosphate buffered saline, a saline and acetate buffer, and the like. In the case of saline, a 0.9% saline solution may be employed. In the case of acetate, phosphate, citrate and the like, a 50 mM solution may be employed. In addition to buffering agents, a suitable preservative may be employed, to prevent or limit bacteria and other microbial growth. One such preservative that may be employed is 0.05% benzalkonium chloride.
  • Preparations for administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of aqueous or non-aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Formulations for topical administration may include ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. It will be expected that the actual preferred amounts of active compound in a specified case will vary according to the specific compound being utilized, the particular compositions formulated, the mode of application, and the particular situs and mammal being treated.
  • In preparing the compositions for oral dosage form, typical pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, hard and soft capsules and tablets.
  • For solid administration formulations, any of a variety of thickening, filler, bulking and carrier additives may be employed, such as starches, sugars, fatty acids and the like. Formulation excipients may include polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol, mannitol, sodium chloride and sodium citrate.
  • For pharmaceutical formulations, it is also contemplated that any of a variety of measured-release, slow-release or time-release formulations and additives may be employed, so that the dosage may be formulated so as to effect delivery of a provided compound over a period of time, commonly referred to as controlled, delayed, extended, slow, or sustained, release formulations. For example, gelatin, sodium carboxymethylcellulose and/or other cellulosic excipients may be included to provide time-release or slower-release formulations, especially for administration by subcutaneous and intramuscular injection.
  • The formulation may be such that an application, administration, or injection is required on a daily, weekly, monthly or other periodic basis, depending on the concentration and amount of a provided compound, the biodegradation rate of a polymer used in the formulation, and other factors known to those of skill in the art.
  • Dosages for a given host can be determined using conventional considerations, e.g. by customary comparison of the differential activities of the subject compounds and of a known agent, e.g., by means of an appropriate conventional pharmacological protocol. Physicians and formulators, skilled in the art of determining doses of pharmaceutical compounds, will have no problems determining dose according to standard recommendations (Physician's Desk Reference, Barnhart Publishing (1999).
  • The following examples are given for the purpose of illustrating various embodiments of the invention and are not meant to limit the present invention in any fashion.
  • EXAMPLE 1 Chemistry of Retinoic Acid Analogues
  • 1H NMR spectra were obtained at 400.1 MHz (Bruker DRX spectrometer) in CDCl3. NOE experiments were performed on degassed samples using 2D phase-sensitive NOESY experiments with different mixing times (between 250 and 2000 ms). Typically, 1 s mixing times were used with 16 pulses for phase cycling and 2 dummy scans. The data were processed with line broadening of 0.3 Hz in each dimension and zero-filled to yield 512×4096 2D contour plots. The integrated intensities of the negative cross-peaks were determined using standard Bruker NMR software features.
  • UV/vis spectra were recorded on an AVIV 14DS spectrophotometer in cyclohexane or methanol solutions (Fisher, Spectrograde). IR spectra were recorded using a Nicolet FT IR spectrometer on thin films. HPLC separations were performed on a Gilson HPLC gradient system using 25-ml pump heads and an ISCO V.sup.4 variable wavelength detector. The column employed was a Whatman Partisil 10 M20/50 (500×22-mm i.d.) with a flow rate of 5 ml/min and monitoring by UV/vis detection at 340 nm. TLC chromatography was performed on pre-coated 250 μm silica gel GF glass plates (Analtech, Inc.; 5×10 cm). Solvents and liquid starting materials were distilled prior to use. Reactions and purifications were conducted with deoxygenated solvents, under inert gas (N2) and subdued lighting. Synthesis methods for retinoid compounds of the subject invention are described in U.S. Pat. No. 6,172,112 and U.S. Pat. Application, Publication No. 2010/0204327, which are incorporated herein by reference in their entirety.
  • (2E,4E,6E,8E)- and (2Z,4E,6E,8E)-Ethyl 8-(3′,4′-Dihydro-1′(2′H)-naphthalen-1′-ylidene-3,7-dimethyl-2,4,6-octatrie noate ((all-E)- and (13Z)-5)
  • This preparation employed a suspension of NaH (0.050 g, 2.1 mmol) in dry THF (1 ml), a solution of triethyl phosphonoseneciaote (0.11 g, 0.40 mmol) in dry THF (1 ml), HMPA (0.048 g, 0.22 mmol), and a solution of (all-E)-4 (0.065 g, 0.31 mmol) in dry THF (1 ml) to give a 2:1 mixture of esters (all-E) and (13)-5 (0.12 g, 78% yield). This mixture was separated by HPLC on silica gel using 1% Et.sub.2 O, 0.5% THF in hexane.
  • Procedure for the Hydrolysis of Individual Isomers of Ester 5
  • To a solution of the ester 5 (1 equiv.) in methanol (final concentration 0.061 M) was added an aqueous solution of 2 M KOH (10 equiv.). This solution was heated at reflux, and the reaction progress was monitored by TLC. After 90 min the hot solution was poured into a beaker of ice (40 g) and acidified with 10% HCl until pH2. The mixture was then extracted with Et2O, which was washed with brine, dried (Na2SO4), and concentrated under reduced pressure to give the product. NMR revealed that the hydrolysis occurred without isomerization. The following acids were synthesized by this method.
  • (2E,4E,6E,8E)-8-(3′,4′-Dihydro-1′(2′H)-naphthalen-1′-ylidene)3,7-dimethyl-2,4,6-octatrienoic acid ((all-E)-UAB30)
  • This preparation utilized a solution of KOH (0.258 g, 4.61 mmol) in water (2 ml) and a warm solution of ester (all-E)-5 (0.122 g, 0.378 mmol) in methanol (10 ml) to provide acid (all-E)-UAB30 (0.104 g, 93% yield) as a yellow solid: mp 192-197° C. (cyclohexane).
  • (2E,4E,6E,8E)-8-(3′,4′-Dihydro-1′(2′H)-naphthalen-1′-ylidene)3,7-dimethyl-2,4,6-octatrienoic acid ((9Z)-UAB30)
  • This preparation utilized a solution of KOH (0.15 g, 2.8 mmol) in water (1 ml) and a warm solution of ester (9Z)-5 (0.073 g, 0.23 mmol) in methanol (3 ml) to give acid (9Z)-UAB30 (0.065 g, 98% yield) as a yellow solid: mp 182-185° C. (cyclohexane).
  • (2E,4E,6E,8E)-8-(3′,4′-Dihydro-1′(2′H)-naphthalen-1′-ylidene)3,7-dimethyl-2,4,6-octatrienoic acid ((13Z)-UAB30)
  • This preparation employed a solution of KOH (0.085 g, 1.50 mmol) in water (1 ml) and a warm solution of ester (13Z)-5 (0.040 g, 0.12 mmol) in methanol (4 ml) to give acid (13Z)-UAB30 (0.034 g, 93% yield) as a yellow solid: mp 180-185° C. (cyclohexane).
  • EXAMPLE 2 Biology of Retinoic Acid Analogues (Skin Testing)
  • a. Summary
  • Compounds UAB30, 4Me-UAB30, UAB111 and Tazarotene were evaluated for possible skin effects (epidermal and dermal thickening as well as skin irritation) following a 14-day topical application in hairless SKH1-E mice. Known RAR-selective retinoids, such as tazarotene, stimulate proliferation of epidermal keratinocytes in human and rodent skin when applied topically. The stimulation the keratinocyte proliferation (reported histologically as epidermal hyperplasia), is believed to be due to increase in the epidermal growth factor receptor activation and is known to be reflective of retinoid effect (Fisher G., Vorhees J., Molecular mechanism of retinoid actions in skin. FASEB J. 1996; 10: 1002-1013). Along with the epidermal hyperplasia, RAR-selective retinoids are associated with significant skin irritation. Previously, RXR-selective retinoids have been reported not to efficiently induce epidermal hyperplasia. Surprisingly, the RXR-selective compound of the subject invention were identified here as exhibiting keratinocyte proliferation effects comparable to a potent RAR-selective retinoid (tazarotene). An additional advantage exhibited by the RXR-selective compounds of the subject invention, compared to tazarotene, was the observation of minimal to no irritation when applied topically.
  • UAB30, 4Me-UAB30 and UAB111 at 0.1% and 0.3%, tazarotene at 0.1% and vehicle were applied topically on approximately 8 cm2 (2×4 cm) of the dorsal skin (100 μL) and on the right ear (20 μL).
  • UAB30 topical application resulted in epidermal hyperplasia grading an average of 1.8 and 2 (in a scale of 0=none to 5=marked) for the 0.1% and 0.3% concentrations, respectively. The degree of epidermal hyperplasia induced by UAB30 was comparable to the hyperplasia induced by the potent RAR retinoid tazarotene, 0.1%, that reported an average of 2. 4Me-UBA30 0.1% and 0.3% and UAB111 0.1% and 0.3% treatments caused comparable epidermal hyperplasia ranging from 2.2 to 2.4 in the 5 point scale. Utriculus formation and sebaceous gland hyperplasia, normally present in this mice strain, was also mildly reduced with all the active treatments.
  • Tazarotene at 0.1% caused severe irritation, measured as erythema, desquamation and skin abrasion in the dorsal skin of all treated mice (Mean skin irritation score=3; scale of 0-4). Vehicle did not cause skin irritation. UAB30 at 0.1% did not cause any irritation whereas the 0.3% concentration was associated with only mild erythema (Mean irritation score=1). 4Me-UAB30 at 0.1% caused mild erythema in 2/5 animals only, however the 0.3% concentration was associated with severe irritation in all mice (Mean irritation score=3.6). UAB111 at 0.1% and 0.3% caused severe irritation (Mean irritation score=3.8) Tazarotene at 0.1%, 4Me-UAB30 at 0.3%, and UAB111 at 0.1% and 0.3% were associated with marked increase in ear Myeloperoxidase (MPO), a marker of neutrophil infiltration, whereas UAB30 at 0.1% and 0.3% and 4Me-UAB30 at 0.1% had no effect in this variable.
  • It is concluded that UAB30 at 0.1% and 0.3% surprisingly caused epidermal hyperplasia comparable to a potent RAR retinoid but with significantly less irritation when applied topically for 14 days in this hairless mice model.
  • b. Materials
  • 1. Test Substances and Dosing Pattern
  • The test compounds were UAB30, 4Me-UAB30 and UAB111. Tazarotene (0.1% cream) was purchased by Ricerca Taiwan Ltd. The three compounds at 0.1% and 0.3%, Tazarotene (0.1% cream) and vehicle (70% ethanol/30% propylene glycol) were each administered topically on the back (100 μL) and on the right ear (20 μL) once daily for two weeks.
  • The formulations are summarized as follows:
  • Formulation
    Test Compound Vehicle Solubility(a) Color Light Protection(b) Temperature (mg/mL)
    UAB30 70% ethanol/ S Lt. yellow Y 4° C. 1 and 3
    30% propylene
    glycol
    4Me-UAB30 70% ethanol/ S Lt. yellow Y 4° C. 1 and 3
    30% propylene
    glycol
    UAB111 70% ethanol/ S Lt. yellow Y 4° C. 1 and 3
    30% propylene
    glycol
    (a)Based upon visual observation: S: soluble; SS: slight soluble I: insoluble (suspension or ppt.)
    (b)Y: kept in tube or vial with brown color, or covered with aluminum foil; N: no protection from light
    (c) RT: prepared fresh and stored between 20-25° C.; 4° C.: prepared fresh and stored in the refrigerator or kept on ice.
  • 2. Animals
  • Female SKH1-E mice weighing 22±2 g, were provided by Charles River, USA. The animals were housed in Individually Ventilated Cages Racks (IVC Racks, 36 Mini Isolator systems) under clean area throughout the experiment. Every 2 or 3 mice were kept in an animal cage (in cm, 26.7 length×20.7 width×14.0 height) and maintained under controlled temperature (20-24° C.) and humidity (50%-80%) with 12-hour light/dark cycles. The animals were given free access to sterilized standard lab chow [MF-18 (Oriental Yeast Co., Ltd. Japan)] and sterile tap water ad libitum. All aspects of this work, i.e., housing, experimentation and disposal of animals, were performed in general accordance with the Guide for the Care and Use of Laboratory Animals (National Academy Press, Washington, D.C., 2011).
  • 3. Chemicals
  • Absolute ethanol (Merck, Germany), Mouse MPO ELISA kit (HK210, Hycult® biotech, The Netherlands), Propylene glycol (Sigma, USA) and Zorac® (Tazarotene, 1% cream) (Allergan, USA).
  • 4. Equipment
  • Animal cage (Techniplast, Italy), Dyer model micrometer gauge (Peacock, Japan), Pipetman (Gilson, France) and Timer (Wisewind, Taiwan).
  • c. Methods
  • 1. Skin, Topical, Hyperplasia, Irritation Assay.
  • Female SKH1-E hairless mice (Crl:SKH1-Hrhr) weighing 22±2 g were used. The animals were divided into groups of 5 each. Once a day, 100 μL of vehicle, tazarotene, and test articles were applied on the back with a micropipette and smeared evenly with gloved finger on approximately 8 cm2 (2×4 cm) of the dorsal skin; 20 μL was applied to the right ear. The tested animals were treated for two consecutive weeks. Body weight was monitored twice a week.
  • 2. Retinoid Activity.
  • Epidermal hyperplasia on the dorsal skin was used as a marker of retinoid activity. For histological evaluation, mice were sacrificed on day 15. The dorsal skin was dissected and stretched to the natural size on the filter. Punch biopsies (ED 8 mm) were obtained, fixed in 10% buffered formalin, and embedded in paraffin. Five micro thick sections were cut, stained with H&E, and a representative section of each biopsy was selected for histological evaluation.
  • 3. Epidermal Measurement.
  • The thickness of the epidermis and dermis was measured using a microscope with a micrometer in the eyepiece. The details are described in the histopathology report (see d. Histopathology, below).
  • 4. Skin Irritation.
  • Erythema, desquamation (skin flaking), and skin lesions/abrasion on dorsal skin were assessed visually in individual animals at the end of a 2-week treatment and scored on a scale of 0 to 4+, with 4+ being maximal.
  • 0: Normal
  • 1+: Mild erythema
  • 2+: Moderate erythema
  • 3+: Severe erythema/Desquamation
  • 4+: Lesions/abrasion.
  • 5. Histology.
  • For histological evaluation, the dorsal skin was dissected and stretched to the natural size on the filter. Punch biopsies (ED 8 mm) were obtained, fixed in 10% buffered formalin, and embedded in paraffin. Five micro thick sections were cut, stained with H&E, and a representative section of each biopsy was selected for histological evaluation. Right ear was harvested on day 15 for measurements of myeloperoxidase activity by mouse MPO ELISA kit, a measure of neutrophil infiltrates.
  • d. Histopathology
  • The histopathology findings were as follows:
      • 1) Hyperplasia of the epidermis was noted in all treatment groups except for the vehicle-treated group. Compared to the commercial product 0.1% Tazarotene, UAB30, 4Me-UBA30 and UAB111 treatment resulted in comparable incidence of epidermal hyperplasia.
      • 2) Hyperkeratosis and/or serocellular crust was seen in most of mice treated (3-5 of 5) with 0.1% Tazarotene, UAB111 and 0.3% 4Me-UBA30 but this finding was not reported in the vehicle and 0.3% UAB30 treated mice and only reported in 1 animal of the 0.1% UAB30 group and in 2 mice of the 0.1% 4Me-UBA30 group.
        e. Conclusions:
  • In female SKH1-E hairless mice 14-Day topical application of 0.1% and 0.3% UAB30, 4Me-UBA30 and UAB111 resulted in epidermal hyperplasia comparable to 0.1% tazarotene treatment. UAB30 at 0.1 and 0.3% as well as 4Me-UBA30 at 0.1% caused noticeable less irritation and hyperkeratosis than tazarotene treatment.
  • Those skilled in the art will readily appreciate that the present invention, or obvious modification thereof, are well adapted to carry out the objects and obtain the advantages mentioned or inherent therein. The present examples along with the methods, procedures, treatments, and compounds or compositions are representative of preferred or exemplary embodiments, and are not intended as limitations to the scope or spirit of the invention.

Claims (28)

1. A method for treating, preventing or lessening the severity of a non-neoplastic skin condition or disorder in a patient in need thereof, comprising the step of administering to said patient a compound, or a composition comprising the compound, said compound having the chemical formula selected from Formula I and Formula II, below:
Figure US20140057982A1-20140227-C00007
wherein
R1 is H, CF3 or a C1-C5 or greater branched or straight chain alkyl group;
R2 is H, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group, or
R1 and R2 collectively form a substituted or unsubstituted fused aryl group; and
R3 is one or more groups comprising, independently, H, F, diflouro, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
R6 and R7 are, independently, H, CF3, a C1-C15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
RH is H, or a C1-C15 branched or straight chain alkyl group.
n is from 0 to 3;
wherein one or more carbons in the ring structures of Formula I or II can optionally be replaced with a heteroatom;
or a pharmaceutically acceptable salt or isomer thereof.
2. The method of claim 1 wherein the compound has the chemical formula of Formula III, below:
Figure US20140057982A1-20140227-C00008
wherein,
R3 is one or more groups comprising, independently, H, F, diflouro, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
R6 and R7 are, independently, H, CF3, a C1-C15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
R9 is one or more groups comprising, independently, H, F, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, or a substituted or unsubstituted cycloalkyl group;
R11 is H, or a C1-C15 branched or straight chain alkyl group. n is from 0 to 3;
wherein one or more carbons in the fused ring structure can optionally be replaced with a heteroatom;
or a pharmaceutically acceptable salt or isomer thereof.
3. The method of claim 2 wherein the compound has the chemical formula shown as Formula IV:
Figure US20140057982A1-20140227-C00009
4. The method of claim 1 wherein the skin condition or disorder is selected from dermatological afflictions associated with excessive skin sebum production, and/or cell differentiation and/or proliferation disorders, and/or keratinization disorders and/or skin pigmentation disorders, and/or skin inflammatory disorders.
5. The method of claim 1 wherein the skin condition or disorder is selected from the dermatological afflictions associated photo-induced skin damage or chronological skin aging.
6. The method of claim 4 wherein the skin condition or disorder is selected from the group consisting of acne vulgaris, psoriasis, actinic keratosis, rosacea, seborrheic dermatitis, actinic lentigines, eczema, warts, keratosis, xerosis, icthyosis, lichen, keratoderma, folliculitis, vitiligo and melasma.
7. The method of claim 4 wherein the skin condition is acne vulgaris.
8. The method of claim 4 wherein the skin condition is psoriasis.
9. The method of claim 4 wherein the skin condition is actinic keratosis.
10. A pharmaceutically acceptable dermal composition for treating, preventing or ameliorating a non-neoplastic dermal condition, said composition comprising a vehicle or carrier pharmaceutically acceptable for dermal application and a compound having the chemical formula selected from Formula I and Formula II:
Figure US20140057982A1-20140227-C00010
wherein
R1 is H, CF3 or a C1-C5 or greater branched or straight chain alkyl group;
R2 is H, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group, or
R1 and R2 collectively form a substituted or unsubstituted fused aryl group; and
R3 is one or more groups comprising, independently, H, F, difluoro, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
R6 and R7 are, independently, H, CF3, a C1-C15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
RH is H, or a C1-C15 branched or straight chain alkyl group.
n is from 0 to 3;
wherein one or more carbons in the ring structures of Formula I or II can optionally be replaced with a heteroatom;
or a pharmaceutically acceptable salt or isomer thereof.
11. The composition of claim 10 wherein the compound has the chemical formula shown as Formula III:
Figure US20140057982A1-20140227-C00011
wherein,
R3 is one or more groups comprising, independently, H, F, diflouro, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, a substituted or unsubstituted cycloalkyl group;
R6 and R7 are, independently, H, CF3, a C1-C15 branched or straight chain alkyl group, or a substituted or unsubstituted cycloalkyl group;
R9 is one or more groups comprising, independently, H, CF3, a C1-C15 branched or straight chain alkyl group, a substituted or unsubstituted aryl group, or a substituted or unsubstituted aralkyl group, or a substituted or unsubstituted cycloalkyl group;
RH is H, or a C1-C15 branched or straight chain alkyl group. n is from 0 to 3;
wherein one or more carbons in the fused ring structure can optionally be replaced with a heteroatom;
or a pharmaceutically acceptable salt or isomer thereof.
12. The composition of claim 11 wherein the compound has the chemical formula shown as Formula IV, below:
Figure US20140057982A1-20140227-C00012
13. The composition of claim 10, wherein the composition is formulated for topical administration.
14. The composition of claim 10 comprising at least two active ingredients.
15. The composition of claim 14, wherein at least one of the active ingredients is a sunscreen.
16. The composition of claim 14 comprising at least two retinoid compounds.
17. The composition of claim 14 comprising at least one non-retinoid compound.
18. The composition of claim 16 further comprising a non-retinoid compound.
19. The composition of claim 10 wherein said compound is provided at a concentration of about 0.01% to about 10% (w/w).
20. The composition of claim 19 wherein the concentration of the compound is about 0.1% to about 3.0% (w/w).
21. The composition of claim 10, wherein the composition is formulated for oral administration.
22. The composition of claim 21 wherein the composition is in the form of a tablet.
23. The composition of claim 21 wherein the composition is in the form of a capsule.
24. The composition of claim 21 comprising at least two active ingredients.
25. The composition of claim 24 comprising at least two retinoid compounds.
26. The composition of claim 24 comprising at least one non-retinoid compound.
27. The composition of claim 21 wherein the compound is administered at a dose of about 0.01 mg/Kg to about 300 mg/kg of body weight.
28. The composition of claim 27 wherein the compound is administered at a dose of about 0.1 mg/Kg to about 30 mg/kg of body weight.
US13/932,294 2012-08-24 2013-07-01 Retinoids and use thereof Abandoned US20140057982A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/932,294 US20140057982A1 (en) 2012-08-24 2013-07-01 Retinoids and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13/594,177 US8475775B1 (en) 2012-08-24 2012-08-24 Retinoids and use thereof
US13/932,294 US20140057982A1 (en) 2012-08-24 2013-07-01 Retinoids and use thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/594,177 Continuation US8475775B1 (en) 2012-08-24 2012-08-24 Retinoids and use thereof

Publications (1)

Publication Number Publication Date
US20140057982A1 true US20140057982A1 (en) 2014-02-27

Family

ID=48671166

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/594,177 Active US8475775B1 (en) 2012-08-24 2012-08-24 Retinoids and use thereof
US13/932,294 Abandoned US20140057982A1 (en) 2012-08-24 2013-07-01 Retinoids and use thereof
US14/422,645 Abandoned US20150202174A1 (en) 2012-08-24 2013-07-01 Retinoids and use thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/594,177 Active US8475775B1 (en) 2012-08-24 2012-08-24 Retinoids and use thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/422,645 Abandoned US20150202174A1 (en) 2012-08-24 2013-07-01 Retinoids and use thereof

Country Status (18)

Country Link
US (3) US8475775B1 (en)
EP (1) EP2887928B1 (en)
JP (1) JP6382817B2 (en)
KR (1) KR20150045450A (en)
CN (1) CN104540502A (en)
AU (1) AU2013306281B2 (en)
BR (1) BR112015003561A8 (en)
CA (1) CA2882330A1 (en)
CL (1) CL2015000434A1 (en)
CO (1) CO7350653A2 (en)
ES (1) ES2670499T3 (en)
HU (1) HUE037075T2 (en)
IL (1) IL236991B (en)
IN (1) IN2015DN01524A (en)
MX (1) MX366050B (en)
NZ (1) NZ704496A (en)
RU (1) RU2687285C2 (en)
WO (1) WO2014031242A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220102239A1 (en) * 2020-09-30 2022-03-31 Kioxia Corporation Semiconductor device and manufacturing method

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10328040B2 (en) 2014-01-17 2019-06-25 Arizona Board Of Regents On Behalf Of Arizona State University Therapeutic methods
US9908856B2 (en) 2014-02-26 2018-03-06 Arizona Board Of Regents On Behalf Of Arizona State University Therapeutic compounds
US10112917B2 (en) 2015-03-03 2018-10-30 Arizona Board Of Regents On Behalf Of Arizona State University Therapeutic compounds
US10238655B2 (en) 2017-01-23 2019-03-26 Arizona Board Of Regents On Behalf Of Arizona State University Dihydroindene and tetrahydronaphthalene compounds
US10238626B2 (en) 2017-01-23 2019-03-26 Arizona Board Of Regents On Behalf Of Arizona State University Therapeutic compounds
US10231947B2 (en) 2017-01-23 2019-03-19 Arizona Board Of Regents On Behalf Of Arizona State University Isochroman compounds and methods of use thereof
US11696897B2 (en) * 2019-10-02 2023-07-11 Georg-August-Universitat Gottingen Stiftung Offentlichen Rechts, Universitatsmedizin Method for the treatment of diseases associated with sulfatase deficiencies
CN113207799B (en) * 2021-03-19 2022-03-15 中山大学 Construction method of type II diabetes mouse rapid heart failure model

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6172112B1 (en) * 1998-04-06 2001-01-09 Uab Research Foundation Retinoids and use thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5094783A (en) * 1990-06-29 1992-03-10 Uab Research Foundation Retinoid compounds
JP4699347B2 (en) * 2003-01-17 2011-06-08 ウィスコンシン・アルムニ・リサーチ・ファウンデーション Modified retinoid compounds and uses thereof
JP2008510822A (en) 2004-08-24 2008-04-10 ザ ユーエービー リサーチ ファウンデーション Method for producing retinoid and use thereof
EP2389357A4 (en) * 2009-01-20 2013-01-23 Signum Biosciences Inc Anti-inflammatory complexes
WO2011141822A2 (en) * 2010-05-11 2011-11-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for induction, predictive diagnosis, and treatment of affective behaviors by modulation of ppar and rxr receptors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6172112B1 (en) * 1998-04-06 2001-01-09 Uab Research Foundation Retinoids and use thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Jonette E. Keri; titel: Rosacea; downloaded from https://www.merckmanuals.com/professional/dermatologic-disorders/acne-and-related-disorders/rosacea on 06/21/17. *
Shinjita Das; title:Vitiligo; downloaded from https://www.merckmanuals.com/professional/dermatologic-disorders/pigmentation-disorders/vitiligo#v965720 on 06/21/17. *
Tutrone et al.; title: Topical therapy for actinic keratoses, II: Diclofenac, colchicine, and retinoids; Cutis, vol. 71(5), Abstract, May 2003 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220102239A1 (en) * 2020-09-30 2022-03-31 Kioxia Corporation Semiconductor device and manufacturing method

Also Published As

Publication number Publication date
MX2015002340A (en) 2015-08-05
RU2687285C2 (en) 2019-05-13
BR112015003561A2 (en) 2017-07-04
EP2887928A1 (en) 2015-07-01
RU2015103469A (en) 2016-10-20
IL236991B (en) 2019-03-31
BR112015003561A8 (en) 2018-01-23
KR20150045450A (en) 2015-04-28
EP2887928B1 (en) 2018-02-21
CA2882330A1 (en) 2014-02-27
WO2014031242A1 (en) 2014-02-27
HUE037075T2 (en) 2018-08-28
JP6382817B2 (en) 2018-08-29
ES2670499T3 (en) 2018-05-30
AU2013306281B2 (en) 2018-08-02
CL2015000434A1 (en) 2015-07-17
EP2887928A4 (en) 2016-03-23
US20150202174A1 (en) 2015-07-23
MX366050B (en) 2019-06-26
JP2015526464A (en) 2015-09-10
NZ704496A (en) 2017-01-27
CO7350653A2 (en) 2015-08-10
CN104540502A (en) 2015-04-22
IN2015DN01524A (en) 2015-07-03
US8475775B1 (en) 2013-07-02
AU2013306281A1 (en) 2015-02-26

Similar Documents

Publication Publication Date Title
US8475775B1 (en) Retinoids and use thereof
CA2866067C (en) Uses of certain f-series prostaglandin analogs for treating diabetes and dyslipidemia
US9592217B2 (en) Method for treating skin inflammatory diseases
JP2013529668A (en) Metronidazole esters for treating rosacea
JP2647880B2 (en) Topical alternative for the treatment of acne and dermatosis
CA3060365A1 (en) Amide derivatives of polycaffeoylquinic acids, process for producing same and uses thereof
US10653659B2 (en) Compositions comprising a compound from the family of avermectins and an agonist compound for at least one of the retinoic acid receptors for treating acne
AU2004251697A1 (en) Methods of treating hyperproliferative cell disorders
JP2013529667A (en) Metronidazole esters for treating rosacea
CN106176707B (en) Application of terpenoid camphene composite ester
AU2012321108B2 (en) Treatment of seborrhoea

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE UAB RESEARCH FOUNDATION, ALABAMA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BROUILLETTE, WAYNE J.;MUCCIO, DONALD D.;REDDY, ATIGADDA VENKATRAM;AND OTHERS;SIGNING DATES FROM 20120814 TO 20130823;REEL/FRAME:031301/0749

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF ALABAMA AT BIRMINGHAM;REEL/FRAME:055637/0313

Effective date: 20200121

AS Assignment

Owner name: NATIONAL INSTITUTE OF HEALTH - DIRECTOR DEITR, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF ALABAMA AT BIRMINGHAM;REEL/FRAME:064955/0591

Effective date: 20230919