US20050152859A1 - Inhibitors of melanocyte tyrosinase as topical skin lighteners - Google Patents

Inhibitors of melanocyte tyrosinase as topical skin lighteners Download PDF

Info

Publication number
US20050152859A1
US20050152859A1 US11/075,922 US7592205A US2005152859A1 US 20050152859 A1 US20050152859 A1 US 20050152859A1 US 7592205 A US7592205 A US 7592205A US 2005152859 A1 US2005152859 A1 US 2005152859A1
Authority
US
United States
Prior art keywords
alkyl
mammalian
compound
hydrogen
halogen
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
US11/075,922
Inventor
Thomas Dooley
Ernest Curto
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.)
MediQuest Therapeutics Inc
Original Assignee
MediQuest Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by MediQuest Therapeutics Inc filed Critical MediQuest Therapeutics Inc
Priority to US11/075,922 priority Critical patent/US20050152859A1/en
Publication of US20050152859A1 publication Critical patent/US20050152859A1/en
Priority to US12/206,334 priority patent/US7718185B2/en
Priority to US12/206,290 priority patent/US7858105B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/02Preparations for care of the skin for chemically bleaching or whitening the skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/095Sulfur, selenium, or tellurium compounds, e.g. thiols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/136Amines having aromatic rings, e.g. ketamine, nortriptyline having the amino group directly attached to the aromatic ring, e.g. benzeneamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/145Amines having sulfur, e.g. thiurams (>N—C(S)—S—C(S)—N< and >N—C(S)—S—S—C(S)—N<), Sulfinylamines (—N=SO), Sulfonylamines (—N=SO2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/17Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine
    • 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
    • A61K31/235Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids having an aromatic ring attached to a carboxyl group
    • A61K31/24Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids having an aromatic ring attached to a carboxyl group having an amino or nitro group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/357Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having two or more oxygen atoms in the same ring, e.g. crown ethers, guanadrel
    • A61K31/36Compounds containing methylenedioxyphenyl groups, e.g. sesamin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/428Thiazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/40Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing nitrogen
    • A61K8/41Amines
    • A61K8/411Aromatic amines, i.e. where the amino group is directly linked to the aromatic nucleus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/46Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing sulfur
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/49Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/49Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds
    • A61K8/4906Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with one nitrogen as the only hetero atom
    • A61K8/4913Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with one nitrogen as the only hetero atom having five membered rings, e.g. pyrrolidone carboxylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/49Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds
    • A61K8/494Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with more than one nitrogen as the only hetero atom
    • A61K8/4946Imidazoles or their condensed derivatives, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/49Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds
    • A61K8/4973Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with oxygen as the only hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/49Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds
    • A61K8/4986Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds with sulfur as the only hetero atom
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/74Biological properties of particular ingredients
    • A61K2800/78Enzyme modulators, e.g. Enzyme agonists
    • A61K2800/782Enzyme inhibitors; Enzyme antagonists

Definitions

  • the present invention relates to compounds and methods for inhibiting the activity of melanocyte tyrosinase in mammalian skin, in order to reduce the expression and production of skin pigmentation, and thereby lighten the color of mammalian skin.
  • Melanogenesis is the process of production and subsequent distribution of melanin by melanocytes within the skin and hair follicles [1, 2].
  • Melanocytes have specialized lysosome-like organelles, termed melanosomes, which contain several enzymes that mediate the production of melanin.
  • the copper-containing enzyme tyrosinase catalyzes the oxidation of the amino acid tyrosine into DOPA and subsequently DOPA-quinone.
  • At least two additional melanosomal enzymes are involved in the eumelanogenesis pathway that produces brown and black pigments, including TRP-1 (DHICA oxidase), and TRP-2 (DOPAchrome tautomerase).
  • TRP-1 DHICA oxidase
  • TRP-2 DOPAchrome tautomerase
  • the perceived color of skin and hair is determined by the ratio of eumelanins to pheomelanins, and in part on blood within the dermis.
  • the balance in skin hue is genetically regulated by many factors, including but not limited to: (a) the levels of expression of tyrosinase, TRP-2, and TRP-1; (b) thiol conjugation (e.g.
  • Vitiligo is the converse of hyperpigmentation, in which cutaneous melanocytes are either ablated or fail to produce sufficient pigment. [17, 18, 20] Although it would be desirable to restore lost pigmentation in vitiligo-affected skin with topical therapies, this has proven to be quite difficult to accomplish in a high proportion of subjects. As an alternative to PUVA therapy or cosmetic camouflage with dihydroxyacetone sunless-tanning lotions, [18] one might reduce the normal pigmentation of the unaffected skin to reduce contrast. Furthermore, a global market demand has developed for skin-lightening agents as “vanity” cosmeceutical products, because lighter skin color is preferred by some dark-skinned individuals in many countries and races, for psychological or sociological reasons. [4, 5].
  • MG Methyl gentisate
  • GA the methyl ester of gentisic acid
  • G 2,5-dihydroxybenzoic acid
  • GA is a moderately potent inhibitor of melanin accumulation in a murine melanocyte cell culture primary screen [6, 25].
  • GA is a natural product from the root of the genus Gentiana , named after Gentius , an Illyrian (Greco-Roman) king of the 2 nd century B.C., said to have first discovered the medicinal properties of the plant [7].
  • the sodium salt of GA is thought to be an analgesic and an anti-inflammatory agent.
  • GA is a ubiquitous metabolite, produced not only by plants, but also by Penicillium patulum and Polyporus tumulosus , and is excreted into the urine of mammals following ingestion of salicylates [8, 9].
  • MG and GA are simple phenolic compounds structurally similar to HQ, yet lacking the mutagenic activity of HQ [25]. MG has not been developed as a commercially available topical skin lightener product to date.
  • Another object is to provide methods and compositions for reducing pigmentation of skin for cosmetic, beauty-enhancing, or esthetic effects.
  • hyperpigmentation-related medical conditions such as melasma, age spots, freckles, ochronosis, postinflammatory hyperpigmentation, lentigo, and other pigmented skin blemishes.
  • Another object of the present invention is to provide methods and compositions for inhibiting mammalian melanocyte tyrosinase, the rate-limiting enzyme in the production of melanin from tyrosine and DOPA.
  • Still another object of the invention is to provide methods and compositions to absorb ultraviolet radiation (UVR), and thus to protect skin from UVR and photoaging.
  • UVR ultraviolet radiation
  • An additional object of the invention is to provide antioxidant compositions that protect skin from oxidative damage, and/or to prevent oxidative decomposition of product formulations.
  • Another object is to facilitate discovery of compounds that inhibit mammalian tyrosinase in cell-free extracts from mammalian melanocyte or melanoma cells, using either a colorometric DOPA oxidation or a radiolabeled tyrosine or DOPA substrate assay (IC 50 ⁇ 300 EM).
  • Another object is to facilitate discovery of compounds that inhibit de novo pigment production (synthesis and/or accumulation) in cultured mammalian melanocyte or melanoma cells (IC 50 ⁇ 300 ⁇ M).
  • Another object is to facilitate evaluation of compounds for toxicity in mammalian melanocyte, melanoma, or other cell cultures (IC 50 ⁇ 300 ⁇ M).
  • Another object is to provide composition of matter and/or identity of compounds that are efficacious and/or exhibit reduced toxicity using one or more of the bioassays described in other objects, with biochemical characteristics equivalent to or superior to hydroquinone or methyl gentisate.
  • Another object is to provide active and/or functional compounds from diverse structural classes, including but not limited to the following examples: benzoimidazoles, phenylamines, phenylthioureas, phenols, and phenylthiols.
  • Still another object is to provide synthesis of derivatives of active and/or functional compounds of the invention, including by organic synthesis, combinatorial chemistry, medicinal chemistry, X-ray crystallography, rational drug design, and other methods.
  • Another object is to provide the use of formulations of the present invention for cosmetic, cosmeceutical, over-the-counter drug, and prescription drug products.
  • Another object is to provide formulations of the present invention for the purpose of reducing or preventing pigmentation in hair, albeit during the biosynthesis of hair, as a result of blocking pigment production within the melanocytes of hair follicles.
  • Another object is to provide the active and/or functional compounds of the present invention for use in inhibiting tyrosinase or tyrosinase-like enzymes from non-mammalian species, for instance for use in the food science industry for the inhibition of enzymatic browning.
  • Still another object is to provide the active and/or functional compounds of the present invention for use in inhibiting tyrosine hydroxylase enzymes, in order to reduce the biosynthesis of DOPA and/or catecholamines.
  • the compounds preferably inhibit the enzymatic activity of melanocyte tyrosinase, though some compounds control pigment production in melanocyte cells without being potent inhibitors of the enzyme. Therefore, the compounds can be used in applications wherein controlling or preventing the production of pigments in mammalian skin is desired.
  • a few examples of such applications include:
  • active skin lightening compounds exhibit activity in the mammalian tyrosinase and/or melanocyte cell culture pigmentation assays, yet with minimal or no cytotoxicity. These compounds exhibit characteristics that are equivalent to or superior to the known standard skin-bleaching agent, hydroquinone, or the known standard tyrosinase inhibitor, methyl gentisate.
  • the compounds are typically applied topically to the skin wherein tyrosinase activity is sought to be reduced through a lotion or occlusive patch.
  • the compounds can be spread over a larger area to produce an even skin tone fade, or they can be applied locally to skin blemishes and other localized conditions to minimize skin irregularities.
  • the compounds can also be administered systemically by methods including oral, intradermal, transdermal, intraveneous, and parenteral administrations. The product works by inhibiting the production of melanin in cells beneath the skin surface. Because the skin naturally renews itself every ca.
  • the compounds of the present invention are benzimidazole and phenylthiourea related compounds represented by the following formula (I): wherein:
  • the compounds of the present invention are benzimidazole and phenylthiourea related compounds represented by the following formula (II): wherein:
  • the compounds of the present invention are phenylthiol, phenylamine, and multicyclic-phenolic related compounds of the following structure (III): wherein:
  • the compounds of the present invention are benzothiamide and thiophene amine derivatives defined by structures (IV) or (V): wherein:
  • the compound can be used as a “vanity” product, to lighten the skin of an individual, especially of dark skinned individuals.
  • the compound can be used to reduce uneven pigmentation marks and surface color irregularities, or to diminish pigmented skin blemishes such as freckles and age spots and hyperpigmentation-related medical conditions such as melasma, ochronosis, and lentigo.
  • the compounds can also be used to lighten hair when applied to skin containing pigmented hair follicles, and to lessen postinflammatory hyperpigmentation resulting from trauma or invasive surgery from a face lift, laser treatment, or cosmetic surgery.
  • the active or functional compounds can also be used to reduce skin pigmentation in normal skin adjacent to areas affected by vitiligo, thereby diminishing the contrast in color between normal and vitiligo affected skin.
  • the invention thus provides a method for lightening mammalian skin that includes applying or otherwise administering an effective treatment amount of an active skin-lightening compound selected from a benzimidazole, a phenylthiourea, a phenylthiol, a bi- or multicyclic phenol, thiopheneamine, a benzothiamide, a phenylamine, or a pharmaceutically acceptable salt or ester thereof, optionally in a pharmaceutically acceptable carrier, to a mammalian subject in need thereof.
  • an active skin-lightening compound selected from a benzimidazole, a phenylthiourea, a phenylthiol, a bi- or multicyclic phenol, thiopheneamine, a benzothiamide, a phenylamine, or a pharmaceutically acceptable salt or ester thereof, optionally in a pharmaceutically acceptable carrier, to a mammalian subject in need thereof.
  • the invention also includes a pharmaceutical composition for topical or general systemic administration, including oral, intradermal, transdermal, occlusive patch, intraveneous, and parenteral formulations, that includes an effective pigment inhibiting amount of the compound.
  • a pharmaceutical composition for topical or general systemic administration including oral, intradermal, transdermal, occlusive patch, intraveneous, and parenteral formulations, that includes an effective pigment inhibiting amount of the compound.
  • the present invention is principally concerned with compositions that inhibit mammalian tyrosinase activity, and which thus have medicinal and/or cosmetic value. However, the present invention can also extend to compounds that inhibit melanin formation within melanocytes through mechanisms other than tyrosinase activity.
  • the compounds also possess other activities that are beneficial when integrated into the compositions of the present invention. For example, many of the compounds also absorb UV light, and can thus be used to block the harmful effects of the sun's rays. Some of the compounds also possess antioxidant properties, and thus can inhibit oxidative damage to the skin, or contribute to the stability of the formulation.
  • TH tyrosine hydroxylase
  • the compounds of the present invention are benzimidazolethiol and phenylthiourea related compounds represented by the following formula (I):
  • a first series of subembodiments of the first principal embodiment is defined when R 1 , R 2 , and R′ are as defined above, R 4 , R 5 , R 6 , and R 7 are independently CR 8 , R′′ is CH, and:
  • R 1 , R 2 , and R′ are as defined above, R′′ is CH, and:
  • R 1 and R 2 are as defined above, R′′ is CH, and:
  • a fourth series of subembodiments of the first principal embodiment is defined when R 1 and R 2 are as defined above, R′′ is C, R′ is N or NH, and:
  • R 1 and R 2 are as defined above, R′′ is C, R′ is CH or CH 2 , and:
  • a first series of preferred species of the first principal embodiment are defined when R 1 and R 2 are as defined above, R′′ is C, R′ is NH or N, and:
  • R 1 and R 2 are as defined above, R′′ is CH, R′ is NH 2 , and:
  • a third series of preferred species of the first principal embodiment are defined when R′′ is CH, R′ is CH 3 , and R 4 , R 5 , R 6 , and R 7 are as defined in any one of the second series of preferred species.
  • the compounds of the present invention are benzimidazoles and phenylthiourea related compounds represented by the following formula (II): wherein:
  • a first series of subembodiments of the second principal embodiment are defined when R 1 , R 2 , R′ and R′′ are as defined above, R 4 , R 5 , R 6 , and R 7 are CR 8 , and:
  • R 1 , R 2 , R′ and R′′ are as defined above, and:
  • R 1 and R 2 are as defined above, R′′ is C, and:
  • R 1 and R 2 are as defined above, R′′ is CH, and:
  • a first series of preferred species of the second principal embodiment are defined when R 1 and R 2 are as defined above, R′′ is C, R′ is NH, and:
  • a second series of preferred species of the second principal embodiment are defined when R′′ is CH, R′ is NH 2 , and R 4 , R 5 , R 6 , and R 7 are as defined in any one of the first series of preferred species.
  • a third series of preferred species of the present invention are defined when R′′ is CH, R′ is CH 3 , and R 4 , R 5 , R 6 , and R 7 are as defined in any one of the first series of preferred species.
  • the compounds of the present invention are phenylthiol, phenylamine, and multicyclic-phenolic related compounds of the following structure (III): wherein:
  • R 1 is (CH 2 ) n SR 7 , n is 0, 1, 2, or 3 but preferably 0, and:
  • a second series of subembodiments is defined when R 1 is (CH 2 ) n SR 7 , n is 0, 1, 2, or 3, R 7 is C 1-5 alkyl optionally substituted with —OH, or C(O)C 1-3 alkyl; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • R 1 is SR 7
  • R 7 is C 1-5 alkyl optionally substituted with —OH, or C(O)C 1-3 alkyl
  • R 4 is —NHCO—C 1-3 alkyl
  • R 2 , R 3 , R 5 and R 6 are CH.
  • R 1 is (CH 2 ) n SR 7 , n is 0, 1, 2, or 3, R 7 is C 1-3 alkyl, or C(O)C 1-3 alkyl; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • R 4 is —NHCO—C 1-3 alkyl; and R 2 , R 3 , R 5 and R 6 are CH.
  • R 1 is (CH 2 ) n SR 7 , n is 0, 1, 2, or 3, R 7 is hydrogen; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • R 4 is —NHCO—C 1-3 alkyl; and R 2 , R 3 , R 5 and R 6 are CH.
  • R 1 is SR 7 , R 7 is C 1-5 alkyl optionally substituted with —OH; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • R 4 is —NHCO—C 1-3 alkyl; and R 2 , R 3 , R 5 and R 6 are CH.
  • R 1 is SR 7 , R 7 is C 1-3 alkyl; and R 2 , R 3 , R 4 , R 5 and R 4 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • R 4 is —NHCO—C 1-3 alkyl; and R 2 , R 3 , R 5 and R 6 are CH.
  • a seventh series of subembodiments is defined when R 1 is SR 7 , R 7 is hydrogen; and R 2 , R 3 , R 4 , R 5 and R 4 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • R 4 is —NHCO—C 1-3 alkyl; and R 2 , R 3 , R 5 and R 6 are CH.
  • R 1 is (CH 2 ) n NHR 7 , n is 0, 1, 2, or 3, R 7 is C 1-5 alkyl optionally substituted with —OH; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • a ninth series of subembodiments is defined when R 1 is (CH 2 ) n NHR 7 , n is 0, 1, 2, or 3, R 7 is C 1-3 alkyl; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • R 1 is (CH 2 ) n NHR 7 , n is 0, 1, 2, or 3, R 7 is hydrogen; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • R 1 is NHR 7
  • R 7 is C 1-5 alkyl optionally substituted with —OH
  • R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • a twelfth series of subembodiments is defined when R 1 is NHR 7 , R 7 is C 1-3 alkyl; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • R 1 is NHR 7 , R 7 is hydrogen; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 4 th through 23d subembodiments of the first series of subembodiments.
  • R 1 is OR 7
  • R 7 is C 1-5 alkyl optionally substituted with —OH
  • R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 8 th through 23d subembodiments of the first series of subembodiments.
  • R 1 is OR 7 , R 7 is C 1-3 alkyl; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 8 th through 23d subembodiments of the first series of subembodiments.
  • R 1 is OR 7 , R 7 is hydrogen; and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of the 8 th through 23d subembodiments of the first series of subembodiments.
  • a first series of species of the second principal embodiment are defined when R 1 is SH or SC(O)CH 3 , and:
  • a second series of preferred species are defined when R 1 is NH 2 , and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of species 1-17 of the first series of preferred embodiments.
  • a third series of preferred species are defined when R 1 is NHC(O)CH 3 , and R 2 , R 3 , R 4 , R 5 and R 6 are as defined in any one of species 1-17 of the first series of preferred embodiments.
  • a first series of subembodiments of the fourth principal embodiment are defined by structure (IV) when Y and Y′ are as described above, and:
  • a second series of subembodiments of the fourth principal embodiment are defined by structure (V) when:
  • Preferred species are defined for structure (IV) when Y is sulfur, Y′ is oxygen, R 1 and R 2 are hydrogen, and R 3 is methyl, and for structure (V) when X is sulfur, R 4 is ethylene and R 1 and R 2 are hydrogen.
  • the compounds of this invention can be optionally substituted with substituents that do not adversely affect the activity of the compound as a skin lightener.
  • substituents include, but are not limited to, alkyl (including lower alkyl), heteroalkyl, aryl, heterocyclic (including heteroaryl and heterocycloalkyl), halo, hydroxyl, carboxyl, acyl, acyloxy, amino, alkylamino, arylamino, alkoxy, aryloxy, alkylthio, alkylamido, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al., Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991. It will be understood that the present invention also covers “prodrugs” for such compositions, and pharmaceutically
  • one or all of three in vitro bioassays can be utilized to evaluate the efficacy and toxicity of candidate skin-lightening compounds.
  • the three bioassays characterize the compounds with regard to mammalian tyrosinase enzyme inhibition (cell free), pigmentation in melanocyte cultured cells, and cytotoxicity of mammalian cultured cells.
  • cell-based pigmentation and cell-free enzymatic assays have been developed [5, 6, 25] using the mammalian melanocyte cell line, Mel-Ab, a C57BL/6 mouse-derived cell line that produces high levels of melanin.
  • a distinct advantage of this approach is that humans share substantial sequence similarities in their genes (DNA) and proteins (such as tyrosinase) with mice, relative to non-mammalian species (e.g., mushrooms). So, mouse Mel-Ab melanocytes can serve as adequate surrogates for human melanocytes for many pharmacologic purposes.
  • Multi-well plate assays have been validated [5, 6, 25] for enzyme inhibition (e.g., DOPA oxidation by colorimetric measurement or radiolabeled substrate incorporation into melanin) and for pigmentation assays on cultured Mel-Ab cells. After 4 days of treatment of cultured cells, melanin content is determined using a spectrophotometer at 400+ nm. [6, 25]
  • This assay can detect an apparent loss in pigmentation resulting from either inhibition of de novo synthesis (e.g. via inhibition of tyrosinase, or the adenylate cyclase pathway, or another pathway) or a cytostatic/cytotoxic mechanism. It is therefore a broad primary screen. It is used in parallel with the tyrosinase enzymatic assay to determine whether an inhibitor of pigmentation at the cellular level is acting primarily at the enzyme level.
  • crystal violet or other staining methods may be used to quantify adherent cell numbers following a period of treatment by an agent.
  • HQ is typically used as a positive control in the assay, since it exhibits an IC 50 in the low micrograms per milliliter range on Mel-Ab culture using this assay, albeit owing to cytotoxicity and not inhibition of pigmentation per se.
  • inhibitors identified in cell-free enzymatic assays might have subsequent difficulties with toxicity or delivery in melanocyte cell-based assays. Therefore, all three in vitro assays in combination provide an excellent characterization of candidate skin lightening compounds.
  • a distinct advantage of the screening systems is the focus on mammalian tyrosinase, as opposed to non-mammalian enyzmes often used by other investigators, such as mushroom tyrosinase. Since the biochemical and pharmacologic characteristics of an enzyme or isozyme can vary dramatically between species of organisms (e.g., due to dissimilarities in primary, secondary, and tertiary structure), it is highly preferable that candidate topical skin lighteners intended for human use be discovered based on their biochemical action against a mammalian source of the enzyme.
  • Mushroom tyrosinase (and in some instances plant polyphenol oxidases) has been used in the vast majority of prior inhibitor studies. [28, 29] Yet fungal tyrosinase exhibits substantial dissimilarities from mammalian tyrosinase(s), and is viewed as a substantially inferior strategy for pharmacologic screening. Thus, the methods reported by the inventors of the present invention for screening against mammalian tyrosinase or within melanocytes is highly preferred over other possible screening strategies. [5, 6, 25].
  • a potentially effective candidate skin lightening agent is considered to be desirable, active, and/or functional if it renders 50% inhibition of mammalian tyrosinase enzyme activity, at concentrations below half the enzyme's “affinity” for tyrosine in cell-free enzyme extracts (IC 50 ⁇ 300 ⁇ M) and pigment production in melanocyte cell cultures (IC 50 ⁇ 300 ⁇ M).
  • the agent has an IC 50 against tyrosinase in cell-free enzyme extracts of less then 200, 100, 50, or 25 ⁇ M, and/or an IC 50 against pigment production in melanocyte cell cultures of less than 200, 100, 50, or 25 ⁇ M.
  • the agent exhibits toxicity at greater than 500, 750, or 1000 ⁇ M.
  • methyl gentisate is an “effective” candidate skin-lightening agent based on in vitro bioassays, because it has an IC 50 of 11.2 ⁇ 4 (ug/mL) against tyrosinase activity in cell-free assays, an IC 50 of 30.9 ⁇ 5 (ug/mL) in melanocyte cell cultures, and melanocyte cytotoxicity IC 50 of 118.7 ⁇ 12 (ug/mL).
  • Methyl gentisate thus poses a standard, against which the efficacy and cytotoxicity of other tyrosinase inhibiting compounds can be evaluated.
  • hydroquinone is an inferior standard, exhibiting potent cytotoxicity and minimal enzymatic inhibition. [5, 6, 25].
  • the invention provides methods for inhibiting pigment production that includes administering an effective treatment amount of a pigment inhibiting compound wherein (i) the compound inhibits tyrosinase activity equivalent to or greater than methyl gentisate in cell-free enzyme extracts from mammalian melanocyte or melanoma cells, when evaluated using either a colorometric DOPA oxidation or a radiolabeled tyrosine or DOPA substrate assay as described in Curto, E. V., et al.
  • the compound inhibits de novo pigment production (synthesis and/or accumulation) equivalent to or greater than methyl gentisate when evaluated in cultured mammalian melanocyte or melanoma cells.
  • Curto, E. V., et al. (1999) [25] the toxicity of the compound in mammalian melanocyte, melanoma, or other cell cultures is equivalent to or less than the toxicity of methyl gentisate. Curto, E. V., et al. (1999) [25].
  • computer-based molecular orbital predictions can aid in the understanding and predictability of structure-activity relationships, such that other effective compounds can be identified and evaluated. See Sakurada, J., et al., “Kinetic and molecular orbital studies on the rate of oxidation of monosubstituted phenols and anilines by horseradish peroxidase compound II.” Biochemistry 29: 4093-4098 (1990) [26].
  • Halo is fluoro, chloro, bromo, or iodo.
  • Alkyl, alkoxy, alkenyl, alkynyl, etc. denote both straight and branched groups; but reference to an individual radical such as “propyl” embraces only the straight chain radical, a branched chain isomer such as “isopropyl” being specifically referred to.
  • alkyl refers to a saturated straight, branched, or cyclic, primary, secondary, or tertiary hydrocarbon of C 1 to C 10 , and specifically includes methyl, ethyl, propyl, isopropyl, cyclopropyl, butyl, isobutyl, t-butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, cyclohexylmethyl, 3-methylpentyl, 2,2-dimethylbutyl, and 2,3-dimethylbutyl.
  • the moieties with which the alkyl group can be substituted are selected from the group consisting of hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, aryl, heterocycle, halo, carboxy, acyl, acyloxy, amido, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al., Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference.
  • lower alkyl refers to a C 1 to C 4 saturated straight, branched, or if appropriate, a cyclic (for example, cyclopropyl) alkyl group, including both substituted and unsubstituted forms. Unless otherwise specifically stated in this application, when alkyl is a suitable moiety, lower alkyl is preferred. Similarly, when alkyl or lower alkyl is a suitable moiety, unsubstituted alkyl or lower alkyl is preferred.
  • alkenyl and alkynyl refer to alkyl moieties, including both substituted and substituted forms, wherein at least one saturated C—C bond is replaced by a double or triple bond.
  • (C 2 -C 6 )alkenyl can be vinyl, allyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, or 5-hexenyl.
  • (C 2 -C 6 )alkynyl can be ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, or 5-hexynyl.
  • alkylene refers to a saturated, straight chain, divalent alkyl radical of the formula —(CH 2 ) n —, wherein n can be 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • aryl is intended to mean any stable monocyclic, bicyclic or tricyclic carbon ring of up to 8 members in each ring, wherein at least one ring is aromatic as defined by the Huckel 4n+2 rule.
  • aryl ring systems include phenyl, naphthyl, tetrahydronaphthyl, and biphenyl.
  • the aryl group can be substituted with one or more moieties selected from the group consisting of hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, alkyl, heterocycle, halo, carboxy, acyl, acyloxy, amido, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al., Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • heterocycle or heterocyclic as used herein except where noted represents a stable 5- to 7-membered monocyclic or stable 8- to 1-membered bicyclic heterocyclic ring which is either saturated or unsaturated, including heteroaryl, and which consists of carbon atoms and from one to three heteroatoms selected from the group consisting of N, O, S, and P; and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heteroaryl and heterocyclic groups include furyl, furanyl, pyridyl, pyrimidyl, thienyl, isothiazolyl, imidazolyl, tetrazolyl, pyrazinyl, benzofuranyl, benzothiophenyl, quinolyl, isoquinolyl, benzothienyl, isobenzofuryl, pyrazolyl, indolyl, isoindolyl, benzimidazolyl, purinyl, carbazolyl, oxazolyl, thiazolyl, isothiazolyl, 1,2,4-thiadiazolyl, isooxazolyl, pyrrolyl, quinazolinyl, cinnolinyl, phthalazinyl, xanthinyl, hypoxanthinyl, thiophene, furan, pyrrole, isopyrrole, pyrazole, imidazole
  • heteroaromatic and heterocyclic moieties can be optionally substituted as described above for aryl, including substituted with one or more substituents selected from hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, alkyl, heterocycle, halo, carboxy, acyl, acyloxy, amido, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al., Protective Groups in Organic Synthesis , John Wiley and Sons, Second Edition, 1991.
  • the heteroaromatic can be partially or totally hydrogenated as desired.
  • dihydropyridine can be used in place of pyridine.
  • Functional oxygen and nitrogen groups on the heteroaryl group can be protected as necessary or desired.
  • Suitable protecting groups are well known to those skilled in the art, and include trimethylsilyl, dimethylhexylsilyl, 1-butyldi-methylsilyl, and t-butyldiphenylsilyl, trityl or substituted trityl, alkyl groups, acyl groups such as acetyl and propionyl, methanesulfonyl, and p-toluenesulfonyl.
  • acyl refers to a carboxylic acid ester in which the non-carbonyl moiety of the ester group is selected from straight, branched, or cyclic alkyl or lower alkyl, alkoxyalkyl including methoxymethyl, aralkyl including benzyl, aryloxyalkyl such as phenoxymethyl, aryl including phenyl optionally substituted with halogen, C 1 to C 4 alkyl or C 1 to C 4 alkoxy, sulfonate esters such as alkyl or aralkyl sulphonyl including methanesulfonyl, the mono, di or triphosphate ester, trityl or monomethoxytrityl, substituted benzyl, trialkylsilyl (e.g.
  • esters dimethyl-t-butylsilyl or diphenylmethylsilyl.
  • Aryl groups in the esters optimally comprise a phenyl group.
  • lower acyl refers to an acyl group in which the non-carbonyl moiety is lower alkyl.
  • alkoxy refers to a moiety of the structure —O-alkyl, wherein alkyl is as defined above.
  • the 1,2-phenylenediamine derivatives (V) can be prepared by twice nitration/reduction reactions on substituted benzene (I), some substituents may need protection under above reaction conditions. Cyclization of (V) with CS 2 , or CH 3 NCS, or thiourea, or S, or (C 2 H 5 ) 2 NCS 2 Na can give the desired 2-mercaptobenzimidazole derivatives (VI). Reaction of (VI) with R 5 X (R 5 can be alkyl or acyl group; X is Cl, Br, I) can produce alkylated products (VIII). 2-Benzimidazoline-selenium derivatives (VI) and (DC) can be synthesized similarly by reacting selenourea with (V).
  • arylthiourea compounds (IV) can be prepared by reaction of corresponding aniline (III) with NH 4 SCN or KSCN in aqueous HCl solution. Alkylation of (IV) by R 6 X (R 6 can be alkyl or acyl group; X is Cl, Br, I) can yield monoalkylated product (VI). By replacing KSCN with KSeCN, the selenium analogous (V) can also be prepared.
  • Substituted arylsulfonyl chlorides (II) can be easily prepared from substituted aromatic compounds (I) by reaction with excess chlorosulfonic acid. Reduction of (II) with dichlorodimethylsilane/zinc will give desired phenylthiole derivatives (E).
  • the substituted phenylalkyl mercaptans (VI) can be prepared from the corresponding chloro compounds (V) which can be obtained from alkylation reaction of (1) (Friedel-Crafts reaction). Both thiole compounds (II) and (VI) can react with alkyl halide R 6 X to form the corresponding sulfides (IV) and (VII).
  • 2-thienyllithium salt (II) Protection may be necessary for some substituents.
  • Substituted 2-thiophenealkylamine (III) can be prepared by reaction of (E) with 1-chloro-2-dimethylaminoalkane.
  • the products (III), (IV) and (V) can be converted to each other by alkylation/dealkylation reactions using alkyl halide R 4 X and ClCO 2 Et, respectively.
  • Benzothioamide derivatives (II) can be prepared from substituted benzene (I) in one single step by reaction with KSCN in polyphosphoric acid or sulfuric acid.
  • the alkylated product (III can be obtained by using alkyl halide R 6 X (X is Cl, Br, I).
  • a compound of this invention is applied or administered to the skin during an appropriate period and using a sufficient number of dosages to achieve skin lightening.
  • concentration of active compound in the composition will depend on absorption, inactivation, and excretion rates of the compound as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.
  • the active ingredient may be administered as a single dose, or may be divided into a number of smaller doses to be administered at varying intervals of time.
  • Topical and other formulations of these active and/or functional compounds are of utility in lightening skin pigmentation in humans and other animals. These formulations may be useful for pure cosmetic purposes, simply to obtain a lighter skin color for perceived beautification.
  • the formulations also have medicinal value and can, for example, decrease hyperpigmentation of melasma, age spots, freckles, and other skin blemishes.
  • the compounds of this invention act primarily by inhibiting mammalian melanocyte tyrosinase, the rate-limiting enzyme in the production of melanin from tyrosine and DOPA. Some compounds also absorb ultraviolet radiation (UVR), and may thus protect skin from UVR and photoaging. In addition, some compounds may be antioxidants that protect skin from oxidative damage, and/or may prevent oxidative decomposition of product formulations.
  • UVR ultraviolet radiation
  • these formulations could also be used to reduce pigmentation in hair, albeit during the biosynthesis of hair, by blocking pigment production within the melanocytes of hair follicles.
  • the formulations would likely not affect the already emerged pigmented portions of hair, unlike a bleaching agent.
  • the formulations useful in the present invention contain biologically effective amounts of the functional and/or active compound(s).
  • a biologically effective amount of the active compound is understood by those skilled in the art to mean that a sufficient amount of the compound in the composition is provided such that upon administration to the human or animal by, for example, topical route, sufficient active agent is provided on each application to give the desired result.
  • the biologically effective amount of the active compound is at a level that it is not toxic to the human or animal during the term of treatment.
  • a suitable biologically compatible carrier when the compound is topically applied, it is understood that the carrier may contain any type of suitable excipient in the form of cosmetic compositions, pharmaceutical adjuvants, sunscreen lotions, creams, and the like.
  • the active compound is administered in a liposomal carrier.
  • the active compound is administered for a sufficient time period to alleviate the undesired symptoms and the clinical signs associated with the condition being treated, or to achieve the level of desired skin lightening.
  • the individual dosage, dosage schedule, and duration of treatment may be determined by clinical evaluations by those of skill in the art.
  • Solutions or suspensions for topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates; and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants such as ascorbic acid
  • Suitable vehicles, carriers, or formulations for topical application include lotions, suspensions, ointments, oil-in-water emulsions, water-in-oil emulsions, creams, gels, tinctures, sprays, powders, pastes, and slow-release transdermal or occlusive patches.
  • Thickening agents, emollients, and stabilizers can be used to prepare topical compositions. Examples of thickening agents include petrolatum, beeswax, xanthan gum, or polyethylene glycol, humectants such as sorbitol, emollients such as mineral oil, lanolin and its derivatives, or squalene.
  • thickening agents include petrolatum, beeswax, xanthan gum, or polyethylene glycol, humectants such as sorbitol, emollients such as mineral oil, lanolin and its derivatives, or squalene.
  • the compounds can be provided in the form of pharmaceutically-acceptable salts.
  • pharmaceutically-acceptable salts or complexes refers to salts or complexes that retain the desired biological activity of the parent compound and exhibit minimal, if any, undesired toxicological effects.
  • salts examples include (a) acid addition salts formed with inorganic acids (for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like), and salts formed with organic acids such as acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acids, naphthalenedisulfonic acids, and polygalacturonic acid; (b) base addition salts formed with polyvalent metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, and the like, or with an organic cation formed from N,N-dibenzylethylene-diamine or ethylenediamine; or (c) combinations of (a) and (b); e.g., a zinc t
  • the compounds can be modified in order to enhance their usefulness as pharmaceutical compositions.
  • various modifications of the active molecule such as alteration of charge, can affect water and lipid solubility and thus alter the potential for percutaneous absorption.
  • the vehicle, or carrier can also be modified to enhance cutaneous absorption, enhance the reservoir effect, and minimize potential irritancy or neuropharmacological effects of the composition. See, in general, Arndt, et al. [27].
  • the invention provides various formulations as topical skin lighteners containing the active and/or functional compounds described above.
  • the invention further provides formulations as topical anti-oxidants containing the active and/or functional compounds described above.
  • the invention provides formulations as topical sunscreens containing the active and/or functional compounds described above.
  • Such formulations can be made in combination with other active and/or functional ingredients used in skincare products (e.g. organic or inorganic sunscreen, antioxidant, anti-inflammatory, anti-erythema, antibiotic, antimicrobial, humectant, or other ingredients).
  • Other ingredients can be formulated with the compounds to augment their effect, including but not limited to Vitamin C, Vitamin E, magnesium ascorbyl phosphate, aloe vera extract, and retinoic acids.
  • alpha-hydroxy acids can be included to speed up the skin lightening process by exfoliating surface colored skin.
  • the compounds of the present invention can also be formulated for alternative routes of administration other than topical application, including but not limited to general systemic, oral, intradermal, transdermal, occlusive patches, intravenous, or parenteral administration, and pharmaceutical compositions known generally to those skilled in the art.
  • the compounds can also be formulated along with other active and/or functional ingredients used in skincare products, depending on the intended use of the formulation.
  • the compounds can be formulated with organic or inorganic sunscreens, an antioxidant, an anti-inflammatory, an anti-erythema, an antibiotic, an antimicrobial, a humectant, or other ingredients.
  • the active and/or functional compounds described above may also be of use in inhibiting tyrosinase-like enzymes from non-mammalian species, for instance for use in the food science industry for the inhibition of enzymatic browning.
  • Inhibition of plant polyphenol oxidases by agents described here may coincidentally have activity against these non-mammalian enzymes.
  • Suitable formulations for spraying or treatment of fruits are known generally to those skilled in the art. Treatment by these formulations containing the enzyme inhibitors of the present invention might improve shelf life of plant or fungal foods.
  • E is the concentration of compound that produces 50% pigment inhibition in the cell-free mammalian-enzyme assay system.
  • P is for the concentration of compound that produces 50% inhibition in the mammalian-melanocyte-culture pigment assay system.
  • T is the concentration of compound that kills 50% of cells in the # mammalian-melanocyte-culture toxicity assay system.
  • the compound extinction coefficient is ⁇ [OD/M ⁇ cm] at the wavelength of maximum absorbency ⁇ max [nm].
  • P is for the concentration of compound that produces 50% inhibition in the mammalian-melanocyte-culture pigment assay system.
  • T is the concentration of compound that kills 50% of cells # in the mammalian-melanocyte-culture toxicity assay system.
  • the compound extinction coefficient is ⁇ [OD/M ⁇ cm] at the wavelength of maximum absorbency ⁇ max [nm].
  • E is the concentration of compound that produces 50% pigment inhibition in the cell-free mammalian-enzyme assay system.
  • P is for the concentration of compound that produces 50% inhibition in the mammalian-melanocyte-culture pigment assay system.
  • T is the concentration of compound that kills 50% of cells in the # mammalian-melanocyte-culture toxicity assay system.
  • the compound extinction coefficient is ⁇ [OD/M ⁇ cm] at the wavelength of maximum absorbency ⁇ max [nm].
  • E is the concentration of compound that produces 50% pigment inhibition in the cell-free mammalian-enzyme assay system.
  • P is for the concentration of compound that produces 50% inhibition in the mammalian-melanocyte-culture pigment assay system.
  • T is the concentration of compound that kills 50% of cells in the # mammalian-melanocyte-culture toxicity assay system.
  • the compound extinction coefficient is ⁇ [OD/M ⁇ cm] at the wavelength of maximum absorbency ⁇ max [nm].

Abstract

Methods and formulations are provided to reduce pigmentation in skin, using an array of compounds selected from benzimidazoles, phenylthioureas, phenyltiols, phenylamines, bi- and multicyclic phenols, thiopheneamines, and benzothiamides. The compounds preferably inhibit pigment systhesis in melanocytes through the tyrosinase pathway. The methods can be used for lightening skin, and for treating uneven skin complexions which result from hyperpigmentation-related medical conditions such as melasma, age spots, freckles, ochronosis, and lentigo. The compounds can be used medically or cosmetically.

Description

    CROSS REFERENCE TO PRIOR APPLICATIONS
  • This application is a continuation of U.S. Ser. No. 09/795,683, filed Feb. 28, 2001, now abandoned and further claims priority to U.S. provisional patent application No. 60/185,610, filed Feb. 29, 2000. This application incorporates the entire disclosure of U.S. Ser. No. 09/795,683 and 60/185,610 as if fully set forth herein.
  • FIELD OF THE INVENTION
  • The present invention relates to compounds and methods for inhibiting the activity of melanocyte tyrosinase in mammalian skin, in order to reduce the expression and production of skin pigmentation, and thereby lighten the color of mammalian skin.
  • BACKGROUND OF THE INVENTION
  • Melanogenesis is the process of production and subsequent distribution of melanin by melanocytes within the skin and hair follicles [1, 2]. Melanocytes have specialized lysosome-like organelles, termed melanosomes, which contain several enzymes that mediate the production of melanin. The copper-containing enzyme tyrosinase catalyzes the oxidation of the amino acid tyrosine into DOPA and subsequently DOPA-quinone. At least two additional melanosomal enzymes are involved in the eumelanogenesis pathway that produces brown and black pigments, including TRP-1 (DHICA oxidase), and TRP-2 (DOPAchrome tautomerase). Depending on the incorporation of a sulfur-containing reactant (e.g. cysteine or glutathione) into the products, the melanogenesis pathway diverges to produce pheomelanins (amber and red pigments).
  • The perceived color of skin and hair is determined by the ratio of eumelanins to pheomelanins, and in part on blood within the dermis. The balance in skin hue is genetically regulated by many factors, including but not limited to: (a) the levels of expression of tyrosinase, TRP-2, and TRP-1; (b) thiol conjugation (e.g. with glutathione or cysteine) leading to the formation of pheomelanins; (c) the α-melanocyte-stimulating hormone (α-MSH) and melanocortin receptor, which is coupled to the adenylate cyclase/protein kinase A pathway; [15] (d) the product of the agouti locus, agouti signal protein, which has been documented to down-regulate pigmentation of hair melanocytes in rodents; [16] and (e) yet unknown mechanisms that regulate the uptake and distribution of melanosomes in recipient epidermal and hair matrix keratinocytes. [2, 13, 14].
  • Abnormalities of human skin pigmentation occur as a result of both genetic and environmental factors. Exposure of skin (especially Caucasian) to ultraviolet radiation, particularly in the UVB (i.e. intermediate) wavelengths, upregulates synthesis of melanocyte tyrosinase resulting in increased melanogenesis and thus tanning. However, acute or persistent UVB exposure can result in the formation of hyperpigmented lesions or regions of skin, including malignant melanoma skin cancer. [17] Both actinic damage and constitutional abnormalities can produce affected regions such as melasma, age spots, liver spots, freckles and other lentigenes. [3, 18, 19].
  • Vitiligo is the converse of hyperpigmentation, in which cutaneous melanocytes are either ablated or fail to produce sufficient pigment. [17, 18, 20] Although it would be desirable to restore lost pigmentation in vitiligo-affected skin with topical therapies, this has proven to be quite difficult to accomplish in a high proportion of subjects. As an alternative to PUVA therapy or cosmetic camouflage with dihydroxyacetone sunless-tanning lotions, [18] one might reduce the normal pigmentation of the unaffected skin to reduce contrast. Furthermore, a global market demand has developed for skin-lightening agents as “vanity” cosmeceutical products, because lighter skin color is preferred by some dark-skinned individuals in many countries and races, for psychological or sociological reasons. [4, 5].
  • Some purportedly “active” or “functional” agents for lightening skin color (e.g. arbutin, kojic acid, niacinamide, licorice, magnesium ascorbyl phosphate, among others) have not been demonstrated yet to be clinically efficacious when critically analyzed in carefully controlled studies [5, 6, 25]. The U.S. FDA-approved pharmaceutical products containing 24% hydroquinone (“HQ”) are minimally to moderately efficacious. However, HQ has been demonstrated to be cytotoxic to cultured mammalian melanocytes, and mutagenic in Salmonella and mammalian Chinese hamster V79 cells [3-6, 10, 11, 25]. HQ appears to be an important intermediate in the bioactivation of the carcinogen benzene [12]. Although it has been repeatedly asserted in the dermatologic literature for many years, without substantiation, that HQ is an inhibitor of tyrosinase, this compound is not an effective inhibitor of the mammalian enzyme [5, 6, 25]. Hydroquinone's in vitro mechanism of action appears to be primarily a melanocytic cytotoxic effect. Its clinical mechanism of action on whole skin remains uncertain.
  • In view of these biochemical disadvantages of the standard skin bleaching agent, HQ, it is highly desirable to identify other compounds with improved efficacy and safety characteristics. Methyl gentisate (“MG”), the methyl ester of gentisic acid (GA; 2,5-dihydroxybenzoic acid), is a moderately potent inhibitor of melanin accumulation in a murine melanocyte cell culture primary screen [6, 25]. GA is a natural product from the root of the genus Gentiana, named after Gentius, an Illyrian (Greco-Roman) king of the 2nd century B.C., said to have first discovered the medicinal properties of the plant [7]. The sodium salt of GA is thought to be an analgesic and an anti-inflammatory agent. GA is a ubiquitous metabolite, produced not only by plants, but also by Penicillium patulum and Polyporus tumulosus, and is excreted into the urine of mammals following ingestion of salicylates [8, 9]. MG and GA are simple phenolic compounds structurally similar to HQ, yet lacking the mutagenic activity of HQ [25]. MG has not been developed as a commercially available topical skin lightener product to date.
  • Two patent publications of Sansei Seiyaku also disclose a number of compounds, which allegedly are active as either tyrosinase inhibitors or as skin lightening agents, JP 5-124925 and JP 5-124922. The compounds are various benzimidazolethiols, but have not been developed as commercially available topical skin lightener products to date. In addition, phenylthiourea (PTU) has been reported as an inhibitor of tyrosinase, but has not been developed as a commercially available topical skin lightener product to date [30-32].
  • It is an object of this invention to provide methods and compositions for reducing pigmentation in skin from mammals, including humans.
  • Another object is to provide methods and compositions for reducing pigmentation of skin for cosmetic, beauty-enhancing, or esthetic effects.
  • It is another object to provide methods and compositions for treating hyperpigmentation-related medical conditions such as melasma, age spots, freckles, ochronosis, postinflammatory hyperpigmentation, lentigo, and other pigmented skin blemishes.
  • Another object of the present invention is to provide methods and compositions for inhibiting mammalian melanocyte tyrosinase, the rate-limiting enzyme in the production of melanin from tyrosine and DOPA.
  • Still another object of the invention is to provide methods and compositions to absorb ultraviolet radiation (UVR), and thus to protect skin from UVR and photoaging.
  • An additional object of the invention is to provide antioxidant compositions that protect skin from oxidative damage, and/or to prevent oxidative decomposition of product formulations.
  • Another object is to facilitate discovery of compounds that inhibit mammalian tyrosinase in cell-free extracts from mammalian melanocyte or melanoma cells, using either a colorometric DOPA oxidation or a radiolabeled tyrosine or DOPA substrate assay (IC50≦300 EM).
  • Another object is to facilitate discovery of compounds that inhibit de novo pigment production (synthesis and/or accumulation) in cultured mammalian melanocyte or melanoma cells (IC50≧300 μM).
  • Another object is to facilitate evaluation of compounds for toxicity in mammalian melanocyte, melanoma, or other cell cultures (IC50≧300 μM).
  • Another object is to provide composition of matter and/or identity of compounds that are efficacious and/or exhibit reduced toxicity using one or more of the bioassays described in other objects, with biochemical characteristics equivalent to or superior to hydroquinone or methyl gentisate.
  • Another object is to provide active and/or functional compounds from diverse structural classes, including but not limited to the following examples: benzoimidazoles, phenylamines, phenylthioureas, phenols, and phenylthiols.
  • Still another object is to provide synthesis of derivatives of active and/or functional compounds of the invention, including by organic synthesis, combinatorial chemistry, medicinal chemistry, X-ray crystallography, rational drug design, and other methods.
  • Another object is to provide the use of formulations of the present invention for cosmetic, cosmeceutical, over-the-counter drug, and prescription drug products.
  • Another object is to provide formulations of the present invention for the purpose of reducing or preventing pigmentation in hair, albeit during the biosynthesis of hair, as a result of blocking pigment production within the melanocytes of hair follicles.
  • Another object is to provide the active and/or functional compounds of the present invention for use in inhibiting tyrosinase or tyrosinase-like enzymes from non-mammalian species, for instance for use in the food science industry for the inhibition of enzymatic browning.
  • Still another object is to provide the active and/or functional compounds of the present invention for use in inhibiting tyrosine hydroxylase enzymes, in order to reduce the biosynthesis of DOPA and/or catecholamines.
  • SUMMARY OF THE INVENTION
  • Several classes of compounds are provided that reduce or prevent the production of pigment by mammalian melanocytes. The compounds preferably inhibit the enzymatic activity of melanocyte tyrosinase, though some compounds control pigment production in melanocyte cells without being potent inhibitors of the enzyme. Therefore, the compounds can be used in applications wherein controlling or preventing the production of pigments in mammalian skin is desired. A few examples of such applications include:
      • 1. As a vanity product, to lighten the skin of an individual, especially of dark skinned individuals;
      • 2. To lessen the hue of pigmented skin blemishes such as freckles and age spots;
      • 3. To diminish uneven pigmentation marks and surface color irregularities;
      • 4. To treat hyperpigmentation-related medical conditions such as melasma, ochronosis, and lentigo;
      • 5. To lighten hair pigmentation when applied to skin containing pigmented hair follicles;
      • 6. To lessen postinflammatory hyperpigmentation resulting from trauma or invasive surgery from a face lift, laser treatment, or cosmetic surgery; and
      • 7. To reduce skin pigmentation in normal skin adjacent to areas affected by vitiligo, thereby diminishing the contrast in color between normal and vitiligo affected skin.
  • Several classes of active skin lightening compounds have been discovered with which the present invention can be practiced. These compounds exhibit activity in the mammalian tyrosinase and/or melanocyte cell culture pigmentation assays, yet with minimal or no cytotoxicity. These compounds exhibit characteristics that are equivalent to or superior to the known standard skin-bleaching agent, hydroquinone, or the known standard tyrosinase inhibitor, methyl gentisate.
  • The compounds are typically applied topically to the skin wherein tyrosinase activity is sought to be reduced through a lotion or occlusive patch. The compounds can be spread over a larger area to produce an even skin tone fade, or they can be applied locally to skin blemishes and other localized conditions to minimize skin irregularities. Moreover, because most of the compounds are selective against melanocyte tyrosinase, the compounds can also be administered systemically by methods including oral, intradermal, transdermal, intraveneous, and parenteral administrations. The product works by inhibiting the production of melanin in cells beneath the skin surface. Because the skin naturally renews itself every ca. 28 days, when the compounds of the present invention are administered old (differentiated) pigmented keratinocytes cells are gradually sloughed off and keratinocytes with less melanin are eventually brought to the surface giving the skin a lighter, more even toned complexion.
  • In a first principal embodiment the compounds of the present invention are benzimidazole and phenylthiourea related compounds represented by the following formula (I):
    Figure US20050152859A1-20050714-C00001

    wherein:
    • 1) R1 is H or a valence for bonding;
    • 2) R2 is S, or SH;
    • 3) one of the dotted lines (----) represents a bond;
    • 4) R4, R5, R6, and R7 are independently CR8, or N;
    • 5) R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NHSO2—C1-3alkyl, (vii) —NHCO—C1-5 alkyl, (viii) oxime, (ix) hydrazine, (x) —NR9R10, (xi) HSO2, (xii) HSO3, (xiii) thio-C1-5 alkyl, (xiv) C1-5 acyloxy, (xv) H2PO3, (xvi) thiol, (xvii) —COOR9, (xviii) C1-5 alkynyl, or (xix) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy;
    • 6) R9 is hydrogen or C1-3 alkyl;
    • 7) R10 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
    • 8) R″ is C or CH;
    • 9) when R″ is C:
      • a) R′ is CR8, C(R8)2, N, or NH, and forms a bond with R″;
      • b) 1 or 2 of R5 and R6 are NH or COR10 other than COH, the remainder of R4, R5, R6, and R7 being CH; and
    • 10) when R″ is CH, R′ is CH3 or NH2.
  • In a second principal embodiment the compounds of the present invention are benzimidazole and phenylthiourea related compounds represented by the following formula (II):
    Figure US20050152859A1-20050714-C00002

    wherein:
    • 1) R1 is H;
    • 2) R2 is selenium;
    • 3) R″ is C or CH;
    • 4) when R″ is C, R′ is C(R8)2 or NR3, and forms a bond with R″;
    • 5) when R″ is CH, R′ is CH3 or NH2;
    • 6) R4, R5, R6, and R7 are independently CR8, or N;
    • 7) R3 is (i) substituted or unsubstituted alkyl, alkenyl, aryl, or heterocycle, (ii) —C1-5 alkoxy, (iii) —OH, (iv) hydrogen, (v) C(O)—C1-3 alkyl, or (vi) —(CH2)1-5 (O)NR 9R10;
    • 8) R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NHSO2—C1-3alkyl, (vii) —NHCO—C1-5 alkyl, (viii) oxime, (ix) hydrazine, (x) —NR9R10, (xi) HSO2, (xii) HSO3, (xiii) thio-C1-5 alkyl, (xiv) C1-5 acyloxy, (xv) H2PO3, (xvi) thiol, (xvii) —COOR9, (xiii) C1-5 alkynyl, or (xix) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy;
    • 9) R9 is hydrogen or C1-3 alkyl; and
    • 10) R10 is hydrogen, or C1-5 alkyl optionally substituted with —OH.
  • In a third principal embodiment the compounds of the present invention are phenylthiol, phenylamine, and multicyclic-phenolic related compounds of the following structure (III):
    Figure US20050152859A1-20050714-C00003

    wherein:
    • 1) R1 is (CH2)nSR7, (CH2)nNHR7, or OR7;
    • 2) n is 0, 1, 2, or 3,
    • 3) R2, R3, R4, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NHSO2—C1-3alkyl, (vii) —NHCO—C1-5 alkyl, (viii) oxime, (ix) hydrazine, (x) —NR9R10, (xi) HSO2, (xii) HSO3, (xiii) thio-C1-5 alkyl, (xiv) C1-5 acyloxy, (xv) H2PO3, (xvi) thiol, (xvii) —COOR9, (xviii) C1-5 alkynyl, or (xix) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy;
    • 4) alternatively, R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, pyridine, —X—(CH2)n′—X—wherein n′ is 1 and X is nitrogen, sulfur, or oxygen, and —(CH)n″XH— wherein n″ is 2 and X is as defined above;
    • 5) R7 is (i) substituted or unsubstituted alkyl, alkenyl, aryl, or heterocycle, (ii) —C1-5 alkoxy, (iii) hydrogen, (iv) —NR9R10, (v) C(O)—C1-3 alkyl, or (vi) —(CH2)mC(O)NR9R10;
    • 6) R9 is hydrogen or C1-3 alkyl;
    • 7) R10 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
    • 8) m is 1, 2, 3, 4, or 5; and
    • 9) provided that when R1 is OR7, R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, pyridine, —X—(CH2)n′—X— wherein n′ is 1 and X is nitrogen, sulfur, or oxygen, and —(CH)n″—XH— wherein n″ is 2 and X is as defined above.
  • In a fourth principal embodiment the compounds of the present invention are benzothiamide and thiophene amine derivatives defined by structures (IV) or (V):
    Figure US20050152859A1-20050714-C00004

    wherein:
    • 1) R1, R2, and R3 are independently (i) substituted or unsubstituted alkyl, alkenyl, aryl, or heterocycle, (ii) hydrogen, (iii) C(O)—C1-3 alkyl, or (iv) —(CH2)1-5C(O)NR9R10;
    • 2) R9 is hydrogen or C1-3 alkyl;
    • 3) R10 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
    • 4) Y and Y′ are independently oxygen or sulfur,
    • 5) X is oxygen, sulfur, or nitrogen; and
    • 6) R4 is C1-5 alkyl, optionally substituted by —OH, or NR9R9.
    DETAILED DESCRIPTION OF THE INVENTION
  • Discussion
  • As noted above, compounds for inhibiting or preventing melanin formation in skin have been discovered for the treatment of various melanin-associated conditions. For example, the compound can be used as a “vanity” product, to lighten the skin of an individual, especially of dark skinned individuals. Alternatively, the compound can be used to reduce uneven pigmentation marks and surface color irregularities, or to diminish pigmented skin blemishes such as freckles and age spots and hyperpigmentation-related medical conditions such as melasma, ochronosis, and lentigo. The compounds can also be used to lighten hair when applied to skin containing pigmented hair follicles, and to lessen postinflammatory hyperpigmentation resulting from trauma or invasive surgery from a face lift, laser treatment, or cosmetic surgery. The active or functional compounds can also be used to reduce skin pigmentation in normal skin adjacent to areas affected by vitiligo, thereby diminishing the contrast in color between normal and vitiligo affected skin.
  • The invention thus provides a method for lightening mammalian skin that includes applying or otherwise administering an effective treatment amount of an active skin-lightening compound selected from a benzimidazole, a phenylthiourea, a phenylthiol, a bi- or multicyclic phenol, thiopheneamine, a benzothiamide, a phenylamine, or a pharmaceutically acceptable salt or ester thereof, optionally in a pharmaceutically acceptable carrier, to a mammalian subject in need thereof. The invention also includes a pharmaceutical composition for topical or general systemic administration, including oral, intradermal, transdermal, occlusive patch, intraveneous, and parenteral formulations, that includes an effective pigment inhibiting amount of the compound. The present invention is principally concerned with compositions that inhibit mammalian tyrosinase activity, and which thus have medicinal and/or cosmetic value. However, the present invention can also extend to compounds that inhibit melanin formation within melanocytes through mechanisms other than tyrosinase activity.
  • Many of the compounds also possess other activities that are beneficial when integrated into the compositions of the present invention. For example, many of the compounds also absorb UV light, and can thus be used to block the harmful effects of the sun's rays. Some of the compounds also possess antioxidant properties, and thus can inhibit oxidative damage to the skin, or contribute to the stability of the formulation.
  • Furthermore, although unrelated to skin pigmentation per se, some of the compounds of the present invention may also inhibit tyrosine hydroxylase (TH). This enzyme is structurally dissimilar from tryosinase, but also catalyzes the formation of DOPA from tysosine. TH is critical for the formation of catecolamines. Therefore, some of the compounds of the present invention which coincidentally inhibit TH activity may have utility in reducing catecholamine biosynethsis, for instance for use as inhibitor “probes” in laboratory experiments where reduction in catacholamine pools is desirable. [30-32].
  • Compounds of the Present Invention
  • In a first principal embodiment the compounds of the present invention are benzimidazolethiol and phenylthiourea related compounds represented by the following formula (I):
    Figure US20050152859A1-20050714-C00005
      • wherein:
      • a. R1 is H or a valence for bonding;
      • b. R2 is S, or SH;
      • c. one of the dotted lines (----) represents a bond;
      • d. R4, R5, R6, and R7 are independently CR8, or N;
      • e. R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NHSO2—C1-3alkyl, (vii) —NHCO—C1-5 alkyl, (viii) oxime, (ix) hydrazine, (x) —NR9R10, (xi) HSO2, (xii) HSO3, (xiii) thio-C1-5 alkyl, (xiv) C1-5 acyloxy, (xv) H2PO3, (xvi) thiol, (xvii) —COOR9, (xviii) C1-5 alkynyl, or (xix) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy;
      • f. R9 is hydrogen or C1-3 alkyl;
      • g. R10 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
      • h. R″ is C or CH;
      • i. when R″ is C:
        • i. R′ is CR8, C(R8)2, N or NH, and forms a bond with R″;
        • ii. 1 or 2 of R5 and R6 are N or COR10 other than COH, the remainder of R4, R5, R6, and R7 being CH; and
      • j. when R″ is CH, R′ is CH3 or NH2.
  • A first series of subembodiments of the first principal embodiment is defined when R1, R2, and R′ are as defined above, R4, R5, R6, and R7 are independently CR8, R″ is CH, and:
    • 1) R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (viii) —NR9R10, (xi) C1-5 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy;
    • 2) R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR9, (viii) —NR9R9, (xi) C1-3 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-3 alkyl, —C1-3 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R9, C1-3 thioether, or C1-3 alkoxy;
    • 3) R8 is (i) hydrogen, (ii) halogen, (v) —OR9, (viii) —NR9R9, (xii) thiol, or (xiv) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2;
    • 4) R8 is C1-3 alkyl;
    • 5) R8 is OR10 or OR9; or
    • 6) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, C(CF3), and C(CHCHBr).
  • A second series of subembodiments of the first principal embodiment is defined when R1, R2, and R′ are as defined above, R″ is CH, and:
    • 1) R4, R5, R6, and R7 are independently selected from CR8, 2 or 3 of R4, R5, R6, and R7 are CH, and R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NR9R10, (vii) C1-5 acyloxy, (viii) thiol, (ix) COOR9, or (x) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy;
    • 2) R4, R5, R5, and R7 are independently selected from CR8, 2 or 3 of R4, R5, R6, and R7 are CH, and R8 is (i) hydrogen, (ii) halogen, (iii) —OR9, (iv) —OH, (v) —NR9R9, (vi) thiol, or (vii) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2;
    • 3) R4, R5, R6, and R7 are independently selected from CR8, 2 or 3 of R4, R5, R6, and R7 are CH, and R8 is C1-3 alkyl;
    • 4) R4, R5, R6, and R7 are independently selected from CR9, R5 is OR9, and 2 or 3 of R4, R5, R6, and R7 are CH; or
    • 5) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, C(CF3), and C(CHCHBr), and 2 or 3 of R4, R5, R6, and R7 are CH.
  • A third series of subembodiments of the first principal embodiment is defined when R1 and R2 are as defined above, R″ is CH, and:
    • 1) R4, R5, R6, and R7 are independently selected from CR8, and R′ is NH2;
    • 2) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (viii) —NR9R10, (xi) C1-5 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is NH2;
    • 3) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (v) —OR9, (vii) —OH, (viii) —NR9R9, (xii) thiol, or (xiv) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is NH2;
    • 4) R4, R5, R6, and R7 are independently selected from CR8, R8 is C1-3 alkyl, OR10, or OR9, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is NH2;
    • 5) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, CCF3, and C(CHCHBr), 2 or 3 of R4, R5, R6, and R7 are CH; and R′ is NH2;
    • 6) R4, R5, R6, and R7 are independently selected from CR8, and R′ is CH3;
    • 7) R4, R5, R6, and R7 are independently selected from CR9, R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (viii) —NR9R10, (xi) C1-5 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is CH3;
    • 8) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (v) —OR9, (vii) —OH, (viii) —NR9R9, (xii) thiol, or (xiv) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is CH3;
    • 9) R4, R5, R6, and R7 are independently selected from CR9, R9 is R8 is C1-3 alkyl or OR9, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is CH3;
    • 10) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, CCF3, and C(CHCHBr), 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is CH3;
  • A fourth series of subembodiments of the first principal embodiment is defined when R1 and R2 are as defined above, R″ is C, R′ is N or NH, and:
    • 1) 1 or 2 of R5 and R6 are COR10 other than COH, the remainder of R4, R5, R6, and R7 being CH;
    • 2) 1 or 2 of R5 and R6 are COR9 other than COH, the remainder of R4, R5, R6, and R7 being CH;
    • 3) 1 or 2 of R5 and R6 are N, the remainder of R4, R5, R6, and R7 being CH;
    • 4) R5 is COR9 other than COH, and R4, R6, and R7 are CH;
    • 5) R6 is COR9 other than COH, and R4, R5, and R7 are CH; or
    • 6) R5 and R6 are COR9 other than COH, and R4 and R7 are CH.
  • A fifth series of subembodiments of the first principal embodiment are defined when R1 and R2 are as defined above, R″ is C, R′ is CH or CH2, and:
    • 1) 1 or 2 of R5 and R6 are COR10 other than COH, the remainder of R4, R5, R6, and R7 being CH;
    • 2) 1 or 2 of R5 and R6 are COR9 other than COH, the remainder of R4, R5, R6, and R7 being CH;
    • 3) 1 or 2 of R5 and R6 are N, the remainder of R4, R5, R6, and R7 being CH;
    • 4) R5 is COR9 other than COH, and R4, R6, and R7 are CH;
    • 5) R6 is COR9 other than COH, and R4, R5, and R7 are CH; or
    • 6) R5 and R6 are COR9 other than COH, and R4 and R7 are CH.
  • A first series of preferred species of the first principal embodiment are defined when R1 and R2 are as defined above, R″ is C, R′ is NH or N, and:
    • 1) R5 is COCH3, and R4, R6, and R7 are CH;
    • 2) R6 is COCH3, and R4, R5, and R7 are CH;
    • 3) R5 and R6 are COCH3, and R4 and R7 are CH;
  • A second series of preferred species of the first principal embodiment are defined when R1 and R2 are as defined above, R″ is CH, R′ is NH2, and:
    • 1) R4, R5, R6, and R7 are CH;
    • 2) R4 is CCH3, and R5, R6, and R7 are CH;
    • 3) R5 is CCH3, and R4, R6, and R7 are CH;
    • 4) R6 is CCH3, and R4, R5, and R7 are CH;
    • 5) R7 is CCH3, and R4, R5, and R6 are CH;
    • 6) R4 is COCH3, and R5, R6, and R7 are CH;
    • 7) R5 is COCH3, and R4, R6, and R7 are CH;
    • 8) R6 is COCH3, and R4, R5, and R7 are CH;
    • 9) R7 is COCH3, and R4, R5, and R6 are CH;
    • 10) R4 is CF, and R5, R6, and R7 are CH;
    • 11) R5 is CF, and R4, R6, and R7 are CH;
    • 12) R6 is CF, and R4, R5, and R7 are CH;
    • 13) R7 is CF, and R4, R5, and R6 are CH;
    • 14) R4 is COH, and R5, R6, and R7 are CH;
    • 15) R5 is COH, and R4, R6, and R7 are CH;
    • 16) R6 is COH, and R4, R5, and R7 are CH;
    • 17) R7 is COH, and R4, R5, and R6 are CH;
    • 18) 2 of R4, R5, R6 are R7 are CCH3, and 2 of R4, R5, R6, and R7 are CH;
    • 19) 2 of R4, R5, R6 are R7 are COCH3, and 2 of R4, R5, R6, and R7 are CH;
    • 20) 2 of R4, R5, R6 are R7 are CF, and 2 of R4, R5, R6, and R7 are CH; or
    • 21) 2 of R4, R5, R6 are R7 are COH, and 2 of R4, R5, R6, and R7 are CH;
  • A third series of preferred species of the first principal embodiment are defined when R″ is CH, R′ is CH3, and R4, R5, R6, and R7 are as defined in any one of the second series of preferred species.
  • In a second principal embodiment the compounds of the present invention are benzimidazoles and phenylthiourea related compounds represented by the following formula (II):
    Figure US20050152859A1-20050714-C00006

    wherein:
    • 1) R1 is H;
    • 2) R2 is selenium;
    • 3) R″ is C or CH;
    • 4) when R″ is C, R′ is C(R8)2 or NR3, and forms a bond with R″;
    • 5) when R″ is CH, R′ is CH3 or NH2;
    • 6) R4, R5, R6, and R7 are independently CR8, or N;
    • 7) R3 is (i) substituted or unsubstituted alkyl, alkenyl, aryl, or heterocycle, (ii) —C1-5 alkoxy, (iii) —OH, (iv) hydrogen, (v) C(O)—C1-3 alkyl, or (vi) —(CH2)1-5C(O)NR9R10;
    • 8) R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NHSO2—C1-3alkyl, (vii) —NHCO—C1-5 alkyl, (viii) oxime, (ix) hydrazine, (x) —NR9R10, (xi) HSO2, (xii) HSO3, (xiii) thio-C1-5 alkyl, (xiv) C1-5 acyloxy, (xv) H2PO3, (xvi) thiol, (xvii) —COOR9, (xviii) C1-5 alkynyl, or (xix) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy;
    • 9) R9 is hydrogen or C1-3 alkyl; and
    • 10) R10 is hydrogen, or C1-5 alkyl optionally substituted with —OH.
  • A first series of subembodiments of the second principal embodiment are defined when R1, R2, R′ and R″ are as defined above, R4, R5, R6, and R7 are CR8, and:
    • 1) R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (viii) —NR9R10, (xi) C1-5 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy;
    • 2) R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR9, (viii) —NR9R9, (xi) C1-3 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-3 alkyl, —C1-3 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R9, C1-3 thioether, or C1-3 alkoxy;
    • 3) R8 is (i) hydrogen, (ii) halogen, (v) —OR9, (viii) —NR9R9, (xii) thiol, or (xiv) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2;
    • 4) R8 is C1-3 alkyl;
    • 5) R9 is OR10 or OR9; or
    • 6) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, CCF3, and C(CHCHBr).
  • A second series of subembodiments of the second principal embodiment is defined when R1, R2, R′ and R″ are as defined above, and:
    • 1) R4, R5, R6, and R7 are independently selected from CR8, 2 or 3 of R4, R5, R6, and R7 are CH, and R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NR9R10, (vii) C1-5 acyloxy, (viii) thiol, (ix) COOR9, or (x) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy;
    • 2) R4, R5, R6, and R7 are independently selected from CR9, 2 or 3 of R4, R5, R6, and R7 are CH, and R8 is (i) hydrogen, (ii) halogen, (iii) —OR9, (iv) —OH, (v) —NR9R9, (vi) thiol, or (vii) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2;
    • 3) R4, R5, R6, and R7 are independently selected from CR8, 2 or 3 of R4, R5, R6, and R7 are CH, and R8 is C1-3 alkyl;
    • 4) R4, R5, R6, and R7 are independently selected from CR8, R8 is OR9, and 2 or 3 of R4, R5, R6, and R7 are CH; and
    • 5) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, CCF3, and C(CHCHBr), and 2 or 3 of R4, R5, R6, and R7 are CH.
  • A third series of subembodiments of the second principal embodiment are defined when R1 and R2 are as defined above, R″ is C, and:
    • 1) R4, R5, R6, and R7 are independently selected from CR8, and R′ is NR3;
    • 2) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (viii) —NR9R10, (xi) C1-5 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is NR3;
    • 3) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (v) —OR9, (vii) —OH, (viii) —NR9R9, (xii) thiol, or (xiv) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is NR3;
    • 4) R4, R5, R6, and R7 are independently selected from CR8, R8 is R8 is C1-3 alkyl or OR9, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is NR3;
    • 5) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, CCF3, and C(CHCHBr), 2 or 3 of R4, R5, R6, and R7 are CH; and R′ is NR3;
    • 6) R4, R5, R6, and R7 are independently selected from CR8, R′ is NR3, and R3 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
    • 7) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (viii) —NR9R10, (xi) C1-5 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy, 2 or 3 of R4, R5, R6, and R7 are CH, R′ is NR3, and R3 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
    • 8) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (v) —OR9, (vii) —OH, (viii) —NR9R9, (xii) thiol, or (xiv) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2, 2 or 3 of R4, R5, R6, and R7 are CH, R′ is NR3, and R3 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
    • 9) R4, R5, R6, and R7 are independently selected from CR8, R8 is R9 is C1-3 alkyl or OR9, 2 or 3 of R4, R5, R6, and R7 are CH, R′ is NR3, and R3 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
    • 10) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, CCF3, and C(CHCHBr), 2 or 3 of R4, R5, R6, and R7 are CH; R′ is NR3; and R3 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
    • 11) R4, R5, R6, and R7 are independently selected from CR8, R′ is NR3, and R3 is hydrogen or C1-3 alkyl;
    • 12) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (viii) —NR9R10, (xi) C1-5 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy, 2 or 3 of R4, R5, R6, and R7 are CH, R′ is NR3, and R3 is hydrogen or C1-3 alkyl;
    • 13) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (v) —OR9, (vii) —OH, (viii) —NR9R9, (xii) thiol, or (xiv) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2, 2 or 3 of R4, R5, R6, and R7 are CH, R′ is NR3, and R3 is hydrogen or C1-3 alkyl;
    • 14) R4, R5, R6, and R7 are independently selected from CR8, R8 is R8 is C1-3 alkyl or OR9, 2 or 3 of R4, R5, R6, and R7 are CH, R′ is NR3, and R3 is hydrogen or C1-3 alkyl; or
    • 15) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, CCF3, and C(CHCHBr), 2 or 3 of R4, R5, R6, and R7 are CH; R′ is NR3, and R3 is hydrogen or C1-3 alkyl.
  • A fourth series of subembodiments of the second principal embodiment is defined when R1 and R2 are as defined above, R″ is CH, and:
    • 1) R4, R5, R6, and R7 are independently selected from CR8, and R′ is NH2;
    • 2) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (viii) —NR9R10, (xi) C1-5 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is NH2;
    • 3) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (v) —OR9, (vii) —OH, (viii) —NR9R9, (xii) thiol, or (xiv) —C1-3 alkyl or alkenyl optionally substituted with one or more of-OH, —SH, halogen, or NH2, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is NH2;
    • 4) R4, R5, R6, and R7 are independently selected from CR8, R8 is R8 is C1-3 alkyl or OR9, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is NH2;
    • 5) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, CCF3, and C(CHCHBr), 2 or 3 of R4, R5, R6, and R7 are CH; and R′ is NH2;
    • 6) R4, R5, R6, and R7 are independently selected from CR8, and R′ is CH3;
    • 7) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (viii) —NR9R10, (xi) C1-5 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy, 2 or 3 of R4, R5, R6, and R7 are CH, R′ is NR3, and R′ is CH3;
    • 8) R4, R5, R6, and R7 are independently selected from CR8, R8 is (i) hydrogen, (ii) halogen, (v) —OR9, (vii) —OH, (viii) —NR9R9, (xii) thiol, or (xiv) —C1-3 alkyl or alkenyl optionally substituted with one or more of-OH, —SH, halogen, or NH2, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is CH3;
    • 9) R4, R5, R6, and R7 are independently selected from CR8, R8 is R9 is C1-3 alkyl or OR9, 2 or 3 of R4, R5, R6, and R7 are CH, and R′ is CH3; and
    • 10) R4, R5, R6, and R7 are independently selected from CH, C(OH), C(SH), CNH2, C(CH3), C(OCH3), CF, CCF3, and C(CHCHBr), 2 or 3 of R4, R5, R6, and R7 are CH; and R′ is CH3.
  • A first series of preferred species of the second principal embodiment are defined when R1 and R2 are as defined above, R″ is C, R′ is NH, and:
    • 1) R4, R5, R6, and R7 are CH;
    • 2) R4 is CCH3, and R5, R6, and R7 are CH;
    • 3) R5 is CCH3, and R4, R6, and R7 are CH;
    • 4) R6 is CCH3, and R4, R5, and R7 are CH;
    • 5) R7 is CCH3, and R4, R5, and R6 are CH;
    • 6) R4 is COCH3, and R5, R6, and R7 are CH;
    • 7) R5 is COCH3, and R4, R6, and R7 are CH;
    • 8) R6 is COCH3, and R4, R5, and R7 are CH;
    • 9) R7 is COCH3, and R4, R5, and R6 are CH;
    • 10) R4 is CF, and R5, R6, and R7 are CH;
    • 11) R5 is CF, and R4, R6, and R7 are CH;
    • 12) R6 is CF, and R4, R5, and R7 are CH;
    • 13) R7 is CF, and R4, R5, and R6 are CH;
    • 14) R4 is COH, and R5, R6, and R7 are CH;
    • 15) R5 is COH, and R4, R6, and R7 are CH;
    • 16) R6 is COH, and R4, R5, and R7 are CH;
    • 17) R7 is COH, and R4, R5, and R6 are CH;
    • 18) 2 of R4, R5, R6 are R7 are CCH3, and 2 of R4, R5, R6, and R7 are CH;
    • 19) 2 of R4, R5, R6 are R7 are COCH3, and 2 of R4, R5, R6, and R7 are CH;
    • 20) 2 of R4, R5, R6 are R7 are CF, and 2 of R4, R5, R6, and R7 are CH; or
    • 21) 2 of R4, R5, R6 are R7 are COH, and 2 of R4, R5, R6, and R7 are CH;
  • A second series of preferred species of the second principal embodiment are defined when R″ is CH, R′ is NH2, and R4, R5, R6, and R7 are as defined in any one of the first series of preferred species.
  • A third series of preferred species of the present invention are defined when R″ is CH, R′ is CH3, and R4, R5, R6, and R7 are as defined in any one of the first series of preferred species.
  • In a third principal embodiment the compounds of the present invention are phenylthiol, phenylamine, and multicyclic-phenolic related compounds of the following structure (III):
    Figure US20050152859A1-20050714-C00007

    wherein:
    • 1) R1 is (CH2)nSR7, (CH2)nNHR7, or OR7;
    • 2) n is 0, 1, 2, or 3,
    • 3) R2, R3, R4, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NHSO2—C1-3alkyl, (vii) —NHCO—C1-5 alkyl, (viii) oxime, (ix) hydrazine, (x) —NR9R10, (xi) HSO2, (xii) HSO3, (xiii) thio-C1-5 alkyl, (xiv) C1-5 acyloxy, (xv) H2PO3, (xvi) thiol, (xvii) —COOR9, (xviii) C1-5 alkynyl, or (xix) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy;
    • 4) alternatively, R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, pyridine, —X—(CH2)n′—X—wherein n′ is 1 and X is nitrogen, sulfur, or oxygen, and —(CH)n″XH— wherein n″ is 2 and X is as defined above;
    • 5) R7 is (i) substituted or unsubstituted alkyl, alkenyl, aryl, or heterocycle, (ii) —C1-5 alkoxy, (iii) hydrogen, (iv) C(O)—C1-3 alkyl, or (v) —(CH2)mC(O)NR9R10;
    • 6) R9 is hydrogen or C1-3 alkyl;
    • 7) R10 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
    • 8) m is 1, 2, 3, 4, or 5; and
    • 9) provided that when R1 is OR7, R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, pyridine, —X—(CH2)n′—X— wherein n′ is 1 and X is nitrogen, sulfur, or oxygen, and —(CH)n″XH— wherein n″ is 2 and X is as defined above.
  • A first series of subembodiments of the third principal embodiment are defined when R1 is (CH2)nSR7, n is 0, 1, 2, or 3 but preferably 0, and:
    • 1) R2, R3, R4, R5 and R6 are as defined above, and R7 is hydrogen, C1-5 alkyl optionally substituted with —OH, or C(O)C1-3 alkyl;
    • 2) R2, R3, R4, R5 and R6 are as defined above, and R7 is hydrogen, C1-3 alkyl, or C(O)C1-3 alkyl;
    • 3) R2, R3, R4, R5 and R6 are as defined above, and R7 is hydrogen;
    • 4) R2, R3, R4, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (viii) —NR9R10, (xi) C1-5 acyloxy, (xii) thiol, (xiii) COOR9, or (xiv) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy, or (xv) —NHCO—C1-5 alkyl; and R7 is as defined above;
    • 5) R2, R3, R4, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR9, (viii) —NR9R9, (xi) C1-3 acyloxy, (xii) thiol, (xiii) COOR9, (xiv) —C1-3 alkyl, —C1-3 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R9, C1-3 thioether, or C1-3 alkoxy, or (xv) —NHCO—C1-3 alkyl; and R7 is as defined above;
    • 6) R2, R3, R4, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (v) —OR9, (viii) —NR9R9, (xii) thiol, (xiv) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2, or (xv) —NHCO—C1-3 alkyl; and R7 is as defined above;
    • 7) R2, R3, R4, R5 and R6 are independently selected from C1-3 alkyl, OR9, or —NHCO—CH3 alkyl; and R7 is as defined above;
    • 8) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, pyridine, —X—(CH2)n′—X—wherein n′ is 1 and X is nitrogen, sulfur, or oxygen, and —(CH)n″XH—wherein n″ is 2 and X is as defined above; and (b) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi —NR9R10, (vii) C1-5 acyloxy, (viii) thiol, (ix) COOR9, or (x) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy; and R7 is as defined above;
    • 9) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, pyridine, —X—(CH2)n′, —X—wherein n′ is 1 and X is nitrogen, sulfur, or oxygen, and —(CH)n″—XH— wherein n″ is 2 and X is as defined above; and (b) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR9, (vi) —NR9R9, (vii) C1-3 acyloxy, (viii) thiol, (ix) COOR9, or (x) —C1-3 alkyl, —C1-3 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R9, C1-3 thioether, or C1-3 alkoxy; and R7 is as defined above;
    • 10) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, pyridine, —X—(CH2)n′—X—wherein n′ is 1 and X is nitrogen, sulfur, or oxygen, and —(CH)n″XH— wherein n″ is 2 and X is as defined above; and (b) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) —OR9, (iv) —NR9R9, (v) thiol, or (vi) —C1-13 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2; and R7 is as defined above;
    • 11) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, pyridine, —X—(CH2)n′—X—wherein n′ is 1 and X is nitrogen, sulfur, or oxygen, and —(CH)n″XH—wherein n″ is 2 and X is as defined above; and (b) the remainder of R2, R3, R5 and R6 are independently selected from C1-3 alkyl or OR9; and R7 is as defined above;
    • 12) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, and pyridine; and (b) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NR9R10, (vii) C1-5 acyloxy, (viii) thiol, (ix) COOR9, or (x) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy; and R7 is as defined above;
    • 13) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, and pyridine; and (1) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR9, (vi) —NR9R9, (vii) C1-3 acyloxy, (viii) thiol, (ix) COOR9, or (x) —C1-3 alkyl, —C1-3 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R9, C1-3 thioether, or C1-3 alkoxy; and R7 is as defined above;
    • 14) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, or pyridine; and (b) the remainder of R2, R3, R5 and R4 are independently selected from (i) hydrogen, (ii) halogen, (iii) —OR9, (iv) —NR9R9, (v) thiol, or (vi) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2; and R7 is as defined above;
    • 15) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from phenyl, cyclopentyl, cyclohexyl, pyrrole, furan, thiophene, pyrazole, or pyridine; and (b) the remainder of R2, R3, R5 and R6 are independently selected from C1-3 alkyl or OR9; and R7 is as defined above;
    • 16) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from —X—(CH2)n′—X— wherein n′ is 1 and X is nitrogen, sulfur, or oxygen; and (b) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NR9R10, (vii) C1-5 acyloxy, (viii) thiol, (ix) COOR9, or (x) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy; and R7 is as defined above;
    • 17) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from —X—(CH2)n′—X— wherein n′ is 1 and X is nitrogen, sulfur, or oxygen; and (b) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR9, (vi) —NR9R9, (vii) C1-3 acyloxy, (viii) thiol, (ix) COOR9, or (x) —C1-3 alkyl, —C1-3 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R9, C1-3 thioether, or C1-3 alkoxy; and R7 is as defined above;
    • 18) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from —X—(CH2)n′—X— wherein n′ is 1 and X is nitrogen, sulfur, or oxygen; and (b) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) —OR9, (iv) —NR9R9, (v) thiol, or (vi) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2; and R7 is as defined above;
    • 19) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from —X—(CH2)n′—X— wherein n′ is 1 and X is nitrogen, sulfur, or oxygen; and (b) the remainder of R2, R3, R5 and R6 are independently selected from C1-3 alkyl or OR9; and R7 is as defined above;
    • 20) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from —(CH)n″XH— wherein n″ is 2 and X is nitrogen, sulfur, or oxygen; and (b) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR10, (vi) —NR9R10, (vii) C1-5 acyloxy, (viii) thiol, (ix) COOR9, or (x) —C1-5 alkyl, —C1-5 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R10, C1-5 thioether, or C1-5 alkoxy; and R7 is as defined above;
    • 21) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from —(CH)n″XH— wherein n″ is 2 and X is nitrogen, sulfur, or oxygen; and (b) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) NO2, (iv) —CN, (v) —OR9, (vi) —NR9R9, (vii) C1-3 acyloxy, (viii) thiol, (ix) COOR9, or (x) —C1-3 alkyl, —C1-3 alkenyl, aryl, heteroaryl, or heterocycle, optionally substituted with one or more of —OH, —SH, C(O)H, COOR9, C1-5 acyloxy, halogen, NR9R9, C1-3 thioether, or C1-3 alkoxy; and R7 is as defined above;
    • 22) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from —(CH)n″XH— wherein n″ is 2 and X is is nitrogen, sulfur, or oxygen; and (b) the remainder of R2, R3, R5 and R6 are independently selected from (i) hydrogen, (ii) halogen, (iii) —OR9, (iv) —NR9R9, (v) thiol, or (vi) —C1-3 alkyl or alkenyl optionally substituted with one or more of —OH, —SH, halogen, or NH2; and R7 is as defined above;
    • 23) (a) R3 and R4, or R4 and R5, combine to form a fused ring-structure which is cycloalkyl, aryl, or heterocyclic selected from —(CH)n″XH— wherein n″ is 2 and X is nitrogen, sulfur, or oxygen; and (b) the remainder of R2, R3, R5 and R5 are independently selected from C1-3 alkyl or OR9; and R7 is as defined above;
  • A second series of subembodiments is defined when R1 is (CH2)nSR7, n is 0, 1, 2, or 3, R7 is C1-5 alkyl optionally substituted with —OH, or C(O)C1-3 alkyl; and R2, R3, R4, R5 and R6 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments. A subset of the second series of embodiments is defined when R1 is SR7, R7 is C1-5 alkyl optionally substituted with —OH, or C(O)C1-3 alkyl; R4, is —NHCO—C1-3 alkyl; and R2, R3, R5 and R6 are CH.
  • A third series of subembodiments is defined when R1 is (CH2)nSR7, n is 0, 1, 2, or 3, R7 is C1-3 alkyl, or C(O)C1-3 alkyl; and R2, R3, R4, R5 and R6 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments. Preferably, R4, is —NHCO—C1-3 alkyl; and R2, R3, R5 and R6 are CH.
  • A fourth series of subembodiments is defined when R1 is (CH2)nSR7, n is 0, 1, 2, or 3, R7 is hydrogen; and R2, R3, R4, R5 and R6 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments. Preferably, R4, is —NHCO—C1-3 alkyl; and R2, R3, R5 and R6 are CH.
  • A fifth series of subembodiments is defined when R1 is SR7, R7 is C1-5 alkyl optionally substituted with —OH; and R2, R3, R4, R5 and R6 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments. Preferably, R4, is —NHCO—C1-3 alkyl; and R2, R3, R5 and R6 are CH.
  • A sixth series of subembodiments is defined when R1 is SR7, R7 is C1-3 alkyl; and R2, R3, R4, R5 and R4 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments. Preferably, R4, is —NHCO—C1-3 alkyl; and R2, R3, R5 and R6 are CH.
  • A seventh series of subembodiments is defined when R1 is SR7, R7 is hydrogen; and R2, R3, R4, R5 and R4 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments. Preferably, R4, is —NHCO—C1-3 alkyl; and R2, R3, R5 and R6 are CH.
  • An eighth series of subembodiments is defined when R1 is (CH2)nNHR7, n is 0, 1, 2, or 3, R7 is C1-5 alkyl optionally substituted with —OH; and R2, R3, R4, R5 and R6 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments.
  • A ninth series of subembodiments is defined when R1 is (CH2)nNHR7, n is 0, 1, 2, or 3, R7 is C1-3 alkyl; and R2, R3, R4, R5 and R6 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments.
  • An tenth series of subembodiments is defined when R1 is (CH2)nNHR7, n is 0, 1, 2, or 3, R7 is hydrogen; and R2, R3, R4, R5 and R6 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments.
  • An eleventh series of subembodiments is defined when R1 is NHR7, R7 is C1-5 alkyl optionally substituted with —OH; and R2, R3, R4, R5 and R6 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments.
  • A twelfth series of subembodiments is defined when R1 is NHR7, R7 is C1-3 alkyl; and R2, R3, R4, R5 and R6 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments.
  • A thirteenth series of subembodiments is defined when R1 is NHR7, R7 is hydrogen; and R2, R3, R4, R5 and R6 are as defined in any one of the 4th through 23d subembodiments of the first series of subembodiments.
  • An fourteenth series of subembodiments is defined when R1 is OR7, R7 is C1-5 alkyl optionally substituted with —OH; and R2, R3, R4, R5 and R6 are as defined in any one of the 8th through 23d subembodiments of the first series of subembodiments.
  • A fifteenth series of subembodiments is defined when R1 is OR7, R7 is C1-3 alkyl; and R2, R3, R4, R5 and R6 are as defined in any one of the 8th through 23d subembodiments of the first series of subembodiments.
  • A sixteenth series of subembodiments is defined when R1 is OR7, R7 is hydrogen; and R2, R3, R4, R5 and R6 are as defined in any one of the 8th through 23d subembodiments of the first series of subembodiments.
  • A first series of species of the second principal embodiment are defined when R1 is SH or SC(O)CH3, and:
    • 1) R2 is OCH3, and R3, R4, R5 and R6 are CH.
    • 2) R3 is OCH3, and R2, R4, R5 and R6 are CH.
    • 3) R4 is OCH3, and R2, R3, R5 and R6 are CH.
    • 4) R5 is OCH3, and R2, R3, R4 and R6 are CH.
    • 5) R6 is OCH3, and R2, R3, R4 and R5 are CH.
    • 6) 2 of R2, R3, R4, R5, and R6 are OCH3, and the remainder of R2, R3, R4, R5, and R6 are CH.
    • 7) R2 is SCH3, and R3, R4, R5 and R6 are CH.
    • 8) R3 is SCH3, and R2, R4, R5 and R6 are CH.
    • 9) R4 is SCH3, and R2, R3, R5 and R6 are CH.
    • 10) R5 is SCH3, and R2, R3, R4 and R6 are CH.
    • 11) R6 is SCH3, and R2, R3, R4 and R5 are CH.
    • 12) 2 of R2, R3, R4, R5, and R6 are SCH3, and the remainder of R2, R3, R4, R5, and R6 are CH.
    • 13) R2 is NHC(O)CH3, and R3, R4, R5 and R6 are CH.
    • 14) R3 is NHC(O)CH3, and R2, R4, R5 and R6 are CH.
    • 15) R4 is NHC(O)CH3, and R2, R3, R5 and R6 are CH.
    • 16) R5 is NHC(O)CH3, and R2, R3, R4 and R6 are CH.
    • 17) R6 is NHC(O)CH3, and R2, R3, R4 and R5 are CH.
  • A second series of preferred species are defined when R1 is NH2, and R2, R3, R4, R5 and R6 are as defined in any one of species 1-17 of the first series of preferred embodiments.
  • A third series of preferred species are defined when R1 is NHC(O)CH3, and R2, R3, R4, R5 and R6 are as defined in any one of species 1-17 of the first series of preferred embodiments.
  • In a fourth principal embodiment the compounds of the present invention are defined by structures (IV) or (V):
    Figure US20050152859A1-20050714-C00008

    wherein:
    • 1) R1, R2, and R3 are independently (i) substituted or unsubstituted alkyl, alkenyl, aryl, or heterocycle, (ii) hydrogen, (iii) C(O)—C1-3 alkyl, or (iv) —(CH2)1-5C(O)NR9R10;
    • 2) R9 is hydrogen or C1-3 alkyl;
    • 3) R10 is hydrogen, or C1-5 alkyl optionally substituted with —OH;
    • 4) Y and Y′ are independently oxygen or sulfur;
    • 5) X is oxygen, sulfur, or nitrogen; and
    • 6) R4 is C1-5 alkyl, optionally substituted by —OH, or NR9R9.
  • A first series of subembodiments of the fourth principal embodiment are defined by structure (IV) when Y and Y′ are as described above, and:
    • 1. R1 is hydrogen, and R2 and R3 are C1-5 alkyl optionally substituted with —OH; and
    • 2. R1 is hydrogen, and R2 and R3 are hydrogen or C1-3 alkyl.
  • A second series of subembodiments of the fourth principal embodiment are defined by structure (V) when:
    • 1. X is sulfur, R1 is hydrogen, and R2 is C1-5 alkyl optionally substituted with —OH;
    • 2. X is sulfur, R1 is hydrogen, and R2 is hydrogen or C1-3 alkyl;
    • 3. X is sulfur, R1 is hydrogen, R2 is C1-5 alkyl optionally substituted with —OH; and R4 is unsubstituted (CH2)1-5; or
    • 4. X is sulfur, R1 is hydrogen, R2 is hydrogen or C1-3 alkyl, and R4 is unsubstituted (CH2)1-3.
  • Preferred species are defined for structure (IV) when Y is sulfur, Y′ is oxygen, R1 and R2 are hydrogen, and R3 is methyl, and for structure (V) when X is sulfur, R4 is ethylene and R1 and R2 are hydrogen.
  • The compounds of this invention can be optionally substituted with substituents that do not adversely affect the activity of the compound as a skin lightener. Nonlimiting examples of substituents include, but are not limited to, alkyl (including lower alkyl), heteroalkyl, aryl, heterocyclic (including heteroaryl and heterocycloalkyl), halo, hydroxyl, carboxyl, acyl, acyloxy, amino, alkylamino, arylamino, alkoxy, aryloxy, alkylthio, alkylamido, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al., Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991. It will be understood that the present invention also covers “prodrugs” for such compositions, and pharmaceutically acceptable salts and esters thereof.
  • Properties of the Compounds of the Present Invention
  • In the present invention, one or all of three in vitro bioassays can be utilized to evaluate the efficacy and toxicity of candidate skin-lightening compounds. The three bioassays characterize the compounds with regard to mammalian tyrosinase enzyme inhibition (cell free), pigmentation in melanocyte cultured cells, and cytotoxicity of mammalian cultured cells. Both cell-based pigmentation and cell-free enzymatic assays have been developed [5, 6, 25] using the mammalian melanocyte cell line, Mel-Ab, a C57BL/6 mouse-derived cell line that produces high levels of melanin. [21] A distinct advantage of this approach is that humans share substantial sequence similarities in their genes (DNA) and proteins (such as tyrosinase) with mice, relative to non-mammalian species (e.g., mushrooms). So, mouse Mel-Ab melanocytes can serve as adequate surrogates for human melanocytes for many pharmacologic purposes.
  • These adherent murine melanocytes are grown on tissue culture plastic in medium supplemented with fetal bovine serum, 12-O-tetradecanoylphorbol-13-acetate (TPA) to stimulate cell division via down-regulation of protein kinase C, [22, 23] and cholera toxin to stimulate adenylate cyclase activity in the absence of α-MSH. [15, 24] Cellular lysates of Mel-Ab cells may be used as tyrosinase enzyme preparations. Multi-well plate assays have been validated [5, 6, 25] for enzyme inhibition (e.g., DOPA oxidation by colorimetric measurement or radiolabeled substrate incorporation into melanin) and for pigmentation assays on cultured Mel-Ab cells. After 4 days of treatment of cultured cells, melanin content is determined using a spectrophotometer at 400+ nm. [6, 25] This assay can detect an apparent loss in pigmentation resulting from either inhibition of de novo synthesis (e.g. via inhibition of tyrosinase, or the adenylate cyclase pathway, or another pathway) or a cytostatic/cytotoxic mechanism. It is therefore a broad primary screen. It is used in parallel with the tyrosinase enzymatic assay to determine whether an inhibitor of pigmentation at the cellular level is acting primarily at the enzyme level.
  • To determine cytotoxicity, crystal violet or other staining methods may be used to quantify adherent cell numbers following a period of treatment by an agent. HQ is typically used as a positive control in the assay, since it exhibits an IC50 in the low micrograms per milliliter range on Mel-Ab culture using this assay, albeit owing to cytotoxicity and not inhibition of pigmentation per se. [6] It should be noted that many inhibitors identified in cell-free enzymatic assays might have subsequent difficulties with toxicity or delivery in melanocyte cell-based assays. Therefore, all three in vitro assays in combination provide an excellent characterization of candidate skin lightening compounds.
  • A distinct advantage of the screening systems (developed by the inventors of the present invention) is the focus on mammalian tyrosinase, as opposed to non-mammalian enyzmes often used by other investigators, such as mushroom tyrosinase. Since the biochemical and pharmacologic characteristics of an enzyme or isozyme can vary dramatically between species of organisms (e.g., due to dissimilarities in primary, secondary, and tertiary structure), it is highly preferable that candidate topical skin lighteners intended for human use be discovered based on their biochemical action against a mammalian source of the enzyme. Mushroom tyrosinase (and in some instances plant polyphenol oxidases) has been used in the vast majority of prior inhibitor studies. [28, 29] Yet fungal tyrosinase exhibits substantial dissimilarities from mammalian tyrosinase(s), and is viewed as a substantially inferior strategy for pharmacologic screening. Thus, the methods reported by the inventors of the present invention for screening against mammalian tyrosinase or within melanocytes is highly preferred over other possible screening strategies. [5, 6, 25].
  • The substrate kinetic “affinity” of mammalian tyrosinase for L-tyrosine is approximately KM=600 μM. A potentially effective candidate skin lightening agent is considered to be desirable, active, and/or functional if it renders 50% inhibition of mammalian tyrosinase enzyme activity, at concentrations below half the enzyme's “affinity” for tyrosine in cell-free enzyme extracts (IC50≦300 μM) and pigment production in melanocyte cell cultures (IC50≦300 μM). In preferred embodiments the agent has an IC50 against tyrosinase in cell-free enzyme extracts of less then 200, 100, 50, or 25 μM, and/or an IC50 against pigment production in melanocyte cell cultures of less than 200, 100, 50, or 25 μM. In addition, it is desirable for the compounds to exhibit minimal cytotoxicity, e.g., thus retaining viability of 50% or more of the cultured cells (IC50≧300 μM), as evidenced by adherent cell number. In preferred embodiments the agent exhibits toxicity at greater than 500, 750, or 1000 μM.
  • Curto, E. V., et al. (1999) [25] reports that methyl gentisate is an “effective” candidate skin-lightening agent based on in vitro bioassays, because it has an IC50 of 11.2±4 (ug/mL) against tyrosinase activity in cell-free assays, an IC50 of 30.9±5 (ug/mL) in melanocyte cell cultures, and melanocyte cytotoxicity IC50 of 118.7±12 (ug/mL). Methyl gentisate thus poses a standard, against which the efficacy and cytotoxicity of other tyrosinase inhibiting compounds can be evaluated. By contrast to MG, hydroquinone is an inferior standard, exhibiting potent cytotoxicity and minimal enzymatic inhibition. [5, 6, 25].
  • Significantly, many of the particular compounds of this invention are comparable to or a more effective candidate skin lightening agents than methyl gentisate. Thus, in another embodiment the invention provides methods for inhibiting pigment production that includes administering an effective treatment amount of a pigment inhibiting compound wherein (i) the compound inhibits tyrosinase activity equivalent to or greater than methyl gentisate in cell-free enzyme extracts from mammalian melanocyte or melanoma cells, when evaluated using either a colorometric DOPA oxidation or a radiolabeled tyrosine or DOPA substrate assay as described in Curto, E. V., et al. (1999) [25], or (ii) the compound inhibits de novo pigment production (synthesis and/or accumulation) equivalent to or greater than methyl gentisate when evaluated in cultured mammalian melanocyte or melanoma cells. Curto, E. V., et al. (1999) [25]. In a preferred embodiment the toxicity of the compound in mammalian melanocyte, melanoma, or other cell cultures is equivalent to or less than the toxicity of methyl gentisate. Curto, E. V., et al. (1999) [25].
  • In another embodiment computer-based molecular orbital predictions can aid in the understanding and predictability of structure-activity relationships, such that other effective compounds can be identified and evaluated. See Sakurada, J., et al., “Kinetic and molecular orbital studies on the rate of oxidation of monosubstituted phenols and anilines by horseradish peroxidase compound II.” Biochemistry 29: 4093-4098 (1990) [26].
  • Definitions and Use of Terms
  • The following definitions and term construction are intended, unless otherwise indicated:
  • Specific and preferred values listed below for radicals, substituents, and ranges, are for illustration only; they do not exclude other defined values or other values within defined ranges for the radicals and substituents.
  • Halo is fluoro, chloro, bromo, or iodo.
  • Alkyl, alkoxy, alkenyl, alkynyl, etc. denote both straight and branched groups; but reference to an individual radical such as “propyl” embraces only the straight chain radical, a branched chain isomer such as “isopropyl” being specifically referred to.
  • The term alkyl, as used herein, unless otherwise specified, refers to a saturated straight, branched, or cyclic, primary, secondary, or tertiary hydrocarbon of C1 to C10, and specifically includes methyl, ethyl, propyl, isopropyl, cyclopropyl, butyl, isobutyl, t-butyl, pentyl, cyclopentyl, isopentyl, neopentyl, hexyl, isohexyl, cyclohexyl, cyclohexylmethyl, 3-methylpentyl, 2,2-dimethylbutyl, and 2,3-dimethylbutyl. When the context of this document allows alkyl to be substituted, the moieties with which the alkyl group can be substituted are selected from the group consisting of hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, aryl, heterocycle, halo, carboxy, acyl, acyloxy, amido, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al., Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference.
  • The term lower alkyl, as used herein, and unless otherwise specified, refers to a C1 to C4 saturated straight, branched, or if appropriate, a cyclic (for example, cyclopropyl) alkyl group, including both substituted and unsubstituted forms. Unless otherwise specifically stated in this application, when alkyl is a suitable moiety, lower alkyl is preferred. Similarly, when alkyl or lower alkyl is a suitable moiety, unsubstituted alkyl or lower alkyl is preferred.
  • The terms alkenyl and alkynyl refer to alkyl moieties, including both substituted and substituted forms, wherein at least one saturated C—C bond is replaced by a double or triple bond. Thus, (C2-C6)alkenyl can be vinyl, allyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, or 5-hexenyl. Similarly, (C2-C6)alkynyl can be ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, or 5-hexynyl.
  • The term “alkylene” refers to a saturated, straight chain, divalent alkyl radical of the formula —(CH2)n—, wherein n can be 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • As used herein, with exceptions as noted, “aryl” is intended to mean any stable monocyclic, bicyclic or tricyclic carbon ring of up to 8 members in each ring, wherein at least one ring is aromatic as defined by the Huckel 4n+2 rule. Examples of aryl ring systems include phenyl, naphthyl, tetrahydronaphthyl, and biphenyl. The aryl group can be substituted with one or more moieties selected from the group consisting of hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, alkyl, heterocycle, halo, carboxy, acyl, acyloxy, amido, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al., Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • The term heterocycle or heterocyclic, as used herein except where noted represents a stable 5- to 7-membered monocyclic or stable 8- to 1-membered bicyclic heterocyclic ring which is either saturated or unsaturated, including heteroaryl, and which consists of carbon atoms and from one to three heteroatoms selected from the group consisting of N, O, S, and P; and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • Nonlimiting examples of heteroaryl and heterocyclic groups include furyl, furanyl, pyridyl, pyrimidyl, thienyl, isothiazolyl, imidazolyl, tetrazolyl, pyrazinyl, benzofuranyl, benzothiophenyl, quinolyl, isoquinolyl, benzothienyl, isobenzofuryl, pyrazolyl, indolyl, isoindolyl, benzimidazolyl, purinyl, carbazolyl, oxazolyl, thiazolyl, isothiazolyl, 1,2,4-thiadiazolyl, isooxazolyl, pyrrolyl, quinazolinyl, cinnolinyl, phthalazinyl, xanthinyl, hypoxanthinyl, thiophene, furan, pyrrole, isopyrrole, pyrazole, imidazole, 1,2,3-triazole, 1,2,4-triazole, oxazole, isoxazole, thiazole, isothiazole, pyrimidine or pyridazine, and pteridinyl, aziridines, thiazole, isothiazole, 1,2,3-oxadiazole, thiazine, pyridine, pyrazine, piperazine, pyrrolidine, oxaziranes, phenazine, phenothiazine, morpholinyl, pyrazolyl, pyridazinyl, pyrazinyl, quinoxalinyl, xanthinyl, hypoxanthinyl, pteridinyl, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, pyrazolopyrimidinyl, adenine, N6-alkylpurines, N6-benzylpurine, N6-halopurine, N6-vinypurine, N6-acetylenic purine, N6-acyl purine, N6-hydroxyalkyl purine, N6-thioalkyl purine, thymine, cytosine, 6-azapyrimidine, 2-mercaptopyrmidine, uracil, N5-alkyl-pyrimidines, N5-benzylpyrimidines, N5-halopyrimidines, N5-vinyl-pyrimidine, N5-acetylenic pyrimidine, N5-acyl pyrimidine, N5-hydroxyalkyl purine, and N6-thioalkyl purine, and isoxazolyl. The heteroaromatic and heterocyclic moieties can be optionally substituted as described above for aryl, including substituted with one or more substituents selected from hydroxyl, amino, alkylamino, arylamino, alkoxy, aryloxy, alkyl, heterocycle, halo, carboxy, acyl, acyloxy, amido, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al., Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
  • The heteroaromatic can be partially or totally hydrogenated as desired. As a nonlimiting example, dihydropyridine can be used in place of pyridine. Functional oxygen and nitrogen groups on the heteroaryl group can be protected as necessary or desired. Suitable protecting groups are well known to those skilled in the art, and include trimethylsilyl, dimethylhexylsilyl, 1-butyldi-methylsilyl, and t-butyldiphenylsilyl, trityl or substituted trityl, alkyl groups, acyl groups such as acetyl and propionyl, methanesulfonyl, and p-toluenesulfonyl.
  • The term acyl refers to a carboxylic acid ester in which the non-carbonyl moiety of the ester group is selected from straight, branched, or cyclic alkyl or lower alkyl, alkoxyalkyl including methoxymethyl, aralkyl including benzyl, aryloxyalkyl such as phenoxymethyl, aryl including phenyl optionally substituted with halogen, C1 to C4 alkyl or C1 to C4 alkoxy, sulfonate esters such as alkyl or aralkyl sulphonyl including methanesulfonyl, the mono, di or triphosphate ester, trityl or monomethoxytrityl, substituted benzyl, trialkylsilyl (e.g. dimethyl-t-butylsilyl) or diphenylmethylsilyl. Aryl groups in the esters optimally comprise a phenyl group. The term “lower acyl” refers to an acyl group in which the non-carbonyl moiety is lower alkyl.
  • The term alkoxy, as used herein, and unless otherwise specified, refers to a moiety of the structure —O-alkyl, wherein alkyl is as defined above.
    Synthetic Methods
    Figure US20050152859A1-20050714-C00009
    • Precursor: Mono- or multiple-substituted benzene. Most are commercially available or can be easily prepared from commercial compounds. The definition of benzene ring substituents R1, R2, R3 and R4 is given in formulas (1) and (II) in section of Summary of The Invention.
    • Reactants: Nitric acid, Zinc, Hydrochloric acid, Carbon disulfide, Methyl isothiocyanate, Thiourea, Sulfur, Sodium diethyldithiocarbamate, Selenourea.
    • Solvents: 1,4-Dioxane, Toluene, Pyridine, Dichloromethane, Tetrahydrofuran, Water.
    • References: Saxena, D. B.; Khajuria, R. K; Suri, O. P. Synthesis and Spectral Studies of 2-Mercaptobenzimidazole Derivatives. J. Heterocycl. Chem., 19, 681-683, (1982).
  • The 1,2-phenylenediamine derivatives (V) can be prepared by twice nitration/reduction reactions on substituted benzene (I), some substituents may need protection under above reaction conditions. Cyclization of (V) with CS2, or CH3NCS, or thiourea, or S, or (C2H5)2NCS2Na can give the desired 2-mercaptobenzimidazole derivatives (VI). Reaction of (VI) with R5X (R5 can be alkyl or acyl group; X is Cl, Br, I) can produce alkylated products (VIII). 2-Benzimidazoline-selenium derivatives (VI) and (DC) can be synthesized similarly by reacting selenourea with (V).
    Figure US20050152859A1-20050714-C00010
    • Precursor: Substituted benzene. Most are commercially available or can be easily prepared from commercial compounds. The definition of benzene ring substituents R1, R2, R3, R4 and R5 is given in formulas (I) and (II) in section of Summary Of The Invention.
    • Reactants: Nitric acid, Zinc, Hydrochloric acid, Ammonium thiocyanate, Potassium thiocyanate, Potassium selenocyanate.
    • Solvents: Acetonitrile, Pyridine, Dichloromethane, Tetrahydrofuran, Water.
    • References: Rasmussen, C. R.; Villani, F. J., Jr.; Weaner, L. E.; Reynolds, B. E.; Hood, A. R.; Hecker, L. R.; Nortey, S. O.; Hanslin, A.; Costanzo, M. J.; et al. Improved Procedures for the Preparation of Cycloalkyl-, and Arylalkyl-, and Arylthioureas. Synthesis, 6,456-459, (1988).
  • Various arylthiourea compounds (IV) can be prepared by reaction of corresponding aniline (III) with NH4SCN or KSCN in aqueous HCl solution. Alkylation of (IV) by R6X (R6 can be alkyl or acyl group; X is Cl, Br, I) can yield monoalkylated product (VI). By replacing KSCN with KSeCN, the selenium analogous (V) can also be prepared.
    Figure US20050152859A1-20050714-C00011
    • Precursor: Substituted benzene. Most are commercially available or can be easily prepared from commercial compounds. The definition of benzene ring substituents R1, R2, R3, R4 and R5 is given in formulas (1) and (II) in section of Summary Of The Invention.
    • Reactants: Chlorosulfonic acid, Dichlorodimethylsilane, Zinc, Cl(CH2)nCl (n is 1-3), Aluminum chloride, Thiourea, Sodium hydroxide.
    • Solvents: Tetrahydrofuran, Benzene, Dimethyl sulfoxide, Water.
    • References: Uchiro, H.; Kobayashi, S. Non-aqueous Reduction of Aromatic Sulfonyl Chlorides to Thios Using a Dichlorodimethylsilane-zinc-dimethylacetamide System. Tetrahedron Lett., 40, 3179-3182, (1999).
  • Substituted arylsulfonyl chlorides (II) can be easily prepared from substituted aromatic compounds (I) by reaction with excess chlorosulfonic acid. Reduction of (II) with dichlorodimethylsilane/zinc will give desired phenylthiole derivatives (E). The substituted phenylalkyl mercaptans (VI) can be prepared from the corresponding chloro compounds (V) which can be obtained from alkylation reaction of (1) (Friedel-Crafts reaction). Both thiole compounds (II) and (VI) can react with alkyl halide R6X to form the corresponding sulfides (IV) and (VII).
    Figure US20050152859A1-20050714-C00012
    • Precursor: Substituted benzene. Most are commercially available or can be easily prepared from commercial compounds. The definition of benzene ring substituents R1, R2, R3, R4 and R5 is given in formulas (1) and (II) in section of Summary Of The Invention.
    • Reactants: Nitric acid, Zinc, Hydrochloric acid, Br(CH2)nBr (n is 1-3), Aluminum chloride.
    • Solvents: Benzene, Tetrahydrofuran, Diethyl ether, Water.
  • The preparation of products (II), (IV) and (V) is same as described previously. Reaction of (V) with alkyl amine R6NH2 (R6 is hydrogen or alkyl) can give arylalkylamine derivatives (VI).
    Figure US20050152859A1-20050714-C00013
    • Precursor: Substituted thiophene. Most are commercially available or can be easily prepared from commercial sources. The definition of ring substituents R1, R2 and R3 is same as that given in formulas (I) and (II) in section of Summary of The Invention.
    • Reactants: Butyllithium, Cl(CH2)nNMe2 (n is 1-3), Ethyl chloroformate.
    • Solvents: Diethyl ether, Tetrahydrofuran, Benzene.
    • References: Hallberg, A.; Gronowitz, S. On The Reaction of Some Thienyllithium Derivatives with 1-Chloro-2-dimethylaminoethane. Chem. Scr., 16, 42-46, (1980).
  • Reaction of substituted thiophene with butyllithium can yield 2-thienyllithium salt (II), protection may be necessary for some substituents. Substituted 2-thiophenealkylamine (III) can be prepared by reaction of (E) with 1-chloro-2-dimethylaminoalkane. The products (III), (IV) and (V) can be converted to each other by alkylation/dealkylation reactions using alkyl halide R4X and ClCO2Et, respectively.
    Figure US20050152859A1-20050714-C00014
    • Precursor: Substituted benzene. Most are commercially available or can be easily prepared from commercial compounds. The definition of benzene ring substituents R1, R2, R3, R4 and R5 is given in formulas (I) and (M) in section of Summary Of The Invention.
    • Reactants: Potassium thiocyanate, Polyphosphoric acid, Sulfuric acid.
    • Solvents: Benzene, Water.
    • References: Sastry, S.; Kudav, N. A. One-step Synthesis of Aromatic Thio Amides: Reaction of Aromatic Compounds with Potassium Thiocyanate in Polyphosphoric Acid or Sulfuric Acid. Indian J. Chem., Sect. B, 18B, 455, (1979).
  • Benzothioamide derivatives (II) can be prepared from substituted benzene (I) in one single step by reaction with KSCN in polyphosphoric acid or sulfuric acid. The alkylated product (III can be obtained by using alkyl halide R6X (X is Cl, Br, I).
  • Pharmaceutical Formulations and Dosing Regimes
  • In one embodiment, a compound of this invention is applied or administered to the skin during an appropriate period and using a sufficient number of dosages to achieve skin lightening. The concentration of active compound in the composition will depend on absorption, inactivation, and excretion rates of the compound as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. The active ingredient may be administered as a single dose, or may be divided into a number of smaller doses to be administered at varying intervals of time.
  • Topical and other formulations of these active and/or functional compounds are of utility in lightening skin pigmentation in humans and other animals. These formulations may be useful for pure cosmetic purposes, simply to obtain a lighter skin color for perceived beautification. The formulations also have medicinal value and can, for example, decrease hyperpigmentation of melasma, age spots, freckles, and other skin blemishes. The compounds of this invention act primarily by inhibiting mammalian melanocyte tyrosinase, the rate-limiting enzyme in the production of melanin from tyrosine and DOPA. Some compounds also absorb ultraviolet radiation (UVR), and may thus protect skin from UVR and photoaging. In addition, some compounds may be antioxidants that protect skin from oxidative damage, and/or may prevent oxidative decomposition of product formulations.
  • If desirable these formulations could also be used to reduce pigmentation in hair, albeit during the biosynthesis of hair, by blocking pigment production within the melanocytes of hair follicles. The formulations would likely not affect the already emerged pigmented portions of hair, unlike a bleaching agent.
  • The formulations useful in the present invention contain biologically effective amounts of the functional and/or active compound(s). A biologically effective amount of the active compound is understood by those skilled in the art to mean that a sufficient amount of the compound in the composition is provided such that upon administration to the human or animal by, for example, topical route, sufficient active agent is provided on each application to give the desired result. However, the biologically effective amount of the active compound is at a level that it is not toxic to the human or animal during the term of treatment. By a suitable biologically compatible carrier, when the compound is topically applied, it is understood that the carrier may contain any type of suitable excipient in the form of cosmetic compositions, pharmaceutical adjuvants, sunscreen lotions, creams, and the like. In one embodiment the active compound is administered in a liposomal carrier.
  • The active compound is administered for a sufficient time period to alleviate the undesired symptoms and the clinical signs associated with the condition being treated, or to achieve the level of desired skin lightening. The individual dosage, dosage schedule, and duration of treatment may be determined by clinical evaluations by those of skill in the art.
  • Solutions or suspensions for topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates; and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • Suitable vehicles, carriers, or formulations for topical application are known, and include lotions, suspensions, ointments, oil-in-water emulsions, water-in-oil emulsions, creams, gels, tinctures, sprays, powders, pastes, and slow-release transdermal or occlusive patches. Thickening agents, emollients, and stabilizers can be used to prepare topical compositions. Examples of thickening agents include petrolatum, beeswax, xanthan gum, or polyethylene glycol, humectants such as sorbitol, emollients such as mineral oil, lanolin and its derivatives, or squalene. A number of solutions and ointments are commercially available, especially for dermatologic applications.
  • The compounds can be provided in the form of pharmaceutically-acceptable salts. As used herein, the term “pharmaceutically-acceptable salts or complexes” refers to salts or complexes that retain the desired biological activity of the parent compound and exhibit minimal, if any, undesired toxicological effects. Examples of such salts are (a) acid addition salts formed with inorganic acids (for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like), and salts formed with organic acids such as acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acids, naphthalenedisulfonic acids, and polygalacturonic acid; (b) base addition salts formed with polyvalent metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium, and the like, or with an organic cation formed from N,N-dibenzylethylene-diamine or ethylenediamine; or (c) combinations of (a) and (b); e.g., a zinc tannate salt or the like.
  • The compounds can be modified in order to enhance their usefulness as pharmaceutical compositions. For example, it is well know in the art that various modifications of the active molecule, such as alteration of charge, can affect water and lipid solubility and thus alter the potential for percutaneous absorption. The vehicle, or carrier, can also be modified to enhance cutaneous absorption, enhance the reservoir effect, and minimize potential irritancy or neuropharmacological effects of the composition. See, in general, Arndt, et al. [27].
  • Thus, the invention provides various formulations as topical skin lighteners containing the active and/or functional compounds described above. The invention further provides formulations as topical anti-oxidants containing the active and/or functional compounds described above. In still another embodiment the invention provides formulations as topical sunscreens containing the active and/or functional compounds described above. Such formulations can be made in combination with other active and/or functional ingredients used in skincare products (e.g. organic or inorganic sunscreen, antioxidant, anti-inflammatory, anti-erythema, antibiotic, antimicrobial, humectant, or other ingredients). Other ingredients can be formulated with the compounds to augment their effect, including but not limited to Vitamin C, Vitamin E, magnesium ascorbyl phosphate, aloe vera extract, and retinoic acids. In addition, alpha-hydroxy acids can be included to speed up the skin lightening process by exfoliating surface colored skin.
  • The compounds of the present invention can also be formulated for alternative routes of administration other than topical application, including but not limited to general systemic, oral, intradermal, transdermal, occlusive patches, intravenous, or parenteral administration, and pharmaceutical compositions known generally to those skilled in the art.
  • The compounds can also be formulated along with other active and/or functional ingredients used in skincare products, depending on the intended use of the formulation. For example, the compounds can be formulated with organic or inorganic sunscreens, an antioxidant, an anti-inflammatory, an anti-erythema, an antibiotic, an antimicrobial, a humectant, or other ingredients.
  • The active and/or functional compounds described above may also be of use in inhibiting tyrosinase-like enzymes from non-mammalian species, for instance for use in the food science industry for the inhibition of enzymatic browning. [28, 29] Inhibition of plant polyphenol oxidases by agents described here may coincidentally have activity against these non-mammalian enzymes. Suitable formulations for spraying or treatment of fruits are known generally to those skilled in the art. Treatment by these formulations containing the enzyme inhibitors of the present invention might improve shelf life of plant or fungal foods.
  • EXAMPLES Example 1 Benzoimidazolethiols
  • A first class of compounds based upon the template compound benzimidazolethiol (lower left structure) were tested for tyrosinase inhibition, cell culture pigment inhibition, and toxicity, by methods described in Curto, E. V., et al. (1999) [25]. Results of the tests are given in Table 1.
    TABLE 1
    Figure US20050152859A1-20050714-C00015
    Figure US20050152859A1-20050714-C00016
    ID # R1 R2 R3 R5 R6 R E P T ε λmax
    138 NH SH N H H C 0.25 14300 300
    140 NH SH N CH3 H C 0.12 2.4 >1000 6300 305
    084 NH SH N OCH3 H C 0.07 1.6 >1000 10000 310
    040 S SH N H H C 8
    091 S SH N H OCH2CH3 C >1000 >1000 >1000
    205 NH ═S N(CO)CH3 H H C 0.5 8.3 35
    098 NH ═Se NH H H C 0.8 14 132
    135 NH ═S NH H H N 4 256 >1000

    *Inhibition [μM] as measured in three assays. Here “E” is the concentration of compound that produces 50% pigment inhibition in the cell-free mammalian-enzyme assay system. “P” is for the concentration of compound that produces 50% inhibition in the mammalian-melanocyte-culture pigment assay system. “T” is the concentration of compound that kills 50% of cells in the
    # mammalian-melanocyte-culture toxicity assay system. The compound extinction coefficient is ε [OD/M × cm] at the wavelength of maximum absorbency λmax [nm].
  • Example 2 Thiophenols
  • A second class of compounds based upon the template compound benzenethiol were tested for tyrosinase inhibition, cell culture pigment inhibition, and toxicity, by methods described in Curto, E. V., et al. (1999) [25]. Results of the tests are given in Table 2.
    TABLE 2
    Figure US20050152859A1-20050714-C00017
    ID # R1 R2 R3 R4 R5 E P T ε λmax
    099 H H OCH3 H H 53 85 202 3000 265
    102 H H H SCH3 H 0.24 115 126 2300 280
    083 H H H NH(CO)CH3 H 19 82 542 4700 265
    103 H H OCH3 OCH3 H 8 8 >1000 4300 250
    093 H OCH3 H H OCH3 500 200 200 2700 305
    148 (CO)CH3 H H NH(CO)CH3 H 500 30 125 3300 255

    *Inhibition [μM] as measured in three assays. Here “E” is the concentration of compound that produces 50% pigment inhibition in the cell-free mammalian-enzyme assay system. “P” is for the concentration of compound that produces 50% inhibition in the mammalian-melanocyte-culture pigment assay system. “T” is the concentration of compound that kills 50% of cells
    # in the mammalian-melanocyte-culture toxicity assay system. The compound extinction coefficient is ε [OD/M × cm] at the wavelength of maximum absorbency λmax [nm].
  • Example 3 Phenylthioureas
  • A third class of compounds based upon the template compound phenylthiourea (lower left structure) were tested for tyrosinase inhibition, cell culture pigment inhibition, and toxicity, by methods described in Curto, E. V., et al. (1999) [25]. Results of the tests are given in Table 3.
    TABLE 3
    Figure US20050152859A1-20050714-C00018
    Figure US20050152859A1-20050714-C00019
    ID # R2 R3 R4 R5 R E P T ε λmax
    033 H H H H NH2 2 12 >1000
    181 OCH3 H H H NH2 >1000 >1000
    105 H F H H NH2 1.52 1.78 >1000 11000 255
    104 H OH H H NH2 4 8 >1000
    131 H CH3 H H NH2 0.82 2.28 >1000
    053 H H OCH3 H NH2 8 30 60
    049 H H NH(CS)NH2 H NH2 4 250 >1000
    101 H CH3 H CH3 NH2 250 125 >1000
    054 H H H H CH3 16 16 >1000

    *Inhibition [μM] as measured in three assays. Here “E” is the concentration of compound that produces 50% pigment inhibition in the cell-free mammalian-enzyme assay system. “P” is for the concentration of compound that produces 50% inhibition in the mammalian-melanocyte-culture pigment assay system. “T” is the concentration of compound that kills 50% of cells in the
    # mammalian-melanocyte-culture toxicity assay system. The compound extinction coefficient is ε [OD/M × cm] at the wavelength of maximum absorbency λmax [nm].
  • Example 4 Miscellaneous
  • A fourth group of miscellaneous compounds of diverse structure were also tested for tyrosinase inhibition, cell culture pigment inhibition, and toxicity, by methods described in Curto, E. V., et al. (1999) [25]. Results of the tests are given in Table 4.
    TABLE 4
    Figure US20050152859A1-20050714-C00020
    Figure US20050152859A1-20050714-C00021
    Figure US20050152859A1-20050714-C00022
    Figure US20050152859A1-20050714-C00023
    1 2 3 4
    Figure US20050152859A1-20050714-C00024
    Figure US20050152859A1-20050714-C00025
    Figure US20050152859A1-20050714-C00026
    Figure US20050152859A1-20050714-C00027
    5 6 7 8
    # ID # E P T ε λmax
    1 081 5 81 500 1000 275
    2 100 32 62 >1000
    3 073 >1000 100 >1000
    4 079 73 71 472
    5 006 110 182 >1000
    6 092 79 236 >1000
    7 009 98 209 775
    8 026 54 153 367

    *Inhibition [μM] as measured in three assays. Here “E” is the concentration of compound that produces 50% pigment inhibition in the cell-free mammalian-enzyme assay system. “P” is for the concentration of compound that produces 50% inhibition in the mammalian-melanocyte-culture pigment assay system. “T” is the concentration of compound that kills 50% of cells in the
    # mammalian-melanocyte-culture toxicity assay system. The compound extinction coefficient is ε[OD/M × cm] at the wavelength of maximum absorbency λmax [nm].
  • Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains.
  • It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention. Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.
  • REFERENCES
    • 1. Hearing V J Jr., “Monophenol monooxygenase (tyrosinase): Purification, properties, and reactions catalyzed.” Methods Enzymol 142: 154-165, 1987.
    • 2. Spritz R A et al., “Genetic-disorders of pigmentation,” Adv Hum Genet 22: 1-45, 1994.
    • 3. Frenk E, “Treatment of melasma with depigmenting agents.” Melasma: New Approaches to Treatment, pp. 9-15. Martin Dunitz Ltd., London, 1995.
    • 4. Dooley T P, “Is there room for a moderate level of regularity oversight?” In: Drug Discovery Approaches for Developing Cosmeceuticals: Advanced Skin Care and Cosmetic Products (Ed. Hori W), Chap. 1.4. International Business Communications, Southborough, Mass., 1997.
    • 5. Dooley T P, “Topical skin depigmentation agents: Current products and discovery of novel inhibitors of melanogenesis.” J. Dermatol. Treat. 8: 275-279, 1997.
    • 6. Dooley T P, et al., “Development of an in vitro primary screen for skin depigmentation and antimelanoma agents.” Skin Pharmacol. 7: 188-200, 1994.
    • 7. Morse J L (Ed.), “An Abridgment of The New Funk & Wagnalls Encyclopedia,” The Universal Standard Encyclopedia, Vol, 10, pp. 3662-3663. Unicorn, N.Y., 1955.
    • 8. Budavari S (Ed.), “Gentisic acid,” Merck Index, 11th Edn, Abstract No. 4290, p. 688. Merck & Co., Rahway, N.J., 1989.
    • 9. J-Hua L, et al., “Direct analysis of salicylic acid, salicyl acyl glucuronide, salicyluric acid and gentisic acid in human plasma and urine by high-performance liquid chromatography.” J. Chromatogr. [B] 675: 61-70, 1996.
    • 10. Glatt H R, et al., “Multiple activation pathways of benzene leading to products with varying genotoxic characteristics.” Environ Health Perspect 82: 81-89, 1989.
    • 11. Glatt H R, “Endogenous mutagens derived from amino acids.” Mutat. Res. 238: 235-243, 1990.
    • 12. La Du B N, “Alcaptonuria and ochronotic arthritis.” Mol. Biol. Med. 8: 31-38, 1991.
    • 13 Hearing V J, “Mammalian monophenol monooxygenase (tyrosinase): purification, properties, and reactions catalyzed.” Methods Enzymol. 142: 154-65, 1987.
    • 14. Spritz R A, et al., “Genetic disorders of pigmentation.” Adv. Hum. Genet. 22: 1-45, 1994.
    • 15. Hadley M E et al, “Melanotropic peptides for therapeutic and cosmetic tanning of the skin.” N Y Acad. Sci. 680: 424-39, 1993.
    • 16. Sakai C et al, “Modulation of murine melanocyte function in vitro by agouti signal protein.” EMBO J. 16: 3544-52, 1997.
    • 17. Dooley T P, “Recent advances in cutaneous melanoma oncogenesis research.” Onco. Res. 6: 1-9, 1994.
    • 18. Benmaman O, et al., “Treatment and camouflaging of pigmentary disorders.” Clin. Dermatol. 6: 50-61, 1998.
    • 19. Zaumseil R-P, et al., “Topical azelaic acid in the treatment of melasma:
    • pharmacological and clinical considerations.” In: Castanet J, Frenk E, Gaupe K et al (Eds) Melasma: new approaches to treatment. Martin Dunitz: London, pp 16-40, 1995.
    • 20. Schallreuter K U, “Epidermal adrenergic signal transduction as part of the neuronal network in the human epidermis.” J. Invest. Dermatol. 2: 37-40, 1997.
    • 21. Bennett D C, et al., “A line of non-tumorigenic mouse melanocytes, syngeneic with the B16 melanoma and requiring a tumour promoter for growth.” Int. J. Cancer 349: 414-18, 1987.
    • 22. Dooley T P et al., “Polyoma middle T abrogates TPA requirement of murine melanocytes and induces malignant melanoma.” Oncogene 3: 531-6, 1988.
    • 23. Brooks G et al., “Protein kinase C down-regulation, and not transient activation, correlates with melanocyte growth.” Cancer Res. 51: 3281-8, 1991.
    • 24. O'Keefe E, et al., “Cholera toxin mimics melanocyte stimulating hormone in inducing differentiation in melanoma cells.” Proc. Natl. Acad. Sci. USA 71: 2500-4, 1974.
    • 25. Curto, E. V., et al., “Inhibitors of Mammalian Melanocyte Tyrosinase: In Vitro Comparisons of Alkyl Esters of Gentisic Acid with Other Putative Inhibitors.” Biochem. Pharmacol. 57: 663-672, 1999.
    • 26. Sakurada, J., et al., “Kinetic and molecular orbital studies on the rate of oxidation of monosubstituted phenols and anilines by horseradish peroxidase compound II.” Biochemistry 29: 4093-4098, 1990.
    • 27. Arndt, et al., “The Pharmacology of Topical Therapy”, Dermatology in General Medicine, 1987; T. B. Fitzpatrick, A. Z. Eisen, K. Wolff, I. M. Freedberg and K F. Austen, eds., 3d ed., McGraw Hill, Inc., New York, pp. 2532-2540.
    • 28. Lee, C. Y. and Whitaker, J. R. (Eds.) Enzymatic Browning and its Prevention. Pub. American Chemical Society, Washington, D.C., 1995.
    • 29. Lerch, K. “Tyrosinase: Molecular and active-site structure.” In Lee, C. Y. and Whitaker, J. R. (Eds.) Enzymatic Browning and its Prevention. Pub. American Chemical Society, Washington, D.C., pp. 64-80, 1995.
    • 30. Mishima, H., et al., “Fine structural demonstration of tyrosinase activity in the retinal pigment epithelium of normal and PTU-treated chick embryos.” Albrecht Von Graefes Arch. Klin. Exp. Ophthalmol. 211: 1-10, 1979.
    • 31. Dryja, T. P., et al., “Demonstration of tyrosinase in the adult bovine uveal tract and retinal pigment epithelium.” Invest. Ophthalmol. Vis. Sci. 17: 511-514, 1978.
    • 32. Higashi, Y., et al., “Inhibition of tyrosinase reduces cell viability in catecholaminergic neuronal cells.” J. Neurochem. 75: 1771-1774, 2000.

Claims (26)

1. A method for protecting skin from ultraviolet rays and photoaging comprising administering to a human in need of ultraviolet ray absorption for protection from ultraviolet rays or photoaging, an effective amount of at least one compound defined by the structure.
Figure US20050152859A1-20050714-C00028
and R10 is methyl, ethyl or propyl, or a pharmaceutically acceptable salt or ester thereof.
2. The method of claim 1 wherein R10 is methyl.
3. The method of claim 1 wherein R10 is ethyl.
4. The method of claim 1 wherein R10 is propyl.
5. The method of claim 1 wherein the compound has an IC50 against mammalian tyrosinase activity of less than or equal to 300 uM, and an IC50 of cytotoxicity in mammalian melanocytic cells of greater than 500 uM.
6. The method of claim 1 wherein the compound has an IC50 against melanin production in mammalian melanocytic cells of less than or equal to 300 uM, and an IC50 of cytotoxicity in mammalian melanocytic cells of greater than 500 uM.
7. The method of claim 1 wherein the compound is an antioxidant.
8. The method of claim 1 wherein the administration is through a topical formulation.
9. The method of claim 1 wherein the method further lightens skin pigmentation.
10. A method of inhibiting or preventing pigment production in a mammal comprising administering to the mammal an effective amount of a compound defined by the structure below, or a pharmaceutically acceptable salt or ester thereof:
Figure US20050152859A1-20050714-C00029
and R10 is methyl, ethyl or propyl.
11. The method of claim 10 wherein the compound has an IC50 against mammalian tyrosinase activity of less than or equal to 300 uM, and an IC50 of cytotoxicity in mammalian melanoctyic cells of greater than 500 uM.
12. The method of claim 10 wherein the compound has an IC50 against melanin production in mammalian melanocytic cells of less than or equal to 300 uM, and an IC50 of cytotoxicity in mammalian melanocytic cells of greater than 500 uM.
13. The method of claim 10 wherein the compound absorbs ultraviolet radiation.
14. The method of claim 10 wherein the compound is an antioxidant.
15. The method of claim 10 wherein the mammal is a human.
16. The method of claim 10 wherein the administration is through a topical formulation or an occlusive patch.
17. The method of claim 10 wherein the method is for lightening skin pigmentation
18. The method of claim 10 wherein the method is for treating hyperpigmentation-related medical conditions.
19. The method of claim 10 wherein R10 is methyl.
20. The method of claim 10 wherein R10 is ethyl.
21. The method of claim 10 wherein R10 is propyl.
22. A method of inhibiting tyrosine hydroxylase comprising administering an effective amount of a compound defined by the structure below or a pharmaceutically acceptable salt or ester thereof:
Figure US20050152859A1-20050714-C00030
and R10 is methyl, ethyl or propyl.
23. The method of claim 12 wherein the hyperpigmentation-related medical condition is melasma, age spots, freckles, ochronosis, postinflammatory hyperpigmentation, or lentigo.
24. The method of claim 22 wherein R10 is methyl.
25. The method of claim 23 wherein R10 is ethyl.
26. The method of claim 24 wherein R10 is propyl.
US11/075,922 2000-02-29 2005-03-10 Inhibitors of melanocyte tyrosinase as topical skin lighteners Abandoned US20050152859A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US11/075,922 US20050152859A1 (en) 2000-02-29 2005-03-10 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US12/206,334 US7718185B2 (en) 2000-02-29 2008-09-08 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US12/206,290 US7858105B2 (en) 2000-02-29 2008-09-08 Inhibitors of melanocyte tyrosinase as topical skin lighteners

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US18561000P 2000-02-29 2000-02-29
US09/795,683 US20020044914A1 (en) 2000-02-29 2001-02-28 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US10/427,561 US20030219393A1 (en) 2000-02-29 2003-05-01 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US11/075,922 US20050152859A1 (en) 2000-02-29 2005-03-10 Inhibitors of melanocyte tyrosinase as topical skin lighteners

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US09/795,683 Continuation US20020044914A1 (en) 2000-02-29 2001-02-28 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US10/427,561 Continuation US20030219393A1 (en) 2000-02-29 2003-05-01 Inhibitors of melanocyte tyrosinase as topical skin lighteners

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US12/206,290 Division US7858105B2 (en) 2000-02-29 2008-09-08 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US12/206,334 Continuation US7718185B2 (en) 2000-02-29 2008-09-08 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US12/206,334 Division US7718185B2 (en) 2000-02-29 2008-09-08 Inhibitors of melanocyte tyrosinase as topical skin lighteners

Publications (1)

Publication Number Publication Date
US20050152859A1 true US20050152859A1 (en) 2005-07-14

Family

ID=22681701

Family Applications (5)

Application Number Title Priority Date Filing Date
US09/795,683 Abandoned US20020044914A1 (en) 2000-02-29 2001-02-28 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US10/427,561 Abandoned US20030219393A1 (en) 2000-02-29 2003-05-01 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US11/075,922 Abandoned US20050152859A1 (en) 2000-02-29 2005-03-10 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US12/206,334 Expired - Fee Related US7718185B2 (en) 2000-02-29 2008-09-08 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US12/206,290 Expired - Fee Related US7858105B2 (en) 2000-02-29 2008-09-08 Inhibitors of melanocyte tyrosinase as topical skin lighteners

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/795,683 Abandoned US20020044914A1 (en) 2000-02-29 2001-02-28 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US10/427,561 Abandoned US20030219393A1 (en) 2000-02-29 2003-05-01 Inhibitors of melanocyte tyrosinase as topical skin lighteners

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/206,334 Expired - Fee Related US7718185B2 (en) 2000-02-29 2008-09-08 Inhibitors of melanocyte tyrosinase as topical skin lighteners
US12/206,290 Expired - Fee Related US7858105B2 (en) 2000-02-29 2008-09-08 Inhibitors of melanocyte tyrosinase as topical skin lighteners

Country Status (8)

Country Link
US (5) US20020044914A1 (en)
EP (2) EP1267868B1 (en)
AT (2) ATE481967T1 (en)
AU (1) AU2001243334A1 (en)
CA (1) CA2401336C (en)
DE (2) DE60143135D1 (en)
ES (1) ES2352974T3 (en)
WO (1) WO2001064206A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060160880A1 (en) * 2003-12-23 2006-07-20 Jan Kehler 2-(1H-indolylsulfanyl)-benzyl amine derivatives as SSRI
US20060287386A1 (en) * 2005-06-17 2006-12-21 Jan Kehler Benzo[b]furane and benzo[b]thiophene derivatives
US20060287382A1 (en) * 2005-06-17 2006-12-21 Jan Kehler 2-(1H-indolylsulfanyl)-aryl amine derivatives
WO2007078034A1 (en) * 2006-01-06 2007-07-12 Amorepacific Corporation A composition for inhibiting the c-kit portein containing benzimidazole amine derivatives or aminoquinoline derivatives
US20080027074A1 (en) * 2004-07-16 2008-01-31 Jan Kehler 2-(1H-Indolysulfanyl)-Aryl Amine Derivatives for Use in the Treatment of Affective Disorders, Pain, Adhd and Stress Urinary Incontinence
US20080070982A1 (en) * 2006-09-15 2008-03-20 Graham Timmins System and methods of melanoma prevention
US8293218B2 (en) 2010-07-29 2012-10-23 Conopco, Inc. Skin care compositions comprising substituted monoamines
US8476251B2 (en) * 2010-07-29 2013-07-02 Conopco, Inc. Skin care compositions comprising substituted diamines

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030199558A1 (en) * 2001-12-28 2003-10-23 Integriderm, Inc. Hydroxamic acid and its derivatives as inhibitors of melanocyte tyrosinase for topical skin lighteners
FR2845599B1 (en) * 2002-10-11 2005-01-07 Lmd MEDICAMENT COMPRISING A THIOUREE FOR ITS USE AS A DEPIGMENTING
DE10252772A1 (en) * 2002-11-13 2004-05-27 Beiersdorf Ag Cosmetic or dermatological composition used for treating e.g. against skin sensitivity, inflammation, psoriasis, pruritis and dandruff contains diethyl dithiocarbamate
PL1748767T3 (en) 2004-05-28 2012-08-31 Unigen Inc 1-(3-methyl-2,4-dimethoxyphenyl)-3-(2',4'-dihydroxyphenyl)-propane as a potent tyrosinase inhibitor
FR2877672B1 (en) * 2004-11-10 2007-04-13 Oreal USE OF UTP TO PROMOTE IN VITRO CULTIVATION OF MELANOCYTES, PARTICULARLY HIGHLY PIGMENTED MELANOCYTES
FR2880022B1 (en) * 2004-12-24 2007-08-24 Mayoly Spindler Soc Par Action NOVEL DERIVATIVES OF N-HYDROXY-N'-PHENYLUREE AND N-HYDROXY-N'-PHENYLTHIOUREE AND THEIR USE AS INHIBITORS OF MELANIN SYNTHESIS
DE102005022292A1 (en) * 2005-05-13 2006-11-16 Basf Ag Use of sulfur containing compound, as a peroxide composite, in cosmetic and pharmaceutical agent to treat skin
FR2890070A1 (en) * 2005-08-31 2007-03-02 Galderma Res & Dev New imidazole compounds are tyrosinase inhibitors useful to treat e.g. melasma, chloasma, lentigines, senile lentigo, and vitiligo, and to prevent and/or treat the signs of ageing
WO2007039821A2 (en) * 2005-08-31 2007-04-12 Galderma Research & Development Novel inhibitors of tyrosinase, their method of preparation and their use in human medicine and in cosmetics
KR102397221B1 (en) * 2008-07-21 2022-05-12 유니젠, 인크. Series of skin-whitening (lightening) compounds
EP2193779A1 (en) * 2008-12-05 2010-06-09 Cognis IP Management GmbH Skin whitener
CN102378622A (en) 2009-03-31 2012-03-14 株式会社资生堂 Skin-whitening agent and cosmetic method for whitening skin
KR101246682B1 (en) * 2010-02-23 2013-04-01 충북대학교 산학협력단 Composition for whitening of the skin comprising arylthiourea derivatives
FR2958845B1 (en) * 2010-04-16 2012-06-01 Clc Technologie NEW DEPIGMENTING COMPOSITIONS
US20110269852A1 (en) * 2010-04-30 2011-11-03 Hampton University Methods and mechanisms involving hyperpigmentation particularly for african american skin
KR102113823B1 (en) 2011-03-24 2020-05-21 유니젠, 인크. Compounds and methods for preparation of diarylpropanes
US20130273183A1 (en) * 2012-04-13 2013-10-17 Eastman Chemical Company 2-Pyrimidine Thioesters and Thiocarbonates as Skin Brightening Agents
US20150174034A1 (en) 2013-03-13 2015-06-25 Avon Products, Inc. Tyrosinase inhibitors
WO2014164195A1 (en) * 2013-03-13 2014-10-09 Avon Products, Inc Tyrosinase inhibitors
WO2014186344A2 (en) 2013-05-13 2014-11-20 New York University Methods and agents for treating tyrosinase-positive albinism
JP6946412B2 (en) 2016-07-18 2021-10-06 ヤンセン ファーマシューティカ エヌ.ベー. Tau PET Imaging Ligand
MX2019002572A (en) 2016-09-06 2019-06-06 Unilever Nv Compounds for reducing cellular melanin content.
CN109180587A (en) * 2018-09-20 2019-01-11 五邑大学 A kind of new application of pyrazole analogs
WO2022064083A1 (en) 2021-01-11 2022-03-31 Mesoestetic Pharma Group, S.L Depigmenting dermopharmaceutical or cosmetic composition and use thereof
KR102543627B1 (en) * 2021-01-22 2023-06-20 한국과학기술연구원 Skin whitening composition containing 2,4-Dimethylphenylthiourea
CN116963710A (en) 2021-02-12 2023-10-27 西姆莱斯股份公司 Medicine for preventing and treating hyperpigmentation
WO2022129651A1 (en) 2021-04-01 2022-06-23 Mesoestetic Pharma Group, S.L Tyrosinase-inhibiting molecules and dermopharmaceutical composition that includes them

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6514486B1 (en) * 1997-11-04 2003-02-04 L'oreal Composition for topical application comprising at least one iminophenol compound

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT1205114B (en) * 1987-05-29 1989-03-15 Boehringer Biochemia Srl 2-SELENOMETHYL-1,4-DIHYDROPYRIDINE, PROCEDURE FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
JPH05124922A (en) * 1991-04-09 1993-05-21 Sansho Seiyaku Co Ltd External preparation with melanin production-inhibitory activity
JPH05124925A (en) * 1991-04-09 1993-05-21 Sansho Seiyaku Co Ltd External preparation with melanin production-inhibitory activity
WO1994002456A1 (en) * 1992-07-15 1994-02-03 The Governors Of The University Of Alberta Radioimaging and radiochemotherapy phenolic thioether amines and acyl derivatives thereof for use in diagnosing and treating pigmentation disorders
JP3545097B2 (en) * 1995-04-19 2004-07-21 御木本製薬株式会社 Skin cosmetics
JPH09323955A (en) * 1996-06-03 1997-12-16 Kao Corp Benzoic derivative and medicine containing the same
US6337320B1 (en) * 1996-10-11 2002-01-08 Thione International, Inc. Reparatives for ultraviolet radiation skin damage
JPH1121225A (en) * 1997-06-27 1999-01-26 Tanabe Seiyaku Co Ltd Ingredient for beautifying and whitening and skin preparation for external use for beautifying and whitening containing the same ingredient

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6514486B1 (en) * 1997-11-04 2003-02-04 L'oreal Composition for topical application comprising at least one iminophenol compound

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080214645A1 (en) * 2003-12-23 2008-09-04 H. Lundbeck A/S Uses of 2-(1h-indolylsulfanyl)-benzyl amine derivatives as ssris
US7737171B2 (en) 2003-12-23 2010-06-15 H. Lundbeck A/S Uses of 2-(1H-indolylsulfanyl)-benzyl amine derivatives as SSRIS
US7737170B2 (en) 2003-12-23 2010-06-15 H. Lundbeck A/S Uses of 2-(1H-indolylsulfanyl)-benzyl amine derivatives as SSRIS
US20060160880A1 (en) * 2003-12-23 2006-07-20 Jan Kehler 2-(1H-indolylsulfanyl)-benzyl amine derivatives as SSRI
US7652150B2 (en) 2003-12-23 2010-01-26 H. Lundbeck A/S 2-(1H-indolylsulfanyl)-benzyl amine derivatives as SSRIs
US7563908B2 (en) 2003-12-23 2009-07-21 Jan Kehler 2-(1H-indolylsulfanyl)-benzyl amine derivatives as SSRI
US20080214644A1 (en) * 2003-12-23 2008-09-04 H. Lundbeck A/S 2-(1h-indolylsulfanyl)-benzyl amine derivatives as ssri
US20080176922A1 (en) * 2003-12-23 2008-07-24 H. Lundbeck A/S Uses of 2-(1h-indolylsulfanyl)-benzyl amine derivatives as ssris
US7678800B2 (en) 2004-07-16 2010-03-16 H. Lundbeck A/S 2-(1H-indolylsulfanyl)-aryl amine derivatives for use in the treatment of affective disorders, pain, ADHD and stress urinary incontinence
US20080027074A1 (en) * 2004-07-16 2008-01-31 Jan Kehler 2-(1H-Indolysulfanyl)-Aryl Amine Derivatives for Use in the Treatment of Affective Disorders, Pain, Adhd and Stress Urinary Incontinence
US7534791B2 (en) 2005-06-17 2009-05-19 H. Lundbeck A/S Benzo[b]furane and benzo[b]thiophene derivatives
US20090192213A1 (en) * 2005-06-17 2009-07-30 H. Lundbeck A/S Benzo[b]furane And Benzo[b]thiophene Derivatives
US7629473B2 (en) 2005-06-17 2009-12-08 H. Lundbeck A/S 2-(1H-indolylsulfanyl)-aryl amine derivatives
US20060287382A1 (en) * 2005-06-17 2006-12-21 Jan Kehler 2-(1H-indolylsulfanyl)-aryl amine derivatives
US20060287386A1 (en) * 2005-06-17 2006-12-21 Jan Kehler Benzo[b]furane and benzo[b]thiophene derivatives
KR100788161B1 (en) 2006-01-06 2007-12-21 (주)아모레퍼시픽 A composition for skin whitening containing benzimidazole amine derivates or aminoquinoline derivatives
WO2007078034A1 (en) * 2006-01-06 2007-07-12 Amorepacific Corporation A composition for inhibiting the c-kit portein containing benzimidazole amine derivatives or aminoquinoline derivatives
US20080070982A1 (en) * 2006-09-15 2008-03-20 Graham Timmins System and methods of melanoma prevention
US20110021623A1 (en) * 2006-09-15 2011-01-27 Regents Of The University Of New Mexico System and methods of melanoma prevention
US8293218B2 (en) 2010-07-29 2012-10-23 Conopco, Inc. Skin care compositions comprising substituted monoamines
US8476251B2 (en) * 2010-07-29 2013-07-02 Conopco, Inc. Skin care compositions comprising substituted diamines

Also Published As

Publication number Publication date
AU2001243334A1 (en) 2001-09-12
DE60143135D1 (en) 2010-11-04
EP1267868B1 (en) 2007-07-18
US20030219393A1 (en) 2003-11-27
CA2401336A1 (en) 2001-09-07
US7858105B2 (en) 2010-12-28
EP1267868A2 (en) 2003-01-02
DE60129420T2 (en) 2008-04-03
CA2401336C (en) 2014-12-23
ATE367157T1 (en) 2007-08-15
DE60129420D1 (en) 2007-08-30
US20020044914A1 (en) 2002-04-18
EP1857109A2 (en) 2007-11-21
US20090023793A1 (en) 2009-01-22
US7718185B2 (en) 2010-05-18
ES2352974T3 (en) 2011-02-24
EP1857109B1 (en) 2010-09-22
WO2001064206A3 (en) 2002-08-29
WO2001064206A2 (en) 2001-09-07
US20090023792A1 (en) 2009-01-22
EP1857109A3 (en) 2008-03-19
ATE481967T1 (en) 2010-10-15

Similar Documents

Publication Publication Date Title
US7858105B2 (en) Inhibitors of melanocyte tyrosinase as topical skin lighteners
Penney et al. Depigmenting action of hydroquinone depends on disruption of fundamental cell processes
US8329149B2 (en) Topical lightening composition and uses thereof
KR100788161B1 (en) A composition for skin whitening containing benzimidazole amine derivates or aminoquinoline derivatives
CZ297669B6 (en) Topical pharmaceutical composition for lightening skin, tyrosinase inhibiting topical composition and topical or transdermal composition for treating inflammatory diseases containing resorcinol derivative and corresponding medicaments
US20030199558A1 (en) Hydroxamic acid and its derivatives as inhibitors of melanocyte tyrosinase for topical skin lighteners
US20150010484A1 (en) Method for anti-skin aging using caffeamide derivative
KR100538417B1 (en) Assay for identification of compounds that promote melanin production and retinoid-like compounds identified by said assay
KR20110097576A (en) Composition for whitening of the skin comprising benzaldehyde thiosemicarbazone derivatives
KR100956698B1 (en) Composition for skin whitening containing N-acetylphytosphingosine
US20220249404A1 (en) Composition containing fluoxetine and vitamin d3 or its derivatives, and application thereof
KR100747042B1 (en) Novel compound of 6-methyl-3-phenethyl-3,4-dihydro-1H-quinazoline-2-thione, its preparation and a depigmentation composition containing the compound as an effective component
US20190388321A1 (en) Methods of treating hyperpigmentation disorders
AU2012243226A1 (en) Use of N-(4-methoxyphenyl)-1-phenyl-1H-pyrazol-3-amine and related compounds
KR100629065B1 (en) Cosmetic compositions
KR100752757B1 (en) Whitening composition containing 2-phenylimino-1,3-thiazoline
KR101628132B1 (en) Novel biaryl amide derivatives, and composition comprising the same as an active ingredient
US20160009755A1 (en) Cyrrhetinic alkyl esters and protected derivatives thereof
KR20050066660A (en) Sphingolipid-rucinol derivatives and composition for skin external use containing the derivatives
EA045810B1 (en) SKIN CARE COMPOSITIONS AND THEIR APPLICATIONS
JPH05222030A (en) Sulfur atom-containing compound

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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