Cosmetic composition

文档序号:1806013 发布日期:2021-11-09 浏览:10次 中文

阅读说明:本技术 化妆品组合物 (Cosmetic composition ) 是由 蒂凡尼·卡尔 吉赛·卡拉哈斯蒂 利沙·万佩尔特 大卫·甘 于 2021-05-06 设计创作,主要内容包括:本发明一般涉及使用方法和组合物,其可用于破坏死皮细胞和新的、健康的细胞之间的连接,以刺激表皮剥落、增强皮肤屏障功能、改善皮肤光彩、改善皮肤纹理外观、加快皮肤更新、加快表皮剥落、提高皮肤平滑度、增加水分含量、吸引水、使表皮剥落、减少或消除表皮剥落造成的刺激、更新皮肤、增加皮肤光彩、使皮肤柔软、增加皮肤平滑度、使皮肤水合、使皮肤平滑、使皮肤变亮、减少皮肤老化的迹象和/或增加化妆品组合物表皮剥落、减少或消除表皮剥落造成的刺激、更新皮肤、增加皮肤光彩、使皮肤柔软、增加皮肤平滑度、使皮肤水合、使皮肤平滑、使皮肤变亮和/或减少皮肤老化的迹象的功效。组合物包含乙醇酸和葡糖酸内酯的组合。(The present invention relates generally to methods and compositions of use, which can be used to disrupt the connection between dead skin cells and new, healthy cells, to stimulate exfoliation, enhance skin barrier function, improve skin radiance, improve skin texture appearance, accelerate skin rejuvenation, accelerate exfoliation, improve skin smoothness, increase moisture content, attract water, exfoliate, reduce or eliminate irritation from exfoliation, rejuvenate skin, increase skin radiance, soften skin, increase skin smoothness, hydrate skin, smooth skin, lighten skin, reduce signs of skin aging, and/or increase exfoliation of a cosmetic composition, reduce or eliminate irritation from exfoliation, rejuvenate skin, increase skin radiance, soften skin, increase skin smoothness, hydrate skin, smooth skin, lighten skin, and/or reduce signs of skin aging. The composition comprises a combination of glycolic acid and gluconolactone.)

1. Use of a composition comprising effective amounts of glycolic acid and gluconolactone in the preparation of a formulation for stimulating exfoliation, removing dead skin cells, increasing cell renewal, improving skin radiance, improving skin texture, and/or accelerating skin renewal in a human, wherein the composition is topically applied to the skin of the human, wherein topical application of the composition stimulates exfoliation, removes dead skin cells, increases cell renewal, improves skin radiance, improves skin texture, and/or accelerates skin renewal.

2. Use according to claim 1, wherein the composition comprises from 0.1 to 15% by weight of glycolic acid and from 0.1 to 10% by weight of gluconolactone.

3. The use of claim 1, wherein at least a second skin care composition is applied to the skin prior to applying the composition to the skin.

4. The use of claim 1, wherein the composition is combined with a third skin care composition prior to applying the composition to skin.

5. Use according to claim 4, wherein the third skin care composition has an effect on the smoothing effect of the skin.

6. Use according to claim 4, wherein the third skin care composition has no effect on the smoothing effect of the skin.

7. Use according to claim 1, wherein the composition further comprises an effective amount of one or more of water, glycerin, butylene glycol, potassium hydroxide and/or betaine to moisturize and/or enhance the smoothing effect of the skin product.

8. The use according to claim 7, wherein the composition further comprises:

1 to 95% by weight of water;

0.1 to 20% by weight of glycerol;

0.1 to 10 wt% of butanediol;

0.1 to 5% by weight of potassium hydroxide; and/or

0.01 to 3% by weight of betaine.

9. The use of claim 1, wherein the composition further comprises one or more than one of methyl gluceth-20, PEG-8 dimethicone, phenoxyethanol, hydroxyethyl cellulose, and/or caprylyl glycol.

10. The use according to claim 9, wherein the composition further comprises:

0.01 to 5% by weight of methyl glucitol polyether-20;

0.01 to 5% by weight of PEG-8 polydimethylsiloxane;

0.01 to 1% by weight of phenoxyethanol;

0.01 to 1% by weight of hydroxyethyl cellulose; and/or

0.01 to 1% by weight of caprylyl glycol.

11. The use according to claim 1, wherein the composition further comprises one or more than one of a moisturizer, an emollient, a skin conditioner, and/or a pH adjuster.

12. Use according to claim 1, wherein the composition comprises from 1% to 10% by weight of gluconolactone.

13. Use according to claim 12, wherein the composition comprises from 3 to 7% by weight of gluconolactone.

14. The use of claim 1, wherein the composition comprises from 1% to 7% by weight of glycolic acid.

15. The use according to claim 14, wherein the composition comprises from 2 to 5% by weight of glycolic acid.

16. Use according to claim 1, wherein the composition further comprises from 40 to 85% by weight of water, from 2 to 15% by weight of glycerol and/or opuntia tuna fruit extract.

17. A method of enhancing the activity of a skin care composition, the method comprising combining an enhancing composition with a skin care composition, wherein the enhancing composition comprises glycolic acid and gluconolactone in an amount effective to increase or enhance the ability of the skin care composition to stimulate epidermal exfoliation, remove dead skin cells, increase cell renewal, improve skin radiance, improve skin texture, and/or accelerate skin renewal.

18. The method of claim 17, wherein the skin care composition has an effect on the smoothing effect of the skin.

19. The method of claim 17, wherein the skin care composition has no effect on the smoothing effect of the skin.

20. A product enhancing composition comprising a combination of glycolic acid and gluconolactone in an effective amount to stimulate exfoliation, remove dead skin cells, increase cell renewal, improve skin radiance, improve skin texture, and/or accelerate skin turnover.

Technical Field

The present invention relates generally to cosmetic compositions and methods useful for stimulating the efficacy of skin exfoliation, removing dead skin cells that make the skin appear rough and dull, increasing cell renewal, improving skin radiance, improving skin texture, accelerating skin renewal, and/or increasing cosmetic stimulation of skin exfoliation, removing dead skin cells that make the skin appear rough and dull, increasing cell renewal, improving skin radiance, improving skin texture, accelerating skin renewal. In particular, the composition may comprise glycolic acid and/or gluconolactone.

Background

Various factors can cause different stresses on the skin, including, for example, aging, prolonged exposure to adverse environmental factors, malnutrition, fatigue, stress, seasonal changes, and other extrinsic and intrinsic factors that may damage the skin. These stresses can alter the visual appearance, physical properties, or physiological function of skin and tissue in ways that are considered visually undesirable. Significant and obvious changes include dry skin, rough surface texture, the appearance of fine lines and wrinkles, loss of elasticity, decreased skin barrier function, loss of skin tone uniformity or skin tone, and mottled pigmentation. Many of these pressures are difficult or unavoidable.

Less obvious but measurable changes that occur as skin and tissue age or experience long-term environmental damage include a general decrease in cellular and tissue activity, a decrease in the rate of cellular replication, reduced skin blood flow, reduced moisture content, cumulative errors in structure and function, changes in the normal regulation of common biochemical processes, and a decrease in the ability of skin and tissue to remodel and repair itself. Many changes in the appearance and function of the skin are caused by changes in the outer epidermal layer of the skin, while other changes are caused by changes in the underlying dermis. Regardless of the stimulus that causes the skin injury, when the injury occurs, many natural and complex biochemical mechanisms come into play in an attempt to repair the injury.

Skin cell renewal is natural and necessary to bring fresh new cells to the skin surface and replace dead cells that make the skin look rough and dull and make the skin feel rough. With aging, the process of skin cell renewal slows, and the skin can look and feel rougher, dry and dull. Exfoliation and moisturization can aid in skin cell renewal. Maintaining skin moisture may also help overcome some unwanted changes in the skin. However, maintaining skin moisture can be difficult. This is especially true for subjects with drier skin than average (dry skin type). Exposure to chemicals, solvents, lotions, cosmetics, textiles, or dry environments is some of the many ways skin may lose moisture.

Others have attempted to make compositions and methods for exfoliating and/or rejuvenating skin. However, many attempts have been ineffective, addressing only one or a few undesirable effects, or causing unacceptable side effects, such as skin irritation, by themselves. Thus, there is a need for new products that are effective in exfoliating skin and/or rejuvenating skin without causing skin irritation.

Disclosure of Invention

The inventors have identified a solution to the problems associated with current cosmetics. The solution is based on a combination of ingredients comprising glycolic acid and gluconolactone. The combination can be used for stimulating exfoliation of skin, removing dead skin cells that make skin appear rough and dull, increasing cell renewal, improving skin radiance, improving skin texture, increasing skin turnover and/or enhancing the effectiveness of other cosmetic products to stimulate exfoliation of skin, removing dead skin cells that make skin appear rough and dull, increasing cell renewal, improving skin radiance, improving skin texture, increasing skin turnover. Additional benefits may include reduction or alleviation of unwanted side effects. In some aspects, an effective amount of a combination of glycolic acid and gluconolactone is combined with an effective amount of glycerin to increase skin moisture content and/or further improve exfoliation.

In some aspects, topical compositions comprising glycolic acid and gluconolactone are disclosed. In some aspects, topical compositions comprising any one, any combination, or all of glycolic acid, gluconolactone, and/or glycerin are disclosed. The amount of ingredients within the composition can vary (e.g., the amount can be as low as 0.000001 wt% to as high as 99 wt%, or any range therebetween). In some aspects, the topical composition comprises 0.1% to 15% by weight gluconolactone and 0.1% to 15% by weight glycolic acid. In some aspects, the topical composition comprises 0.1% to 10% by weight gluconolactone and 0.1% to 10% by weight glycolic acid. In some aspects, the topical composition comprises 0.1% to 10% by weight gluconolactone, 0.1% to 10% by weight glycolic acid, and 0.1% to 10% by weight glycerin.

In some aspects, the compositions comprise effective amounts of glycolic acid and gluconolactone, wherein topical application of the composition stimulates exfoliation, removes dead skin cells, increases cell renewal, improves skin radiance, improves skin texture, and/or accelerates skin turnover. In some cases, the composition comprises 0.1% to 15% by weight glycolic acid and 0.1% to 10% by weight gluconolactone. In some cases, the second skin care composition is applied to the skin prior to applying the composition to the skin. In some cases, more than one skin care composition is applied to the skin prior to applying the composition to the skin. In some cases, the composition is combined with a third skin care composition prior to applying the composition to the skin. In some cases, the third skin care composition has an effect on the smoothing effect of the skin. In some cases, the third skin care composition has no effect on the smoothing effect of the skin.

In some cases, the composition further comprises an effective amount of one or more of water, glycerin, butylene glycol, potassium hydroxide, and/or betaine to moisturize and/or enhance the smoothing action of the skin product. In some cases, the composition comprises one or more of 1 to 95 wt% water, 0.1 to 20 wt% glycerin, 0.1 to 10 wt% butanediol, 0.1 to 5 wt% potassium hydroxide, and/or 0.01 to 3 wt% betaine.

In some cases, the composition further comprises one or more than one of methyl gluceth-20, PEG-8 dimethicone, phenoxyethanol, hydroxyethyl cellulose, and/or caprylyl glycol. In some cases, the composition comprises one or more of 0.01 to 5 weight percent methyl gluceth-20, 0.01 to 5 weight percent PEG-8 polydimethylsiloxane, 0.01 to 1 weight percent phenoxyethanol, 0.01 to 1 weight percent hydroxyethyl cellulose, and/or 0.01 to 1 weight percent caprylyl glycol.

In some cases, the composition further comprises one or more than one of a moisturizer, an emollient, a skin conditioner, and/or a pH adjuster. In some instances, the composition comprises from 1% to 10% by weight of gluconolactone. In some instances, the composition comprises from 3% to 7% by weight of gluconolactone. In some instances, the composition comprises from 1% to 7% by weight of glycolic acid. In some instances, the composition comprises from 2% to 5% by weight of glycolic acid.

In some instances, the composition also includes from 40% to 85% by weight water. In some instances, the composition also comprises from 2% to 15% by weight of glycerin. In some instances, the composition also includes Opuntia ficus-indica (Opuntia tuna) extract. In some instances, the composition comprises 0.001% to 2% by weight of opuntia ficus-indica (opuntia ficus-indica) extract. In some instances, the composition is an enhancing composition capable of enhancing the activity of a skin care composition by combining the enhancing composition with the skin care composition, wherein the enhancing composition comprises an effective amount of glycolic acid and gluconolactone to enhance or enhance the ability of the cosmetic composition to stimulate epidermal exfoliation, remove dead skin cells, increase cell renewal, improve skin radiance, improve skin texture, and/or accelerate skin renewal. In some cases, the skin care composition has an effect on the smoothing effect of the skin. In some cases, the skin care composition has no effect on the smoothing effect of the skin.

In some cases, the composition is a product enhancing composition comprising a combination of glycolic acid and gluconolactone in an effective amount to stimulate exfoliation, remove dead skin cells, increase cell renewal, improve skin radiance, improve skin texture, and/or accelerate skin renewal.

In some aspects, the composition is applied to the skin multiple times per week. In some cases, the composition may be applied to the skin 2 to 3 times per week. In some cases, the composition may be applied to the skin 2 times per week. In some cases, the composition may be applied to the skin 3 times per week. In some cases, the composition may be applied to the skin more than 3 times per week. In some aspects, the composition may be used in combination with a second composition for treating skin. In some aspects, the second composition for treating skin does not comprise retinol. In some aspects, the second composition for treating skin is not an exfoliating product. In some aspects, the second composition for treating skin is a product enhancing composition. In some cases, the product enhancing composition comprises ceramide, hyaluronic acid, and/or Verbena (Verbena officinalis) extract.

In some aspects, the composition is applied to clean skin. In some cases, the composition is left on the skin to be absorbed. In some cases, the composition is applied followed by the serum or moisturizer. In some cases, the serum or moisturizer is applied after the composition is absorbed by the skin.

In some aspects, the compositions of the present invention may further comprise surfactants, silicone-containing compounds, UV agents, oils, and/or other ingredients identified herein or known in the art. The composition can be a lotion, cream, body lotion, mask, scrub, lotion, gel, essence, emulsion (e.g., oil-in-water, water-in-oil, silicone-in-water, water-in-oil-in-water, oil-in-water-in-oil, oil-in-water-in-silicone, etc.), solution (e.g., aqueous or hydroalcoholic solution), anhydrous base (e.g., lipstick or powder), ointment, emulsion, paste, aerosol, solid form, eye gel, gel essence, gel emulsion, etc. In some cases, the composition is a serum, cream, gel, cream gel, oil-in-water emulsion, water-in-oil emulsion, or liquid. In some cases, the composition is a liquid. In some cases, the composition is contained in an ampoule. The composition may be formulated for topical skin application at least 1,2, 3, 4, 5,6, 7, or more than 7 times per day during use. In some aspects of the invention, the composition may be storage stable or color stable, or both. It is also contemplated that the viscosity of the composition can be selected to achieve a desired result, for example, the viscosity of such composition can be from about 1cp to well over 1 million cp, or any range or integer available therein (e.g., 2cp, 3cp, 4cp, 5cp, 6cp, 7cp, 8cp, 9cp, 10cp, 20cp, 30cp, 40cp, 50cp, 60cp, 70cp, 80cp, 90cp, 100cp, 200cp, 300cp, 400cp, 500cp, 600cp, 700cp, 800cp, 900cp, 1000cp, 2000cp, 3000cp, 4000cp, 5000cp, 6000cp, 7000cp, 8000cp, 9000cp, 10000cp, 20000cp, 30000cp, 40000cp, 50000cp, 60000cp, 70000cp, 80000cp, 90000cp, 30000cp, 400000cp, 20000000 cp, 800000cp, 80cp, measured at 2.5rpm on a Brookfield viscometer at 25 ℃.(s), 2000000cp, 3000000cp, 4000000cp, 5000000cp, 10000000cp, etc.).

In a non-limiting aspect, the pH of the composition can be from about 6 to about 9. In some aspects, the pH may be 1,2, 3, 4, 5,6, 7, 8, 9, 10, 11, 12, 13, or 14. The composition may comprise triglycerides. Non-limiting examples include short, medium and long chain triglycerides. In certain aspects, the triglyceride is a medium chain triglyceride (e.g., caprylic capric triglyceride). The composition may also comprise a preservative. Non-limiting examples of preservatives include phenoxyethanol, methyl paraben, propyl paraben, iodopropynyl butylcarbamate, potassium sorbate, sodium benzoate, or any mixture thereof. In some embodiments, the composition is free of parabens.

The compositions of the present invention may have UVA and UVB absorption properties. The composition may have a Sun Protection Factor (SPF) of 2, 3, 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, or greater than 60, or any integer or derivative thereof. The composition may be a sunscreen lotion, sunscreen spray or sunscreen cream.

The compositions of the present invention may further comprise any one, any combination, or all of the following additional ingredients: conditioning agent, humectant, pH adjuster, structuring agent, inorganic salt, preservative, thickener, silicone-containing compound, essential oil, fragrance, vitamin, pharmaceutical ingredient or antioxidant, or any combination of these ingredients or mixture of these ingredients. In certain aspects, the composition may comprise at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or more than ten, or all of these additional ingredients identified in the previous sentence. Non-limiting examples of these additional ingredients are identified throughout this specification and are incorporated in this section by reference. As disclosed elsewhere in this specification, the amount of such ingredients may be from 0.0001% to 99.9%, or any integer or range therebetween, by weight or volume of the composition, which is incorporated by reference in this paragraph.

Methods of using the compositions disclosed herein are also disclosed. In some aspects, methods of stimulating exfoliation of skin, removing dead skin cells that make skin appear rough and dull, increasing cell renewal, improving skin radiance, improving skin texture, accelerating skin renewal, and/or enhancing the efficacy of cosmetics stimulating exfoliation of skin, removing dead skin cells that make skin appear rough and dull, increasing cell renewal, improving skin radiance, improving skin texture, accelerating skin renewal are disclosed. In some cases, the method comprises topically applying any of the compositions disclosed herein to skin in need thereof. In one aspect, any of the compositions disclosed herein are topically applied and the composition is left on the area of application, removed from the area of application after a period of time, and/or removed directly after application.

In some aspects, the compositions disclosed herein are used to improve natural skin cell turnover, which can replace dead skin cells with fresh new cells and improve skin texture and radiance. In some aspects, the compositions disclosed herein can be used to exfoliate skin with reduced irritation as compared to other exfoliating products. In some aspects, the compositions disclosed herein are used to increase the moisture content of the upper layers of skin.

It is also contemplated that the compositions disclosed throughout this specification may be used as leave-on or rinse-off compositions. For example, the leave-on composition can be a composition that is topically applied to the skin and left on the skin for a period of time (e.g., at least 5 minutes, at least 6 minutes, at least 7 minutes, at least 8 minutes, at least 9 minutes, at least 10 minutes, at least 20 minutes, or at least 30 minutes, or at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 5 hours, at least 6 hours, at least 7 hours, at least 8 hours, at least 9 hours, at least 10 hours, at least 11 hours, at least 12 hours, at least 13 hours, at least 14 hours, at least 15 hours, at least 16 hours, at least 17 hours, at least 18 hours, at least 19 hours, at least 20 hours, at least 21 hours, at least 22 hours, at least 23 hours, or at least 24 hours, or overnight or all day). Alternatively, the rinse-off composition may be a product that is to be applied to the skin and then removed or washed off the skin (e.g., with water) over a period of time, such as less than 5 minutes, less than 4 minutes, less than 3 minutes, less than 2 minutes, or less than 1 minute. In some cases, the composition is designed to be washed off after 30 seconds, 1 minute, 2 minutes, 3 minutes, 4 minutes, 5 minutes, 6 minutes, 7 minutes, 8 minutes, 9 minutes, 10 minutes, 11 minutes, 12 minutes, 13 minutes, 14 minutes, 15 minutes, 20 minutes, 30 minutes, 40 minutes, 50 minutes, 60 minutes, or any number or range therebetween. Examples of rinse-off compositions may be facial cleansers, shampoos, conditioners or soaps. Examples of leave-on compositions may be skin moisturizers, sunscreens, masks, night creams or day creams.

Kits comprising the compositions of the invention are also contemplated. In certain embodiments, the composition is contained in a container. The container may be a bottle, a dispenser or a package. The container may dispense a predetermined amount of the composition. In certain aspects, the composition is dispensed in the form of a spray, mist, bolus, or liquid. The container may contain indicia on its surface. The indicia may be words, abbreviations, pictures or symbols.

It is contemplated that any embodiment discussed in this specification can be implemented for any method or composition of the invention and vice versa. Furthermore, the compositions of the present invention may be used to carry out the methods of the present invention.

In some embodiments, the compositions of the present invention may be pharmaceutically or cosmetically acceptable, or may have pleasant tactile properties. "pharmaceutically acceptable", "cosmetically acceptable", and/or "pleasant tactile properties" describe compositions having specific tactile properties that are pleasant to the skin (e.g., compositions that are not too water or too oil, compositions that have a silky texture, non-tacky or sticky compositions, etc.). Pharmaceutically acceptable or cosmetically acceptable may also relate to the creaminess or lubricity of the composition, or the moisture retention properties of the composition.

Products comprising the compositions of the present invention are also contemplated. In a non-limiting aspect, the product can be a cosmetic product. The cosmetic products may be those described elsewhere in this specification or known to those skilled in the art. Non-limiting examples of products include moisturizers, creams, lotions, emollients, essences, gels, lotions, body milks, scrubs, foundations, night creams, lipsticks, facial cleansers, toners, sunscreens, masks, anti-aging products, deodorants, antiperspirants, perfumes, colognes, and the like.

In the context of the present invention, at least the following 39 aspects are described. Aspect 1 includes methods of stimulating exfoliation, removing dead skin cells, increasing cell renewal, improving skin radiance, improving skin texture, and/or accelerating skin renewal in a human. The method comprises topically applying to the skin of a human an effective amount of glycolic acid and gluconolactone, wherein topical application of the composition stimulates exfoliation of the epidermis, removes dead skin cells, increases cell renewal, improves skin radiance, improves skin texture, and/or accelerates skin turnover. Aspect 2 depends from aspect 1, wherein the composition comprises from 0.1% to 15% by weight of glycolic acid and from 0.1% to 10% by weight of gluconolactone. Aspect 3 pertains to any one of aspects 1 and 2, wherein the second skin care composition is applied to the skin prior to applying the composition to the skin. Aspect 4 pertains to any one of aspects 1 to 3, wherein more than one skin care composition is applied to the skin prior to applying the composition to the skin. Aspect 5 pertains to any one of aspects 1 to 4, wherein the composition is combined with a third skin care composition prior to application to the skin. Aspect 6 pertains to aspect 5, wherein the third skin care composition has an effect on the smoothing effect of the skin. Aspect 7 pertains to aspect 5, wherein the third skin care composition has no effect on the smoothing effect of the skin. Aspect 8 pertains to any of aspects 1 to 7, wherein the composition further comprises an effective amount of one or more of water, glycerin, butylene glycol, potassium hydroxide, and/or betaine to moisturize and/or enhance the smoothing action of the skin product. Aspect 9 depends from aspect 8, wherein the composition further comprises 1 to 95 wt% water, 0.1 to 20 wt% glycerin, 0.1 to 10 wt% butanediol, 0.1 to 5 wt% potassium hydroxide, and/or 0.01 to 3 wt% betaine. Aspect 10 pertains to any one of aspects 1 to 9, wherein the composition further comprises one or more of methyl gluceth-20, PEG-8 polydimethylsiloxane, phenoxyethanol, hydroxyethyl cellulose, and/or caprylyl glycol. Aspect 11 depends from aspect 10, wherein the composition further comprises 0.01 to 5 wt.% of methyl gluceth-20, 0.01 to 5 wt.% of PEG-8 polydimethylsiloxane, 0.01 to 1 wt.% of phenoxyethanol, 0.01 to 1 wt.% of hydroxyethyl cellulose, and/or 0.01 to 1 wt.% of caprylyl glycol. Aspect 12 pertains to any one of aspects 1 to 11, wherein the composition further comprises one or more than one of a moisturizer, an emollient, a skin conditioner, and/or a pH adjuster. Aspect 13 pertains to any one of aspects 1 to 12, wherein the composition comprises 1% to 10% gluconolactone by weight. Aspect 14 depends from aspect 13, wherein the composition comprises 3% to 7% gluconolactone by weight. Aspect 15 pertains to any one of aspects 1 to 14, wherein the composition comprises 1 to 7 weight percent glycolic acid. Aspect 16 pertains to aspect 15, wherein the composition comprises 2% to 5% by weight of glycolic acid. Aspect 17 depends from any one of aspects 1 to 16, wherein the composition further comprises 40 wt% to 85 wt% water. Aspect 18 pertains to any one of aspects 1 to 17, wherein the composition further comprises 2 to 15% by weight of glycerin. Aspect 19 pertains to any one of aspects 1 to 18, wherein the composition further comprises opuntia ficus-indica (opuntia ficus-indica) extract. Aspect 20 includes a method of enhancing the activity of a skin care composition. The method comprises combining an enhancing composition with a skin care composition, wherein the enhancing composition comprises an effective amount of glycolic acid and gluconolactone to enhance or enhance the ability of the cosmetic composition to stimulate epidermal exfoliation, remove dead skin cells, increase cell renewal, improve skin radiance, improve skin texture, and/or accelerate skin renewal. Aspect 21 pertains to aspect 20, wherein the skin care composition has an effect on the smoothing effect of the skin. Aspect 22 pertains to aspect 20, wherein the skin care composition has no effect on the smoothing effect of the skin. Aspect 23 includes a product enhancing composition. The product enhancing composition comprises a combination of glycolic acid and gluconolactone in an amount effective to stimulate exfoliation, remove dead skin cells, increase cell renewal, improve skin radiance, improve skin texture, and/or accelerate skin turnover. Aspect 24 depends from aspect 23, wherein the product enhancing composition comprises from 0.1% to 15% by weight of glycolic acid and from 0.1% to 10% by weight of gluconolactone. Aspect 25 pertains to any one of aspects 23 to 24, further comprising an effective amount of one or more of water, glycerin, butylene glycol, potassium hydroxide, and/or betaine to moisturize and/or enhance the smoothing action of the skin care product. Aspect 26 pertains to aspect 25, further comprising 1 to 95 wt.% water, 0.1 to 20 wt.% glycerin, 0.1 to 10 wt.% butanediol, 0.1 to 5 wt.% potassium hydroxide, and/or 0.01 to 3 wt.% betaine. Aspect 27 pertains to any one of aspects 23 to 26, further comprising one or more of methyl gluceth-20, PEG-8 polydimethylsiloxane, phenoxyethanol, hydroxyethyl cellulose, and/or caprylyl glycol. Aspect 28 depends from aspect 27, further comprising 0.01 to 5 wt.% methyl gluceth-20, 0.01 to 5 wt.% PEG-8 polydimethylsiloxane, 0.01 to 1 wt.% phenoxyethanol, 0.01 to 1 wt.% hydroxyethyl cellulose, and/or 0.01 to 1 wt.% caprylyl glycol. Aspect 29 pertains to any one of aspects 23 to 28, further comprising one or more than one of a moisturizer, an emollient, a skin conditioner, and/or a pH adjuster. Aspect 30 depends from any one of aspects 23 to 29, wherein the product enhancing composition comprises 1% to 10% gluconolactone by weight. Aspect 31 depends from aspect 30, wherein the product enhancing composition comprises 3% to 7% by weight of gluconolactone. Aspect 32 pertains to any one of aspects 23 to 31, wherein the product enhancing composition comprises from 1% to 7% by weight of glycolic acid. Aspect 33 pertains to aspect 32, wherein the product enhancing composition comprises 2% to 5% by weight of glycolic acid. Aspect 34 pertains to any one of aspects 23 to 33, wherein the product enhancing composition comprises 40 wt.% to 85 wt.% water. Aspect 35 pertains to any one of aspects 23 to 34, wherein the product enhancing composition comprises 2% to 15% by weight of glycerin. Aspect 36 pertains to any one of aspects 23 to 35, wherein the product enhancing composition is a serum, cream, gel, cream gel, oil-in-water emulsion, water-in-oil emulsion, or liquid. Aspect 37 pertains to the product enhancing composition of aspect 36, wherein the product enhancing composition is a liquid. Aspect 38 pertains to any one of aspects 23 to 37, wherein the product enhancing composition is contained in an ampoule. Aspect 39 depends from any one of aspects 23 to 38, further comprising 0.001% to 2% by weight of opuntia ficus-indica (opuntia ficus-indica) extract.

By "topical application" is meant that the composition is applied or spread onto the surface of the lips or keratinous tissue. "topical skin compositions" include compositions suitable for topical application to the skin and/or keratinous tissue. Such compositions are generally dermatologically acceptable in that they do not have excessive toxicity, incompatibility, instability, allergic response, and the like when applied to the skin and/or keratinous tissue. The topical skin care compositions of the present invention may have a viscosity selected to avoid significant dripping or pooling after application to the skin and/or keratinous tissue.

"keratinous tissue" includes keratin-containing layers configured as the outermost protective layer of a mammal, and includes, but is not limited to, lips, skin, hair, and nails.

The term "about" or "approximately" is defined as being close as understood by one of ordinary skill in the art. In one non-limiting embodiment, the term is defined as within 10%, preferably within 5%, more preferably within 1%, and most preferably within 0.5%.

The term "substantially" and variations thereof refer to a range within 10%, within 5%, within 1%, or within 0.5%.

The terms "inhibit" or "reduce" or any variation of these terms includes any measurable reduction or complete inhibition to achieve a desired result. The term "promote" or "increase" or any variant of these terms includes any measurable increase, such as a measurable increase in a protein or molecule (e.g., a matrix protein such as fibronectin, laminin, collagen or elastin, or a molecule such as hyaluronic acid) in order to achieve a desired result.

As used in this specification and/or the claims, the term "effective" means sufficient to achieve a desired, expected, or intended result.

When used in the claims and/or the specification with the terms "comprising," including, "" containing, "or" having, "or any variation of these terms, the foregoing non-use of a quantitative term may mean" one, "but it is also consistent with the meaning of" one or more, "" at least one, "and" one or more than one.

As used in this specification and claims, the words "comprise," "have," "include," or "contain" are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.

The compositions and methods used may "comprise," consist essentially of, "or" consist of any of the ingredients or steps disclosed throughout this specification. With respect to the phrase "consisting essentially of … …," the basic and novel properties of the compositions and methods of the present invention are the ability to stimulate exfoliation of the skin, remove dead skin cells that make the skin look rough and dull, increase cell renewal, improve skin radiance, improve skin texture, accelerate skin renewal and/or enhance the efficacy of other cosmetics to stimulate exfoliation of the skin, remove dead skin cells that make the skin look rough and dull, increase cell renewal, improve skin radiance, improve skin texture, accelerate skin renewal.

Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the examples, while indicating specific embodiments of the invention, are given by way of illustration only. In addition, it is contemplated that variations and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.

Detailed Description

As described above, the present invention provides a solution to the problems associated with current cosmetics. In some embodiments, an effective amount of a composition comprising any one, any combination, or all of glycolic acid, gluconolactone, and/or glycerin is found to exfoliate skin, improve skin radiance, improve the appearance of skin texture, reduce skin roughness, enhance skin smoothness, and/or increase skin moisturization with less irritation than conventional methods. It has also been shown that the combination of ingredients breaks the link between dead skin cells and new, healthy cells to stimulate exfoliation, enhance skin barrier function, and attract water to raise skin moisture.

Gluconolactone has also been shown to disrupt the connection between dead skin cells and new, healthy cells to stimulate exfoliation, enhance skin barrier function, improve skin radiance, and improve the appearance of skin texture. Glycolic acid is shown to accelerate skin turnover, accelerate exfoliation, improve skin radiance and improve skin smoothness. Glycerol was shown to increase skin moisture and attract water.

Particular compositions of the present invention are designed for use as topical compositions. The composition relies on any one, any combination, or a unique combination of all of glycolic acid, gluconolactone, and/or glycerol. These compositions can be used to make topical compositions that disrupt the connection between dead skin cells and new, healthy cells, to stimulate exfoliation, enhance skin barrier function, improve skin radiance, improve skin texture appearance, accelerate skin rejuvenation, accelerate exfoliation, improve skin smoothness, increase moisture content, attract water, exfoliate, reduce or eliminate irritation from exfoliation, rejuvenate skin, increase skin radiance, soften skin, increase skin smoothness, hydrate skin, smooth skin, lighten skin, reduce signs of skin aging, and/or increase the effectiveness of a cosmetic composition to exfoliate skin, reduce or eliminate irritation from exfoliation, rejuvenate skin, increase skin radiance, soften skin, increase skin smoothness, hydrate skin, smooth skin, lighten skin, and/or reduce signs of skin aging. Non-limiting examples of such compositions are provided in table 1 of example 1 below.

Some compositions disclosed herein can be applied to the skin and left on the skin for a period of time (e.g., at least 1 minute, at least 2 minutes, at least 3 minutes, at least 4 minutes, at least 5 minutes, at least 10 minutes, at least 20 minutes, at least 30 minutes, or at least 60 minutes, or more than 60 minutes). Thereafter, the composition can be rinsed or peeled off from the skin, if desired. Some of the compositions disclosed herein can be applied to the skin and rinsed off the skin immediately. Some of the compositions disclosed herein may be applied to and at least partially absorbed by the skin. Some compositions are designed to remain on the skin.

These and other non-limiting aspects of the invention are described in the following sections.

A. Active ingredient

Gluconolactone is one of the compounds known as polyhydroxy acids. Gluconolactone is a crystalline powder prepared by removing water from gluconic acid. Gluconolactone may be prepared by the enzymatic oxidation of D-glucose oxidation. In some aspects, gluconolactone may be formed by crystallization of gluconic acid in a supersaturated aqueous solution, followed by dehydration of the crystals formed. Gluconolactone disrupts the connections between dead skin cells to stimulate exfoliation. Gluconolactone is a humectant that attracts and holds water. Gluconolactone helps to form a moisture barrier on skin tissue by preventing evaporation of water already present in the tissue, and may help to enhance the skin barrier function.

Glycolic acid is water soluble, one of the compounds known as alpha-hydroxy acids. Glycolic acid is a naturally occurring compound that can be derived from plants. In some aspects, glycolic acid is derived from sugar cane. Glycolic acid reacts with the upper layers of the skin, breaking it down by dissolving sebum and other substances that bind the cells together. After application of glycolic acid, dead skin cells can be removed to reveal smoother skin.

Glycerol is an alcohol present in animal, plant and human tissues. The glycerin can be prepared by heating vegetable oil (such as soybean oil, palm oil, coconut oil) or animal fat. Glycerin is a moisturizing agent that absorbs water and increases the moisture content of the skin. After application of glycerin, the skin may be more moist and/or become less irritated.

These combinations of ingredients can be used in different product forms to treat various skin conditions. As non-limiting examples, the combination of ingredients may be formulated as an ampoule, an emulsion (e.g., oil-in-water, water-in-oil), a gel, a serum, a gel emulsion, a gel serum, a lotion, a mask, a scrub, a lotion, a cream, or a body lotion.

The components described herein can be extracts prepared by extraction methods known in the art and combinations thereof. Non-limiting examples of extraction methods include the use of liquid-liquid extraction, solid phase extraction, water extraction, ethyl acetate extraction, alcohol extraction, acetone extraction, oil extraction, supercritical carbon dioxide extraction, heat extraction, pressure drop extraction, ultrasound extraction, and the like. The extract may be a liquid, solid, dried liquid, re-suspended solid, etc.

B. Amount of ingredient

It is contemplated that the compositions of the present invention may contain any amount of the ingredients discussed in this specification. The composition may also contain any number of combinations of additional ingredients (e.g., pigments or additional cosmetic or pharmaceutical ingredients) described throughout this specification. The concentration of any ingredient in the composition may vary. For example, in non-limiting embodiments, the composition in its final form may comprise, consist essentially of, or consist of: for example, at least about 0.0001%, 0.0002%, 0.0003%, 0.0004%, 0.0005%, 0.0006%, 0.0007%, 0.0008%, 0.0009%, 0.0010%, 0.0011%, 0.0012%, 0.0013%, 0.0014%, 0.0015%, 0.0016%, 0.0017%, 0.0018%, 0.0019%, 0.0020%, 0.0021%, 0.0022%, 0.0023%, 0.0024%, 0.0025%, 0.0026%, 0.0027%, 0.0028%, 0.0029%, 0.0030%, 0.0031%, 0.0032%, 0.0033%, 0.0034%, 0.0035%, 0.0036%, 0.0037%, 0.0038%, 0.0039%, 0.0040.0040%, 0051%, 0040.0043%, 0.0040.0040.0070%, 0060.0075%, 0.0070.0070.0070.0060.0060.0075%, 0.0070.0070.0070.0060.0075%, 0.0070.1%, 0.0040.0050.0075%, 0.0070.1%, 0.0050.0070.1%, 0.0040.0040.0050.0075%, 0.8%, 0.0070.0040.8%, 0.0040.0050.0070.8%, 0.0040.0050.8%, 0.8%, 0.0040.8%, 0.0040%, 0.0040.0040%, 0.0040.8%, 0.0040.0050.0070.8%, 0.0040.1%, 0.0050.0050.0050.8%, 0.8%, 0.0050.0050.8%, 0.0070.8%, 0.0070.0050.1%, 0.0050%, 0.0050.1%, 0.0050.0070.0050.0050.1%, 0.0070.1%, 0.0070.0050.1%, 0.1%, 0.0070.0070.1%, 0.0070.1%, 0.0070.0050.0070.1%, 0.1%, 0.0070.1%, 0.0050.0070.0070.0050.0050.0070.0050.0050.0050.0050.0050.0050.0075%, 0%, 0.0075%, 0.1%, 0.0070.0070.0070.0070.0070.0050.0070.0070.0070.1%, 0.0070.1%, 0.1%, 0.0070.1%, 0.0050.1%, 0.0050.0070.0040.1%, 0.1%, 0.0070.1%, 0.1%, 0.0070.1%, 0.1%, 0.0070.0050.1%, 0.1%, 0.0070.0070.1%, 0.0040.1%, 0.0040.0050.0070.0050.0070.0040.0070.0070.0070.1%, 0.1%, 0.0070.0070.1%, 0.1%, 0.0070.0070.0070.0070.0070.1%, 0.1%, 0.0040.0070.1%, 0.0070.1%, 0.0070, 0.0085%, 0.0086%, 0.0087%, 0.0088%, 0.0089%, 0.0090%, 0.0091%, 0.0092%, 0.0093%, 0.0094%, 0.0095%, 0.0096%, 0.0097%, 0.0098%, 0.0099%, 0.0100%, 0.0200%, 0.0250%, 0.0275%, 0.0300%, 0.0325%, 0.0350%, 0.0375%, 0.0400%, 0.0425%, 0.0450%, 0.0475%, 0.0500%, 0.0525%, 0.0550%, 0.0575%, 0.0600%, 0.0625%, 0.0650%, 0.0675%, 0.0700%, 0.0725%, 0.0750%, 0.0775%, 0.0800%, 0.0800.0800.085%, 0.0850%, 0.080.080%, 0.0900%, 0.9%, 0.460%, 0.9%, 9%, 9.590.9%, 0.9%, 9%, 0.9%, 0%, 0.9%, 0%, 0.9%, 0%, 0.9%, 0%, 0.9%, 0%, 0.9%, 0%, 0.9%, 0%, 0.9%, 0%, 0.9%, 0%, 0.9%, 0%, 0.9%, 0%, 0.9%, 0%, 0.9%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2.0%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3.0%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4.0%, 4.1%, 4.2%, 4.3%, 4.4%, 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, 5.0%, 5.1%, 5.2%, 5.3%, 5.4%, 5.5%, 5.6%, 5.7%, 5.8%, 5.9%, 6.0%, 6.1%, 6.2%, 6.3%, 6.4%, 6.5%, 6.6.6%, 6.7%, 7.9.7%, 7.9%, 8%, 7.9.9%, 7.9%, 7%, 8%, 7.9.9%, 7%, 7.9%, 8%, 7.9%, 7%, 7.9%, 8%, 7.9%, 7%, 8%, 7.9%, 7.9.9%, 7%, 8%, 7%, 8%, 7.9%, 7%, 8.8.8%, 7%, 7.9%, 7.8%, 7.8.8%, 7%, 7.9%, 7%, 7.8%, 7%, 7.8%, 7.8.8%, 7.9%, 7%, 7.8.8.8%, 7%, 7.0%, 7.9%, 7%, 7.8.8%, 7.9%, 7%, 7.8.8%, 7%, 7.9%, 8%, 8.9%, 7.9%, 7%, 7.9%, 7%, 8%, 7%, 7.8%, 8%, 7%, 7.9%, 8%, 7%, 7.9%, 7%, 7.8.9%, 7%, 8%, 7.9%, 7%, 7.9%, 7%, 7.9.9% or 8.9%, 7.9% or 8.9%, 7%, 7.9%, 7%, 7.9.9.9%, 7%, 7.9.9% or 8% or 8.9% or 8% or 6.9% or 8.9% or 8% or more, 9.9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 35%, 40%, 45%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% or any range available therein. In a non-limiting aspect, the percentages can be calculated by weight or volume of the entire composition. One of ordinary skill in the art will appreciate that the concentration in a given composition can vary depending on the addition, substitution, and/or subtraction of ingredients.

C. Carrier

The compositions of the present invention may comprise or be incorporated into all types of carriers and vehicles. The carrier or vehicle may be a pharmaceutically or dermatologically acceptable carrier or vehicle. Non-limiting examples of carriers or vehicles include water, glycerin, alcohols, oils, silicon-containing compounds, silicone compounds, and waxes. Variations and other suitable carriers will be apparent to the skilled artisan and are suitable for use in the present invention. In certain aspects, the concentrations and combinations of compounds, ingredients, and agents are selected in such a way that the compositions are chemically compatible and do not form complexes that precipitate out of the final product.

D. Structure of the product

The compositions of the present invention may be constructed or formulated in a variety of different forms. Non-limiting examples include emulsions (e.g., water-in-oil-in-water, silicone-in-water, water-in-silicone, oil-in-water-in-oil-in-water, oil-in-water-in-silicone emulsions), creams, lotions, solutions (aqueous or hydro-alcoholic solutions), anhydrous bases (e.g., lipsticks and powders), gels, masks, scrubs, body lotions, exfoliants, and ointments. Variations and other configurations will be apparent to the skilled artisan and are applicable to the present invention.

E. Additional ingredients

In addition to the combination of ingredients disclosed by the inventors, the composition may also comprise additional ingredients, such as cosmetic ingredients and pharmaceutical active ingredients. Non-limiting examples of these additional ingredients are described in the following subsections.

1. Cosmetic composition

CTFA international cosmetic ingredient dictionaries and manuals (2004 and 2008) describe a variety of non-limiting cosmetic ingredients that may be used in the context of the present invention. Examples of these ingredient classes include: fragrances (artificial and natural, such as gluconic acid, phenoxyethanol, and triethanolamine), dyes and coloring ingredients (e.g., blue 1 lake, red 40, titanium dioxide, D & C blue No. 4, D & C green No. 5, D & C orange No. 4, D & C red No. 17, D & C red No. 33, D & C violet No. 2, D & C yellow No. 10, and D & C yellow No. 11), flavoring/scenting agents (e.g., Stevia rebaudiana (Stevia rebaudiana) extract, and menthol), adsorbents, lubricants, solvents, moisturizers (including, for example, emollients, humectants, film formers, occlusive agents, and agents that affect the natural moisturizing mechanism of the skin), water repellents, UV absorbers (physical and chemical absorbers, such as p-benzoic acid ("PABA") and corresponding PABA derivatives, titanium dioxide, zinc oxide, and the like), essential oils, colorants, and coloring agents, Vitamins (e.g., A, B, C, D, E and K), trace metals (e.g., zinc, calcium, and selenium), anti-irritants (e.g., steroidal and non-steroidal anti-inflammatory drugs), plant extracts (e.g., Aloe vera (Aloe vera), citrus, cucumber extract, Ginkgo biloba (Ginko biloba), ginseng, and rosemary), antibacterial agents, antioxidants (e.g., BHT and tocopherol), chelating agents (e.g., disodium EDTA and tetrasodium EDTA), preservatives (e.g., methyl and propyl parabens), pH modifiers (e.g., sodium and citric acids), absorbents (e.g., aluminum starch octenyl succinate, kaolin, corn starch, oat starch, cyclodextrin, talc, and zeolites), skin bleaching and whitening agents (e.g., hydroquinone and niacinamide lactate), humectants (e.g., sorbitol, urea, methyl gluceth-20, sodium, and magnesium gluconate), and combinations thereof, Saccharide isomerate and mannitol), exfoliants, waterproofing agents (e.g., magnesium hydroxide/aluminum stearate), skin conditioning agents (e.g., aloe vera extract, allantoin, bisabolol, ceramide, polydimethylsiloxane, hyaluronic acid, biogel-1, ethylhexylglycerin, pentylene glycol, hydrogenated polydecene, octyldodecanol oleate, gluconolactone, calcium gluconate, cyclohexasiloxane, and dipotassium glycyrrhizinate). Non-limiting examples of some of these additional ingredients are provided in the subsections below.

UV absorbers and/or reflectors

UV absorbers and/or reflectors that may be used in combination with the compositions of the present invention include chemical and physical sunscreen substances. Non-limiting examples of chemical sunscreen substances that may be used include p-aminobenzoic acid (PABA), PABA esters (PABA glycerides, PABA amyl dimethyl esters, and PABA octyl dimethyl esters), PABA butyl esters, PABA ethyl dihydroxy propyl esters, benzophenones (oxybenzone, sulisobenzone, benzophenones, and benzophenone-1 to benzophenone-12), cinnamates (octyl methoxycinnamate (ethylhexyl methoxycinnamate)), isoamyl p-methoxycinnamate, octyl methoxycinnamate, cinoxate, diisopropyl methyl cinnamate, DEA methoxycinnamate, ethyl diisopropylcinnamate, dimethoxycinnamate and ethyl methoxycinnamate), cinnamates, salicylates (monomethyl salicylate, benzyl salicylate, ethylene glycol salicylate, isopropyl benzyl salicylate, etc.)), Anthranilate, ethyl urocanate, homosalate, octyl salicylate, dibenzoylmethane derivatives (e.g. avobenzone), octocrylene, octyl triazone, galloylgallate trioleate, glyceryl aminobenzoate, 2-hydroxy-1, 4-naphthoquinone and dihydroxyacetone, ethylhexyl triazone, dioctyl butamido triazone, benzylidene malonate polysiloxane, p-xylylene dicamphor sulfonic acid, disodium phenyldibenzoimidazole tetrasulfonate, diethylamino hydroxybenzoyl hexyl benzoate, bis-diethylamino hydroxybenzoyl benzoate, bis-benzoAzolylphenylethylhexyliminotriazine, cresoltrazol trisiloxane, methylenebisbenzotriazolyl tetramethylbutylphenol and bisethylhexyloxyphenol methoxyphenyl triazine, 4-methylbenzylidenecamphor and isoamyl 4-methoxycinnamate.Non-limiting examples of physical sunscreen substances include kaolin, talc, petrolatum, and metal oxides (e.g., titanium dioxide and zinc oxide).

b. Moisture-retaining agent

Non-limiting examples of humectants that can be used with the compositions of the present invention include amino acids, chondroitin sulfate, diglycerin, erythritol, fructose, glucose, glycerol polymers, ethylene glycol, 1,2, 6-hexanetriol, honey, hyaluronic acid, hydrogenated honey, hydrogenated starch hydrolysate, inositol, lactitol, maltitol, maltose, mannitol, natural moisturizing factor, PEG-15 butanediol, polyglycerol sorbitol, salts of pyrrolidone carboxylic acid, potassium PCA, propylene glycol, sodium glucuronate, saccharide isomers, sodium PCA, sorbitol, sucrose, trehalose, urea, and xylitol.

Other examples include acetylated lanolin, acetylated lanolin alcohol, alanine, algae extract, Aloe vera (Aloe barbadensis), Aloe vera extract, Aloe vera gel, althea officinalis (Aloe barbadensis) extract, apricot (prunus armeniaca) kernel oil, arginine aspartate, arnica extract, aspartic acid, avocado (avocado gratissima) oil, barrier sphingolipids, butanol, beeswax, behenyl alcohol, beta-sitosterol, white birch (betula alba) bark extract, borage (Borago officinalis) extract, pseudobulbus pseudolari (ruscus aculeatus) extract, butylene glycol, goldenrod flower extract, calendula oil, candelilla (euphularia cerifera) wax, rapeseed oil, caprylic/capric triglyceride, cardamomum (eletaria cardamonum) oil, palmitic acid (cornucopia officinalis) wax, castor oil (carob oil), carrot oil (carob) wax, carrot oil (carob oil), Ceramide, ceresin, ceteareth-5, ceteareth-12, ceteareth-20, cetearyl octanoate, ceteth-20, ceteth-24, cetoacetate, cetearyl octanoate, cetyl palmitate, chamomile (anthemius nobilis) oil, cholesterol ester, cholesterol hydroxystearate, citric acid, sage (salvia sclarea) oil, cocoa (theobroma) butter, coco-octanoate/decanoate, coconut (coco nucifera) oil, collagen amino acids, corn (zea mays) oil, fatty acids, decanoate, dimethicone copolyol, dimethiconol, dioctyl adipate, dioctyl succinate, pentaerythritol hexaoctanoate/hexadecanoate, pentaerythritol hexadecanoate, DNA, erythritol, ethoxydiglycol, ethyl linoleate, Eucalyptus globulus (Eucalyptus globulus) oil, evening primrose (oenothera biennis) oil, fatty acids, Geranium robustum (Geranium maculosum) oil, glucosamine, polyglutamate, glutamic acid, glyceryl polyether-26, glycerol, glyceryl distearate, glyceryl hydroxystearate, glyceryl laurate, glyceryl linoleate, glyceryl myristate, glyceryl oleate, glyceryl stearate SE, glycine, ethylene glycol stearate SE, glycosaminoglycans, grape (vitis vinifera) seed oil, hazel nut oil, hexylene glycol, hyaluronic acid, mixed safflower (carthamus tinctorius) oil, hydrogenated castor oil, hydrogenated coconut oil glyceride, hydrogenated lanolin, hydrogenated lecithin, Hydrogenated palm oil glyceride, hydrogenated palm kernel oil, hydrogenated soybean oil, hydrogenated tallow fatty acid glyceride, hydrogenated vegetable oil, hydrolyzed collagen, hydrolyzed elastin, hydrolyzed glycosaminoglycans, hydrolyzed keratin, hydrolyzed soybean protein, hydroxylated lanolin, hydroxyproline, isocetyl stearate, isocetyl stearoyloxy stearate, isodecyl oleate, isopropyl isostearate, isopropyl lanolate, isopropyl myristate, isopropyl palmitate, isopropyl stearate, isostearamide DEA, isostearic acid, isostearyl lactate, isostearyl pivalate, jasmine (jasminum officinale) oil, jojoba (buxus chinensis) oil, macroalgae, chestnut (aleurs moluccanana) nut oil, lactamide MEA, lanolin-16, lanolin alcohol polyether-10 acetate, lanolin fatty acid, lanolin alcohol, hydrolyzed collagen, hydrolyzed soybean protein, hydrolyzed lanolin, hydroxyproline, isocetyl stearate, isopropyl myristate, isopropyl palmitate, isopropyl oleate DEA, isopropyl oleate, isopropyl stearate, isopropyl oleate, isopropyl stearate, isopropyl oleate, isopropyl stearate, isopropyl oleate, isopropyl stearate, isopropyl oleate-16, lanolin-10, lanolin-16, lanolin-10, lanolin-10, lanolin-, Lanolin oil, lanolin wax, lavender (lavandala angustifolia) oil, lecithin, lemon (citrus medica limonum) oil, linoleic acid, linolenic acid, Macadamia ternifolia (Macadamia ternifolia) oil, maltitol, chamomile (chamomilla recutita) oil, methylglucotene sesquistearate, methylsilanol PCA ester, mineral oil, mink oil, Mortierella fulva oil, myristyl lactate, myristyl myristate, myristyl propionate, neopentyl glycol dicaprylate/dicaprate, octyldodecanol myristate, octyldodecanol stearate, octyl hydroxystearate, octyl palmitate, octyl salicylate, octyl stearate, oleic acid, olive (olea europaea) oil, orange (citrus aurantium dulcis) oil, palm (elaeisis guinssis) oil, palmitic acid, pantethine, panthenol, paraffin wax, PCA (paraffin wax), and mixtures thereof, Peach (prunus persica) kernel oil, peanut (arachis hypogaea) oil, PEG-8C12-18 acid ester, PEG-15 cocoamine, PEG-150 distearate, PEG-60 glyceryl isostearate, PEG-5 glyceryl stearate, PEG-30 glyceryl stearate, PEG-7 hydrogenated castor oil, PEG-40 hydrogenated castor oil, PEG-60 hydrogenated castor oil, PEG-20 methyl glucose sesquistearate, PEG-40 sorbitan monooleate, PEG-5 soya sterol, PEG-10 soya sterol, PEG-2 stearate, PEG-8 stearate, PEG-20 stearate, PEG-32 stearate, PEG-40 stearate, PEG-50 stearate, PEG-100 stearate, PEG-150 stearate, pentadecanolide, Peppermint (mentha piperita) oil, petrolatum, phospholipids, plankton extracts, polyamino acid polysaccharide condensates, polyglycerol-3 diisostearate, polyquaternium-24, polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, polysorbate 85, potassium myristate, potassium palmitate, propylene glycol dicaprylate/dicaprate, propylene glycol dicaprylate, propylene glycol dinonate, propylene glycol laurate, propylene glycol stearate SE, PVP, pyridoxine dipalmitate, retinol palmitate, rice (oryza sativa) bran oil, RNA, rosemary (rosmarinus officinalis) oil, rose oil, safflower (carthamus tinctorius) oil, sage (salvia officinalis) oil, sandalwood (saalbum) oil, serine, serum albumin, sesame (semum) oil, sesame (semolinum) butter, tallow (tallow) butter, and tallow (tallow) oil, Silk powder, sodium chondroitin sulfate, sodium hyaluronate, sodium lactate, sodium palmitate, sodium PCA, sodium polyglutamate, soluble collagen, sorbitan laurate, sorbitan oleate, sorbitan palmitate, sorbitan sesquioleate, sorbitan stearate, sorbitol, soybean (soybean soja) oil, sphingolipid, squalane, squalene, stearamide MEA-stearate, stearic acid, stearyloxypolydimethylsiloxane, stearyloxytrimethylsilane, stearyl alcohol, stearyl glycyrrhetinate, stearyl heptanoate, stearyl stearate, sunflower (heliothus annuus) seed oil, sweet almond (prunus amygdalus dulcis) oil, synthetic beeswax, tocopherol acetate, tocopherol linoleate, behenyl trimaran, tridecyl alcohol pivalate, tridecyl alcohol stearate, triethanolamine, tristearin, urea, vegetable oil, lecithin, and water, Water, wax, wheat (triticum vulgare) germ oil, and cananga odorata oil.

c. Antioxidant agent

Non-limiting examples of antioxidants that may be used with the compositions of the present invention include acetylcysteine, ascorbyl polypeptide, ascorbyl dipalmitate, ascorbyl methylsilanol pectate, ascorbyl palmitate, ascorbyl stearate, BHA, BHT, t-butylhydroquinone, cysteine HCI, dipentylhydroquinone, di-t-butylhydroquinone, dicetylthiodipropionate, dioleyl tocopheryl methylsilanol, disodium ascorbyl sulfate, distearylthiodipropionate, ditridecylthiodipropionate, lauryl gallate, erythorbic acid, ascorbyl esters, ethyl ferulate, ferulic acid, galloyl, hydroquinone, isooctyl thioglycolate, kojic acid, magnesium ascorbate, magnesium ascorbyl phosphate, methylsilanol ascorbate, natural plant antioxidants such as green tea or grape seed extract, Nordihydroguaiaretic acid, octyl gallate, phenyl thioglycolic acid, ascorbyl phosphate, potassium sulfite, propyl gallate, quinone, rosmarinic acid, sodium ascorbate, sodium bisulfite, sodium erythorbate, sodium metabisulfite, sodium sulfite, superoxide dismutase, sodium thioglycolate, sorbitolide, thiodiglycol, thiodiglycide, thioglycolic acid, thiolactic acid, thiosalicylic acid, tocopherol polyether-5, tocopherol polyether-10, tocopherol polyether-12, tocopherol polyether-18, tocopherol polyether-50, tocopherol, tocoferol acetate, tocopherol linoleate, tocopherol nicotinate, tocopherol succinate and tris (nonylphenol) phosphite.

d. Structuring agent

In other non-limiting aspects, the compositions of the present invention may comprise a structuring agent. In particular aspects, the structuring agent helps provide rheological characteristics to the composition to aid in the stability of the composition. In other aspects, the structuring agent may also function as an emulsifier or surfactant. Non-limiting examples of structuring agents include sodium cocoyl glutamate, hydroxypropyl cyclodextrin, stearic acid, palmitic acid, stearyl alcohol, cetyl alcohol, behenyl alcohol, stearic acid, palmitic acid, the polyethylene glycol ether of stearyl alcohol having an average of about 1 to about 21 ethylene oxide units, the polyethylene glycol ether of cetyl alcohol having an average of about 1 to about 5 ethylene oxide units, and mixtures thereof.

e. Emulsifier

In a particular aspect of the invention, the composition does not comprise an emulsifier. However, in other aspects, the composition may comprise one or more than one emulsifier. Emulsifiers can reduce the surface tension between phases and improve the formulation and stability of the emulsion. The emulsifiers may be nonionic, cationic, anionic and zwitterionic emulsifiers (see U.S. Pat. Nos. 5011681; 4421769; 3755560). Non-limiting examples include glycerol esters, propylene glycol esters, fatty acid esters of ethylene glycol, fatty acid esters of polypropylene glycol, esters of sorbitol, sorbitan esters, carboxylic acid copolymers, esters and ethers of glucose, ethoxylated esters, ethoxylated alcohols, alkyl phosphate esters, polyoxyethylene fatty ether phosphate esters, fatty acid amides, lactyl lactate, fatty acid salts, TEA stearate, DEA oleyl polyether-3 phosphate, polyethylene glycol 20 sorbitan monolaurate (polysorbate 20), polyethylene glycol 5 sterol, steareth-2, steareth-20, steareth-21, ceteth-20, cetearyl glucoside, cetearyl alcohol, C12-13 alkanol polyether-3, PPG-2 methyl glucose ether distearate, PPG-5-cetyl polyether-20, PPG-2 methyl glucose ether distearate, bis-PEG/PPG-20/20 dimethicone, ceteth-10, polysorbate 80, cetyl phosphate, potassium cetyl phosphate, diethanolamine cetyl phosphate, polysorbate 60, glyceryl stearate, PEG-100 stearate, arachidyl alcohol glucoside, and mixtures thereof.

f. Silicone-containing compound

In a non-limiting aspect, the silicone-containing compound includes any member of a family of polymerization products whose molecular backbone is comprised of alternating silicon and oxygen atoms with pendant groups attached to the silicon atoms. By varying the length of the-Si-O-chains, the pendant groups, and the cross-linking, silicones can be synthesized as a wide variety of materials. Their consistency can vary from liquid to gel to solid.

Silicone-containing compounds that may be used in the context of the present invention include those described in the specification or known to one of ordinary skill in the art. Non-limiting examples include silicone oils (e.g., volatile and non-volatile oils), gels, and solids. In certain aspects, the silicon-containing compound comprises a silicone oil, such as a polyorganosiloxane. Non-limiting examples of polyorganosiloxanes include polydimethylsiloxane, cyclomethicone, cyclohexasiloxane, polysiloxane-11, phenyl trimethicone, trimethylsilylaminomethylsiloxane, stearyloxytrimethylsilane, or mixtures thereof, and other organosiloxane materials in any given ratio to achieve a desired consistency and application characteristics depending on the intended application (e.g., to a particular area such as skin, hair, or eye). "volatile silicone oils" include silicone oils having a low heat of vaporization, i.e., generally less than about 50 calories per gram of silicone oil. Non-limiting examples of volatile silicone oils include: cyclopolydimethylsiloxanes such as Dow Corning 344 Fluid, Dow Corning 345 Fluid, Dow Corning 244 Fluid and Dow Corning 245 Fluid, vollate Silicon 7207 (Union Carbide Corp., Danbury, Connecticut); low viscosity polydimethylsiloxanes, i.e., polydimethylsiloxanes having a viscosity of about 50cst or less (e.g., polydimethylsiloxanes such as Dow Corning 200-0.5 cst Fluid). Dow Corning Fluid is commercially available from Dow Corning Corporation of Midland, Mich. In the third edition of the CTFA cosmetic ingredient dictionary, incorporated by reference, cyclomethicones and dimethicones are described as mixtures of cyclic dimethylpolysiloxane compounds and fully methylated linear siloxanes end-capped with trimethylsiloxy units, respectively. Other non-limiting volatile Silicone oils that may be used in the context of the present invention include those available from General Electric co, silicon Products div, walford, new york, and SWS Silicones div, of Stauffer Chemical co, edlien, michigan.

g. Exfoliating agent

Exfoliating agents include ingredients that remove dead skin cells from the outer surface of the skin. These agents may act mechanically, chemically, and/or otherwise. Non-limiting examples of mechanical exfoliants include abrasives such as pumice, silica, cloth, paper, shells, beads, solid crystals, solid polymers, and the like. Non-limiting examples of chemical exfoliants include acid and enzymatic exfoliants. Acids useful as exfoliants include, but are not limited to, glycolic acid, lactic acid, citric acid, alpha hydroxy acids, beta hydroxy acids, and the like. Other exfoliating agents known to those skilled in the art are also contemplated as being useful within the scope of the present invention.

h. Essential oil

Essential oils include oils from herbs, flowers, trees, and other plants. Such oils typically exist as tiny droplets between plant cells and can be extracted by several methods known to those skilled in the art (e.g., steam distillation, floral extraction (i.e., using fat extraction), maceration, solvent extraction, or mechanical pressing). These types of oils tend to volatilize (i.e., volatile oils) when exposed to air. Thus, while many essential oils are colorless, they oxidize and become darker in color over time. Essential oils are insoluble in water, but soluble in alcohols, ethers, fixed oils (vegetable) and other organic solvents. Typical physical characteristics found in essential oils include a boiling point of about 160 ℃ to 240 ℃ and a density of about 0.759 to about 1.096.

Essential oils are generally named by the plant from which the oil is found. For example, rose oil or peppermint oil is from rose or peppermint plants, respectively. Non-limiting examples of essential oils that may be used in the context of the present invention include sesame oil, macadamia nut oil, tea tree oil, evening primrose oil, spanish sage oil, spanish rosemary oil, coriander oil, thyme oil, rose oil, anise oil, balsamine oil, bergamot oil, rosewood oil, cedar oil, chamomile oil, sage oil, clary sage oil, clove oil, cedar oil, eucalyptus oil, fennel oil, sea fennel oil, frankincense oil, geranium oil, ginger oil, grapefruit oil, jasmine oil, juniper oil, lavender oil, lemon oil, lemongrass oil, lime oil, mandarin oil, marjoram oil, myrrh oil, bitter orange flower oil, orange oil, green leaf oil, pepper oil, black pepper oil, bitter orange leaf oil, pine oil, rose oil, rosemary oil, sandalwood oil, spearmint oil, spikenard oil, vetiver oil, wintergreen oil, ylang-ylang oil. Other essential oils known to those skilled in the art are also contemplated to be useful in the context of the present invention.

i. Thickening agent

Thickeners, including thickeners or gelling agents, include substances that can increase the viscosity of the composition. Thickeners include those that can increase the viscosity of the composition without substantially altering the efficacy of the active ingredients within the composition. Thickeners may also increase the stability of the compositions of the present invention. In certain aspects of the invention, the thickener comprises hydrogenated polyisobutylene, trihydroxystearin, ammonium acryloyldimethyl taurate/VP copolymer, or a mixture thereof.

Non-limiting examples of additional thickeners that may be used in the context of the present invention include carboxylic acid polymers, crosslinked polyacrylate polymers, polyacrylamide polymers, polysaccharides, and gums. Examples of carboxylic acid polymers include crosslinked compounds containing one or more than one monomer derived from acrylic acid, substituted acrylic acids and salts and esters of these acrylic acids and substituted acrylic acids, wherein the crosslinking agent contains two or more carbon-carbon double bonds and is derived from a polyol (see U.S. Pat. No. 5087445; No. 4509949; No. 2798053; CTFA International cosmetic ingredient dictionary, fourth edition, 1991, pages 12 and 80). Examples of commercially available carboxylic acid polymers includeCarbomers, which are homopolymers of acrylic acid crosslinked with allyl ethers of sucrose or pentaerythritol (e.g., CARBOPOL available from b.f. goodrich)TM900 series).

Non-limiting examples of crosslinked polyacrylate polymers include cationic and nonionic polymers. Examples are described in U.S. patent nos. 5100660, 4849484, 4835206, 4628078, 4599379.

Non-limiting examples of polyacrylamide polymers (including nonionic polyacrylamide polymers, including substituted branched or unbranched polymers) include polyacrylamide, isoparaffins, and laureth-7, multi-block copolymers of acrylamide and acrylamide substituted with acrylic acid and substituted acrylic acid.

Non-limiting examples of polysaccharides include cellulose, carboxymethyl hydroxyethyl cellulose, cellulose acetate propionate, hydroxyethyl cellulose, hydroxyethyl ethyl cellulose, hydroxypropyl methyl cellulose, methyl hydroxyethyl cellulose, microcrystalline cellulose, sodium cellulose sulfate, and mixtures thereof. Other examples are alkyl substituted celluloses, wherein the hydroxyl groups of the cellulose polymer are hydroxyalkylated (preferably hydroxyethylated or hydroxypropylated) to form a hydroxyalkylated cellulose, which is then further modified with C10 to C30 linear or branched alkyl groups via ether linkages. Typically these polymers are ethers of C10 to C30 linear or branched alcohols with hydroxyalkyl celluloses. Other useful polysaccharides include scleroglucans, which comprise a linear chain of (1-3) linked glucose units having one (1-6) linked glucose per three units.

Non-limiting examples of gums that may be used in the present invention include gum arabic, agar, algin, alginic acid, ammonium alginate, pullulan, calcium alginate, calcium carrageenan, carnitine, carrageenan, dextrin, gelatin, gellan gum, guar hydroxypropyltrimonium chloride, hectorite, hyaluronic acid, hydrated silica, hydroxypropyl chitosan, hydroxypropyl guar, caraya gum, kelp, carob gum, natto gum, potassium alginate, potassium carrageenan, propylene glycol alginate, sclerotium gum, sodium carboxymethyl dextran, sodium carrageenan, tragacanth gum, xanthan gum, and mixtures thereof.

j. Preservative

Non-limiting examples of preservatives that may be used in the context of the present invention include quaternary ammonium preservatives, such as polyquaternium-1 and benzalkonium halides (e.g., benzalkonium chloride ("BAC") and benzalkonium bromide), parabens (e.g., methyl and propyl parabens), phenoxyethanol, benzyl alcohol, chlorobutanol, phenol, sorbic acid, thimerosal, or combinations thereof.

2. Pharmaceutical composition

It is also contemplated that pharmaceutically active ingredients are useful for the compositions of the present invention. Non-limiting examples of pharmaceutically active ingredients include anti-acne agents, agents for treating rosacea, analgesics, anesthetics, anorectic agents, antihistamines, anti-inflammatory agents including non-steroidal anti-inflammatory agents, antibiotics, antifungal agents, antivirals, antimicrobial agents, anticancer agents, anti-sarcoptics, pediculicides, antineoplastics, antiperspirants, antipruritics, antipsoriatics, anti-seborrheics, biologically active proteins and polypeptides, burn treatment agents, cauterizing agents, depigmenting agents, depilatory agents, diaper rash treatment agents, enzymes, hair growth stimulants, hair growth inhibitors including DFMO and its salts and analogs, hemostatic agents, keratolytic agents, aphthous treatment agents, cold sore treatment agents, dental or periodontal treatment agents, light sensitive active agents, skin protectants/barriers, steroids including hormones and corticosteroids, sunburn treatment agents, Sunscreens, transdermal active agents, nasal active agents, vaginal active agents, wart treatment agents, wound healing agents, and the like.

F. Reagent kit

Kits for use in certain aspects of the invention are also contemplated. For example, the compositions of the present invention may be included in a kit. The kit may comprise a container. The container may comprise a bottle, metal tube, laminated tube, plastic tube, dispenser, pressurized container, barrier container, package, compartment, lipstick container, pressurized container, cosmetic tray capable of holding a cosmetic composition, or other type of container, such as an injection or blow molded plastic container, in which the dispersion or composition is held or the desired bottle, dispenser, or package. The kit and/or container may comprise indicia on its surface. For example, the indicia may be words, phrases, abbreviations, pictures or symbols.

The container may dispense a predetermined amount of the composition. In other embodiments, the container (e.g., a metal tube, a laminated tube, or a plastic tube) can be squeezed to dispense the desired amount of the composition. The composition may be dispensed as a spray, aerosol, liquid, fluid, or semi-solid. The container may have a spraying, pumping or squeezing mechanism. The kit may also include instructions for using the kit components and using any other compositions contained within the container. The instructions may include instructions for using and preserving the composition.

Examples

The following examples are set forth to illustrate preferred embodiments of the present invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to establish preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.

All of the compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

Example 1

(exemplary formulations)

Formulations having the ingredients disclosed herein are prepared as topical skin compositions. In some cases, topical skin compositions may be prepared as ampoules, serums, creams, lotions, gels, and gel emulsions. The formulations in table 1 are examples of topical skin compositions prepared as ampoules.

TABLE 1^ s

Composition (I) % concentration (by weight)
Water (W) 79.2
Gluconolactone 5
Glycolic acid 4
Butanediol 3
Glycerol 3
Potassium hydroxide 2.6
Betaine 1.5
Methyl glucitol polyether-20 0.5
PEG-8 polydimethylsiloxane; 0.5
phenoxyethanol 0.3

Composition (I) % concentration (by weight)
Hydroxyethyl cellulose 0.3
Octylene glycol 0.1
Excipients Proper amount of

The formulation can be prepared by mixing the ingredients in a beaker at 70 ℃ to 75 ℃ with heat until homogeneous. The formulation can then be cooled to standard room temperature (20 ℃ to 25 ℃). In addition, if desired, additional ingredients may be added, for example, to modify the rheological properties of the composition or ingredients that provide benefit to the skin.

For example, excipients may be added to modify the rheological properties of the composition. Alternatively, the amount of water may vary, provided that the amount of water in the composition is at least 40% by weight, preferably from 50% to 80% by weight.

Example 2

(clinical efficacy research)

It was unexpected that the use of a combination of glycolic acid and gluconolactone was effective in reducing skin roughness as measured by images captured by visionascan VC 98 and analyzed using visionascan VC 98 software for roughness. The data indicate that the combination of ingredients may act synergistically or that the combination of glycolic acid and gluconolactone may be an effective combination to reduce skin roughness.

A randomized controlled clinical study was conducted to evaluate the efficacy of a treatment product to provide skin smoothness within 15 minutes after use. The study was conducted for more than one hour on each participant for a total of three days, with participants acclimating to environmentally controlled room conditions of 70 ° ± 5 ° F and 35% ± 15% relative humidity for at least fifteen (15) minutes, exposing their forearms prior to each set of measurements of baseline and post-treatment product use tests. The test treatment product used by the participants was the formulation of table 1, which contained 5% gluconolactone and 4% glycolic acid ("test product"). No additional product is used. Evaluation of the skin of each participant was performed at baseline and fifteen (15) minutes after treatment ("post-treatment"). The baseline was measured 15 minutes after acclimatization. The method used for evaluation included using VisioScan VC 98(Courage + Khazaka, germany) to take images to examine the smoothness of the skin in the area 2X2cm of the right forearm of each participant. The images taken were then analyzed using VisioScan VC 98 software measuring the roughness value SE.

Participants were thirty (30) subjects, of which twenty-six (26) healthy volunteers (age 21 to 63 years) were participants who completed the study. The selected participants were: age 21 to 65 years, overall health is good, and the forearms are free of tattoos, scars, and other obstructions. Participants agreed not to use any moisturizing product on the forearms in the morning of the study and during the study, except for the test product provided. Participants agreed not to drink caffeine-containing beverages (e.g., coffee, tea, soda) at least one hour prior to the initial study visit and during the study period. Participants agreed not to perform rigorous exercise activities twenty-four (24) hours prior to and during the study. Volunteers with any known allergic reactions to cosmetics and toiletries, pregnant at the time of study, intended to be pregnant or lactating, taking medications that researchers believe would interfere with the results of the study, or having skin conditions that researchers believe would interfere with the results of the study were excluded from the study.

Three (3) days of rinsing were completed by each participant prior to product testing. For three (3) days of rinsing, until the participants applied (i) only the provided detergent at the test site and (ii) no other product. On the day of product testing, the 2X2cm area on the forearm of the right palm of each participant was marked ("test site"). The study duration for each participant was about one (1) hour. Participants were acclimated to environmentally controlled indoor conditions of 70 ° ± 5 ° F and 35% ± 15% relative humidity for at least fifteen (15) minutes with their forearms exposed. An image of the test site was taken using a Visioscan VC 98(Courage + Khazaka, Germany). The test treatment product was applied to the test site of each participant and the forearms were exposed for fifteen minutes. Fifteen minutes after the test treatment product was applied, an image of the test site was taken using a VisioScan VC 98.

The VisioScan VC 98 is the digitization process of an image, consisting of a black and white video sensor chip with very high resolution, the target (test site) and a UVA light source in a plastic box. When taking an image, the two special halide lights arranged on opposite sides uniformly illuminate the skin. The VisioScan VC 98 uses the arrangement of light illuminating the skin, the intensity and the spectrum of the light to monitor only the stratum corneum and not the reflections from deeper layers of the skin. Images of the skin were taken by an embedded CCD camera over a measurement area of 6X8 cm. At intervals, one copy/image is taken at each test site, which serves as an untreated control and test area, since the image capture area is wider than the test site. Images captured by VisioScan VC 98 were analyzed using embedded software for coarseness SE.

The skin roughness shown in table 2 was measured using images captured by VisioScan VC 98, which were compared to baseline measurements. Table 3 shows the average roughness, the average percent change from baseline, and the p-value for the test treated product and untreated control at baseline and fifteen minutes later. The post-treatment test treatment product showed a significant improvement in skin smoothness (roughness reduction) compared to baseline. The untreated control showed statistically no significant improvement after treatment compared to baseline. The post-treatment test treatment products showed a significant improvement in skin smoothness (reduction in roughness) compared to the untreated control.

TABLE 2

Lambda shows significant change when compared to baseline (p ≦ 0.05).

Display the percent change when compared to baseline.

Example 3

(additional analysis)

Assays that can be used to determine the efficacy of any one ingredient, or any combination of ingredients or combination of ingredients, having a combination of ingredients as disclosed throughout the specification and claims can be determined by methods known to those of ordinary skill in the art. The following are non-limiting assays that may be used in the context of the present invention. It should be appreciated that other testing procedures may be used, including, for example, objective and subjective procedures.

Antioxidant (AO) analysis: antioxidant assays can be performed on skin cells (e.g., epidermal keratinocytes, fibroblasts, and/or dermal endothelial cells) to determine whether any of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed in this specification are inhibited by(2,2' -azino-bis- [ 3-ethylbenzodihydrophthalazinone sulfonic acid)]) Oxidized by ferric myoglobin to ABTS. +, to provide antioxidant (TEAC) capability. The antioxidant system of living organisms may comprise enzymes, e.g. hyper-oxidationSuperoxide dismutase, catalase, and glutathione peroxidase; macromolecules such as albumin, ceruloplasmin, and ferritin; and a large number of small molecules including ascorbic acid, alpha-tocopherol, beta-carotene, reduced glutathione, uric acid, and bilirubin. The total number of endogenous and food-derived antioxidants represents the total antioxidant activity of the extracellular fluid. The cooperation of all the different antioxidants may provide greater protection against attack by reactive oxygen or nitrogen radicals than any single compound alone. Thus, the total antioxidant capacity may give more relevant biological information than the information obtained by measuring the individual components, since it takes into account the cumulative effect of all antioxidants present in plasma and body fluids. The ability of the ingredients in the composition to prevent ABTS oxidation was compared to the water soluble tocopherol analogue Trolox and quantified as molar equivalents of Trolox. Total antioxidant capacity can be measured using antioxidant capacity kit #709001 from Cayman Chemical (Anneaberg, Mich., USA).

Collagen stimulation assay: the collagen stimulation assay can be used to determine the ability of any of the active ingredients, combinations of ingredients, or compositions with the combinations disclosed in this specification to increase expression of procollagen-1 (a precursor of collagen). Collagen (type I, type II, type III, type IV and type V) can be synthesized as precursor molecules called procollagen. These precursor molecules may contain additional peptide sequences, commonly referred to as "propeptides", at the amino-and carboxy-termini. During cellular expression and secretion, procollagen can be assembled in the form of trimers, which are then cleaved by specific endopeptidases at specific N-and C-terminal sites, yielding three fragments: procollagen-1N-terminal propeptide (PINP), type I collagen, and procollagen-1 carboxy-terminal propeptide (PICP).

The function of the pro-peptide is to promote the winding of the pro-collagen molecule into a triple-helical conformation within the endoplasmic reticulum. The propeptide may be cleaved from the collagen triple-helical molecule during its secretion process, after which the triple-helical collagen polymerizes into extracellular collagen fibrils. Thus, the amount of free propeptide reflects stoichiometrically the amount of collagen molecule synthesized (similar to the relationship between the carboxy-terminal peptide of proinsulin and endogenous insulin). Collagen is an extracellular matrix protein that is critical to skin structure. Increased collagen synthesis helps to improve skin firmness and elasticity.

Quantitative detection of PICP in fibroblast extracts and culture supernatants can be performed using an enzyme immunoassay kit (e.g., Takara # MK101) to assess the effect of these components on PICP synthesis in skin. This bioassay can be used to examine the effect of human epidermal fibroblasts on the production of procollagen peptide (collagen precursor). The endpoint of the assay may be a spectrophotometric measurement, which reflects the presence of procollagen peptides and cellular activity. The assay employs a quantitative sandwich enzyme immunoassay technique whereby monoclonal antibodies specific for procollagen peptide are pre-coated onto a microplate. The standard and sample can be pipetted into the well and any procollagen peptide present is bound by the immobilized antibody. After washing away all unbound material, an enzyme-linked polyclonal antibody specific for procollagen peptide was added to the wells. After washing to remove all unbound antibody-enzyme reagent, a substrate solution may be added to the wells to develop a color proportional to the amount of procollagen peptide bound in the initial step. The development was stopped and the intensity of the colour at 450nm was measured using a microplate reader.

For the generation of samples and controls, the samples and controls can be grown in standard DMEM growth medium containing 10% fetal bovine serum (Mediatech) at 37 deg.C with 10% CO2Sub-confluent normal adult epidermal fibroblasts (Cascade Biologics) were cultured. Cells can be treated with each test component and control for 3 days. Following incubation, the cell culture medium can be collected and the amount of type I procollagen peptide secreted quantified using a sandwich enzyme-linked immunosorbent assay (ELISA) from Takara (# MK101) as described above.

Elastin stimulation assay: elastin is a connective tissue protein that helps the skin to recover shape after stretching or contraction. Elastin is also an important load protein for use in locations where storage of mechanical energy is required. Elastin is made by linking a number of soluble tropoelastin molecules in a lysyl oxidase-catalyzed reaction. Elastin secretion and elastin fibers in cultured human fibroblasts can be monitored by direct ELISA sandwich staining of cultured human fibroblasts with immunofluorescent antibodies to elastin. Results can be analyzed using a Meso Scale Discovery system SECTOR 2400 imaging system. The change in elastin secretion and elastin fibers caused by one or more than one component of the composition may be determined by incubating cultured human fibroblasts with the active ingredient for a period of time prior to probing the cells or their lysates with an antibody directed against elastin.

Laminin stimulation assay: laminins are the major protein in the dermal-epidermal junction (DEJ), also known as the basement membrane. The DEJ is located between the dermis and the epidermal junction, which forms a finger-like protrusion called the epidermal process. Epidermal cells receive their nutrients from blood vessels in the dermis. The epidermal process increases the surface area of the epidermis exposed to these blood vessels and the nutrients required. The DEJ provides adhesion of the two tissue compartments and controls the structural integrity of the skin. Laminins are structural glycoproteins located in the DEJ. Laminin, together with fibronectin, is thought to be a glue that holds cells together, both secreted by dermal fibroblasts to help promote intracellular and intercellular adhesion of epidermal cells to DEJ.

Secretion of laminin can be monitored by quantifying laminin in the cell supernatant of cultured human fibroblasts treated for 3 days with or without 1.0% of the final concentration of the test component. After incubation, laminin content can be measured in an enzyme-linked immunosorbent assay (ELISA) using immunofluorescent antibodies directed against each protein.

Enzyme activity of matrix metalloproteinase 1 (MMP-1) assay: MMPs are extracellular proteases that by virtue of their broad substrate specificity play a role in many normal and disease states. MMP-1 substrates include collagen IV. A molecular probe Enz/Chek gelatinase/collagenase detection kit (# E12055) can be used to detect MMP-1 protease activity, which utilizes a fluorescent gelatin substrate and detects proteolytic cleavage of the substrate by purified MMP-1 enzyme. After proteolytic cleavage of the substrate, a bright green fluorescence was exhibited and monitored using a fluorescent microplate reader to measure enzyme activity. The test material is incubated in the presence or absence of purified enzyme and substrate to determine its protease inhibitor ability.

Enzyme Activity analysis of matrix metalloproteinases 3 and 9 (MMP-3; MMP-9): MMPs are extracellular proteases that by virtue of their broad substrate specificity play a role in many normal and disease states. MMP-3 substrates include collagen, fibronectin, and laminin; whereas MMP9 substrates include collagen VII, fibronectin, and laminin. A Colorimetric Drug Discovery kit for MMP3(AK-400) and MMP-9(AK-410) from BioMol International can be used to determine MMP protease activity using thiopeptide as the chromogenic substrate (Ac-PLG- [ 2-mercapto-4-methyl-pentanoyl ] -LG-OC2H5)5, 6. The peptide bond of the MMP cleavage site is replaced by a thioester bond in a thiopeptide. This bond is hydrolyzed by MMP to produce a thiol group which reacts with DTNB [5,5' -dithiobis (2-nitrobenzoic acid), elman reagent ] to produce 2-nitro-5-thiobenzoic acid, which can be detected by its absorbance at 412nm (e.g., 13600M-1cm-1 at pH 6.0 and above 7).

Lipoxygenase (LO) analysis: lipoxygenase assays can be used to determine the ability of any of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed in the present specification to inhibit the expression of Lipoxygenase (LO). LO is an iron-containing dioxygenase, a non-heme enzyme, that catalyzes the addition of molecular oxygen to fatty acids. Linoleate and arachidonate are the main substrates of LO in plants and animals. Arachidonic acid can then be converted to hydroxyeicosatetraenoic acid (HETE) derivatives, which are subsequently converted to leukotrienes, a potent inflammatory mediator. Accurate and convenient screening of lipoxygenase inhibitors can be achieved by measuring the hydroperoxides produced by incubation of lipoxygenase (5-LO, 12-LO or 15-LO) with arachidonic acid. Colorimetric LO inhibitor screening kits (#760700, Cayman Chemical) can be used to determine the ability of a component of a composition to inhibit enzymatic activity.

The purified 15-lipoxygenase and the test components can be mixed in assay buffer and incubated with shaking for 10 minutes at room temperature. After incubation, arachidonic acid may be added to start the reaction and the mixture may be incubated at room temperature for an additional 10 minutes. The catalysis was stopped by the addition of a colorimetric substrate and the color change was assessed by fluorescence plate reading at 490 nm. The percent inhibition of lipoxygenase activity can be calculated as compared to an untreated control to determine the ability of the components of the composition to inhibit the activity of the purified enzyme.

Tumor necrosis factor- α (TNF- α) assay: the prototype ligand of the TNF superfamily, TNF- α, is a pleiotropic cytokine that plays a central role in inflammation. The increase in its expression is associated with an upregulation of pro-inflammatory activity. Bioassays can be used to analyze the effect of the composition's ingredients on the production of TNF-alpha by human epidermal keratinocytes. The endpoint of the assay may be a spectrophotometric measurement reflecting the presence of TNF-alpha and cellular activity. The assay may employ a quantitative sandwich enzyme immunoassay technique whereby monoclonal antibodies specific for TNF-alpha are pre-coated onto a microplate.

Standards and samples can be transferred into the wells of a microplate and any TNF-alpha present is bound by the immobilized antibody. After washing away all unbound material, an enzyme-linked polyclonal antibody specific for TNF- α can be added to the wells. After washing to remove all unbound antibody-enzyme reagent, a substrate solution can be added to the wells and detected at 450nm using a microplate reader, with the color developing in proportion to the amount of TNF-alpha bound in the initial step. The development may be terminated and the intensity of the color may be measured. 5% CO at 37 ℃2In EPILIFETMSecondary confluent normal adult keratinocytes cultured in standard growth medium (Cascade Biologics) can be treated with phorbol-12-myristate 13-acetate (PMA, 10ng/ml, Sigma Chemical, # P1585-1MG) and components of the composition or no test components (for negative control) for 6 hours. PMA was shown to result in a significant increase in TNF- α secretion, which peaked 6 hours after treatment. Following incubation, the cell culture medium can be collected and used from R&Sandwich enzyme-linked immunosorbent assay (ELISA) of D Systems (# DTA00C) quantitated TNF-. alpha.secretion.

Elastase assay: from Molecular Probes (Eugenin)Oregon, USA) ofThe elastase assay (kit # E-12056) can be used as an in vitro enzyme inhibition assay for measuring inhibition of elastase activity in the presence of composition ingredients. The EnzChek kit may contain soluble bovine cervical ligament elastin, which is labeled with a dye to quench the fluorescence of the conjugate. The non-fluorescent substrate can be digested by elastase or other proteases to produce highly fluorescent fragments. The resulting fluorescence enhancement can be monitored with a fluorescent microplate reader. The digestion product from the elastin substrate has an absorption maximum at about 505nm and a fluorescence emission maximum at about 515 nm. When screening elastase inhibitors using the EnzChek elastase assay kit, the peptide N-methoxysuccinyl-Ala-Pro-Val-chloromethyl ketone can be used as a selective collective inhibitor of elastase for a positive control.

Fibronectin stimulation assay: fibronectin is the major protein in the dermal-epidermal junction (DEJ), also known as the basement membrane. The DEJ is located between the dermis and the epidermal junction, forming a finger-like protrusion called the epidermal process. Epidermal cells receive their nutrients from blood vessels in the dermis. The epidermal process increases the surface area of the epidermis exposed to these blood vessels and the nutrients required. The DEJ provides adhesion of the two tissue compartments and controls the structural integrity of the skin. Fibronectin is a structural glycoprotein located in the DEJ. Fibronectin, together with laminin, is thought to be a glue that holds cells together, both secreted by dermal fibroblasts to help promote intra-and intercellular adhesion of epidermal cells to DEJ.

The secretion of fibronectin can be monitored by quantifying fibronectin in the cell supernatants of cultured human fibroblasts treated for 3 days with or without 1.0% of the final concentration of the test component. Following incubation, fibronectin content can be measured in an enzyme-linked immunosorbent assay (ELISA) using immunofluorescent antibodies to each protein.

Lysyl oxidase assay: lysyl oxidase assays can be performed on skin cells (e.g., epidermal keratinocytes, fibroblasts, and/or dermal endothelial cells) to determine the ability of any of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed herein to stimulate lysyl oxidase expression in skin. Lysyl oxidase can catalyze the cross-linking of elastin and collagen, thereby providing a structurally stronger substrate for the skin. By increasing the expression of lysyl oxidase, increased crosslinking of elastin and collagen may occur, which may be beneficial in reducing the appearance of fine lines, wrinkles, skin laxity, and/or inelastic skin.

B16 pigmentation analysis: melanogenesis is the process by which melanocytes produce melanin, a naturally occurring pigment that imparts color to the skin, hair, and eyes. Inhibiting melanogenesis is beneficial for preventing skin dullness associated with aging and reducing dark spots. The bioassay can employ B16-F1 melanocytes (ATCC) (immortalized mouse melanoma cell line) to analyze the effect of compounds on melanogenesis. The end point of the assay may be a spectrophotometric measurement of melanin production and cellular activity. Can be 10% CO at 37 deg.C2B16-F1 melanocytes were cultured in standard DMEM growth medium containing 10% fetal bovine serum (Mediatech) and then treated with any one of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed in the present specification. Following incubation, melanin secretion was measured by absorption at 405nm and cellular activity was quantified.

ORAC analysis: any of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed herein can also be analyzed for oxygen radical absorption (or absorbance) capability (ORAC) by measuring the antioxidant activity of the ingredients or compositions. Antioxidant activity indicates the ability to reduce the oxidizing agent (oxidizer). This assay quantifies the extent and time required to inhibit the activity of oxidizing agents, such as oxygen radicals, known to cause damage to cells (e.g., skin cells). The ORAC value of any of the active ingredients, combinations of ingredients, or compositions having such combinations disclosed in this specification can be determined by methods known to those of ordinary skill in the art (see U.S. patent publication nos. 2004/0109905 and 2005/0163880; and commonly available kits such as the Zen-Bio ORAC antioxidant assay kit (# AOX-2)). Zen-Bio ORAC antioxidant assay kit (# AOX-2) measured the loss of fluorescein fluorescence over time due to the formation of peroxy radicals by decomposition of AAPH (2,2' -azobis-2-methylpropionamidine dihydrochloride). Trolox, a water-soluble vitamin E, acts in a dose-dependent manner as a positive control that inhibits the decay of fluorescein.

Production of hyaluronic acid: changes in the production of hyaluronic acid in human dermal fibroblasts can be determined as a result of any of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed in the present specification. HA is a polysaccharide that is associated with the stability of the matrix structure, and is also associated with providing turgor pressure to tissues and cells. As a non-limiting example, Hyaluronan DuoSet ELISA kit from R can be used&D Systems (DY3614) determines HA production in treated and untreated adult dermal fibroblast (HDFa) cells. In this test, for the generation of the sample, before the treatment, at 37 ℃ and 10% CO2Next, sub-confluent HDFa cells (C-13-5C) obtained from Cascade Biologics were cultured in starvation medium (0.15% bovine fetal serum and 1% penicillin streptomycin solution in Dulbecco's modified Eagle medium) for 72 hours. The cells were then cultured with fresh starvation medium for 24 hours using test compound, positive control (phorbol 12-myristate 13-acetate from Sigma-Aldrich (P1585) and platelets derived from growth factor from Sigma-Aldrich (P3201)) or no additive control. The medium was then collected and frozen at-80 ℃ until use in an ELISA assay.

Briefly, the ELISA assay employs a quantitative sandwich enzyme immunoassay technique, so that capture antibodies specific to HA can be pre-coated on a microplate. Standards, media from treated and untreated cells were pipetted into the microplate to allow any HA present to be bound by the immobilized antibody. After washing away all unbound material, an enzyme-linked detection antibody specific for HA was added to the wells. After washing to remove all unbound antibody-enzyme reagent, a substrate solution is added to the wells, allowing color to develop in proportion to the amount of HA bound in the initial step. The color development is stopped at a specific time and the intensity of the color can be measured at 450nm using a microplate reader.

Production of occludin: changes in keratinocytes due to occludin of each of the active ingredients, any of the combinations of ingredients, or compositions having the combinations disclosed in the specification can be determined. Occludin is a protein important for the formation of tight junctions and the skin's moisture barrier function. A non-limiting example of how occluding protein production can be determined in treated and untreated keratinocytes is by using a biological assay that analyzes the concentration of occluding protein in the keratinocyte lysate. Use ofSIMONTMThe immunoblotting protocol performs this biological analysis. For the samples, at 37 ℃ and 5% CO2In EPILIFE, adult epidermal keratinocytes (HEKa) derived from Life Technologies (C-005-5C)TMGrowth medium was grown for 24 hours, and Epilife Growth medium contained calcium from Life Technologies (M-EP-500-CA) supplemented with Keratinocyte Growth Supplement (HKGS) from Life Technologies (S-101-5). Test compounds/extracts, no compounds/extracts for negative control, or 1mM CaCl were then used2For positive control, HEKa was cultured in growth medium for 24 to 48 hours. HEKa was then washed, collected and stored on ice or colder objects until lysed with lysis buffer. The protein concentration of the sample can be determined and used to normalize the sample. The lysate was stored at-80 ℃ until use in bioassays.

SIMONTMWestern blot bioanalysis uses quantitative western blot immunoassay techniques that use antibodies specific for occludin to quantitatively detect occludin in a sample. Cell samples were lysed and normalized for protein concentration. The normalized sample and molecular weight standards were then loaded onto and run on a denatured protein separation gel using capillary electrophoresis. The proteins in the gel were then immobilized and immunodetected using primary antibodies specific for occludin. The immobilized protein was immunodetected with an enzyme-linked detection antibody that binds to the primary antibody. A chemiluminescent substrate solution is then added to the immobilized protein to make the chemiluminescent coloration proportional to the amount of occludin bound in the immobilization. The chemiluminescent development may be terminated at a specific time and the intensity of the chemiluminescent signal determined and compared to positive and negative controls.

Keratinocyte monolayer permeability: the change in permeability of the keratinocyte monolayer due to any of each active ingredient, combination of ingredients, or composition having the combination disclosed in the present specification can be determined. The permeability of the keratinocyte monolayer is a measure of the integrity of the skin barrier. As a non-limiting example, the permeability of a keratinocyte monolayer in treated and untreated keratinocytes can be determined using the in vitro vascular permeability test of Millipore (ECM 642). This assay analyzes endothelial cell adsorption, transport and infiltration. Briefly, adult epidermal keratinocytes from Life Technologies (C-005-5C) can be seeded onto porous collagen-coated membranes within the collection wells. At 37 ℃ and 5% CO2In (2), keratinocytes were cultured in Epilife Growth medium containing calcium from Life Technologies (M-EP-500-CA) supplemented with Keratinocyte Growth Supplement (HKGS) from Life Technologies (S-101-5) for 24 hours. The incubation time allowed the cells to form a monolayer and close the membrane pores. The medium was then replaced with fresh medium with (test sample) or without (untreated control) test compound/extract, keratinocytes at 37 ℃ and 5% CO2And cultured for another 48 hours. After the culture in the presence/absence of test compounds/extracts, in order to determine the permeability of the keratinocyte monolayer, the culture medium was replaced with fresh medium containing high molecular weight Fluorescein Isothiocyanate (FITC) -Dextran, the keratinocytes were incubated at 37 ℃ and5% CO2And cultured for another 4 hours. During the 4 hour incubation, FITC can pass through the keratinocyte monolayer and the porous membrane into the collection well at a rate proportional to the monolayer membrane permeability. After 4 hours of incubation, cell viability and FITC content in the collection wells can be determined. For FITC content, the media in the collection wells was collected and the fluorescence of the media was measured at 480nm (Em) upon excitation at 520 nm. The percent permeability and percent change compared to the untreated control can be determined by the following equations: percent permeability ═ ((average Ex/Em for test samples)/average Ex/Em for untreated controls) × 100; percent change-percent permeability of the test sample-percent permeability of the untreated control.

Mushroom tyrosinase activity assay: in mammalian cells, tyrosinase catalyzes two steps in the multistep biosynthesis of melanin from tyrosine (and from dopachrome polymerization). Tyrosinase is localized in melanocytes and produces melanin (aromatic quinone compounds) that imparts color to skin, hair, and eyes. Purified mushroom tyrosinase (Sigma) can be incubated with its substrate L-dopa (fisher) in the presence or absence of each active ingredient, any combination of ingredients, or a composition with such a combination as disclosed herein. Pigment formation can be assessed by reading on a color scale at 490 nm. The percent inhibition of mushroom tyrosinase activity can be calculated by comparison to an untreated control to determine the ability of the test ingredient or combination thereof to inhibit purified enzyme activity. The inhibition of the test extracts was compared to that of kojic acid (Sigma).

Cyclooxygenase (COX) assay: cyclooxygenase-1 and cyclooxygenase-2 (COX-1, COX-2) inhibition assays in vitro. COX is a bifunctional enzyme that exhibits both cyclooxygenase and peroxidase activities. Cyclooxygenase activity converts arachidonic acid into hydroperoxides endoperoxide (prostaglandin G2; PGG2), and the peroxidase component reduces endoperoxide (prostaglandin H2; PGH2) to the corresponding alcohol, prostaglandin, blood coagulationPrecursors of alkanes and prostacyclanes. The COX inhibitor screening assay measures the peroxidase component of the cyclooxygenase enzyme. Peroxidase activity was analyzed colorimetrically by monitoring the appearance of oxidized N, N' -tetramethyl-p-phenylenediamine (TMPD). The inhibition screening assay includes both COX-1 and COX-2 enzymes to screen for isozyme specific inhibitors. A colorimetric COX (ovine) inhibitor screening assay (#760111, Cayman Chemical) can be used to assay the effect of the active ingredients disclosed in the specification, a combination of any of the ingredients, or a composition having the combination on the activity of a purified cyclooxygenase enzyme (COX-1 or COX-2). The purified enzyme, heme, and test extract can be mixed in assay buffer and incubated with shaking at room temperature for 15 minutes, as indicated by the manufacturer. After incubation, arachidonic acid and a colorimetric substrate may be added to start the reaction. The color change can be assessed by reading on a color plate at 590 nm. The percent inhibition of COX-1 or COX-2 activity can be calculated by comparison to an untreated control to determine the ability of the test extract to inhibit the activity of the purified enzyme.

Oil control analysis: the assays used to measure the reduction of sebum secretion in sebaceous glands and/or the reduction of sebum production in sebaceous glands can be analyzed by using standard techniques known to those of ordinary skill in the art. In a specific example, a forehead may be used. Each active ingredient, any combination of ingredients, or composition having the combination disclosed in this specification can be administered once or twice daily to a portion of the forehead for a fixed day (e.g., 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, or more than 14 days), while other portions of the forehead are not treated with the composition. After expiration of a fixed period of days, sebum secretion can be analyzed by applying a fine oil absorbing paper to treated and untreated forehead skin. This is accomplished by first removing all sebum from the treated and untreated areas with a wet and dry cloth. An oil absorbing paper is then applied to the treated and untreated forehead area, and a rubber band may be placed around the forehead to gently press the oil absorbing paper against the skin. After 2 hours, the oil absorbing paper can be removed, allowed to dry and then transilluminated. A deeper oil absorbing paper corresponds to more sebum secretion (or a shallower oil absorbing paper corresponds to reduced sebum secretion).

Erythema analysis: the test for measuring the reduction of skin redness can be evaluated using a Minolta Chromometer. Cutaneous erythema may be initiated by applying a 0.2% sodium lauryl sulfate solution to the subject's forearm. The area was protected with a closed patch for 24 hours. After 24 hours, the patch is removed and the a from Minolta Chroma Meter can be used*The values were evaluated for redness induced by stimulation. a is*Values measure the change in skin color in the red region. Immediately after reading, the area is treated with any of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed in the specification. Repeated measurements may be made periodically to determine the ability of the formulation to reduce redness and irritation.

Skin moisture/hydration analysis: the benefit of skin moisture/hydration can be measured using impedance measurements performed with the Nova derm Phase Meter. The impedance meter measures the change in skin moisture content. The outer layers of skin have different electrical properties. When the skin is dry, it conducts electricity poorly. As it becomes more aqueous, increased conductivity results. Thus, changes in skin impedance (related to conductivity) can be used to assess changes in skin hydration. The device can be calibrated for each test day according to the instrument specifications. Temperature and relative humidity may also be marked. The subject may be evaluated as follows: it can be equilibrated in a room with a defined humidity (e.g. 30% to 50%) and temperature (e.g. 68 ℃ to 72 ℃) before measurement. Three independent impedance measurements were taken on each side of the face and recorded and averaged. The impedance meter may be set using T5, which averages the impedance value every five seconds applied to the face. Changes can be reported as statistical variance and significance. Each active ingredient, combination of ingredients, or composition having the combination disclosed in the specification can be analyzed.

Analysis of skin clarity and reduction of freckles and age spots: skin clarity and reduction of freckles and age spots were evaluated using a Minolta Chromometer. Skin color change may be usedA of Minolta Chroma Meter*The values are evaluated to determine the likelihood of irritation due to product handling. a is*Values measure the change in skin color in the red region. This is used to determine whether each active ingredient, combination of any one of the ingredients, or composition with the combination disclosed in the specification induces stimulation. The measurements may be taken on each side of the face and averaged as the left and right face values. Skin clarity can also be measured using a Minolta Meter. The measurement is a for the Minolta Meter*B, and L values, and is related to the brightness of the skin, and very well corresponds to the smoothness and hydration of the skin. Skin measurements were performed as above. In one non-limiting aspect, skin clarity can be described as L/C, where C is chroma and is defined as (a)2+b2)1/2

Dry skin, surface fine lines, skin smoothness and skin color analysis: dry skin, surface fine lines, skin smoothness and skin tone can be assessed using clinical scoring techniques. For example, the clinical score for dry skin can be determined by the five-point standard Kligman scale: (0) the skin is soft and moist; (1) the skin appeared normal with no visible dryness; (2) the skin felt slightly dry to the touch without visible flaking; (3) the skin feels dry, tough and has a whitish appearance of scales; and (4) the skin feels very dry, rough and has a whitish appearance of scales. The assessments may be performed independently by two clinicians and averaged.

Skin color clinical score analysis: clinical scoring of skin tone can be performed by a ten-point analog numerical scale: (10) smooth, even skin, pink-brown color. There were no dark, red or scaly plaques when examined with a hand-held magnifying glass. The micro-texture of the skin is very uniform to the touch; (7) the uniform skin color was observed without a magnifying glass. There were no scaly areas, but there was slight discoloration due to pigmentation or erythema. No discoloration of greater than 1cm in diameter; (4) skin discoloration and uneven texture was easily noticed. Slightly scaly. Some areas feel rough skin; and (1) uneven skin coloration and texture. Areas with scales and discoloration, hypopigmentation, redness or dark spots. Large areas of uneven coloration with diameters exceeding 1 cm. The assessments were performed independently by two clinicians and averaged.

Clinical score analysis of skin smoothness: the clinical score of skin smoothness can be analyzed on a ten point analog numerical scale: (10) smooth, the skin is moist and shiny, there is no resistance to the fingers sliding across the surface; (7) a certain degree of smoothness and slight resistance; (4) rough, visibly changing, frictional when rubbed; and (1) rough, flaky, uneven surfaces. The assessments were performed independently by two clinicians and averaged.

Skin smoothness and wrinkle reduction analysis using the method disclosed by packaman et al (1978): (1978): reduction of skin smoothness and wrinkles can also be visually assessed using the method disclosed by packaman et al (1978). For example, the depth, shallowness and total number of surface lines (SFLs) for each subject can be carefully scored and recorded at the time of visit. The fraction of the number is obtained by multiplying the number factor by the depth/width/length factor. The scores of the eye area and mouth area (left and right) are obtained and added together as the total wrinkle score.

Visualization analysis of lines and wrinkles with replica: the appearance of lines and wrinkles on the skin can be assessed using a replica, which is an impression of the skin surface. Materials such as silicone rubber may be used. The replica can be analyzed by image analysis. The change in line and wrinkle visibility can be objectively quantified by forming the face of the object with a silicon replica and analyzing the replica image with a computer image analysis system. Replicas can be taken from the eye area and neck area and photographed with a digital camera at low illumination angles of incidence. The digital image may be analyzed with an image processing program and the areas of the replica covered by wrinkles and fine lines determined.

Skin firmness analysis with Hargens Ballistometer: skin firmness can be measured using a Hargens Ballistometer, a device that assesses skin elasticity and firmness by dropping a small object on the skin and recording the first two rebound peaks. The Ballistometry is a small lightweight probe using a relatively blunt probe (4 mm square-contact area). The probe penetrates gently into the skin, resulting in measurements that depend on the properties of the outer layers of the skin, including the stratum corneum and the outer epidermis, as well as a portion of the dermis.

Skin softness/flexibility analysis with Gas Bearing electrodynameter: skin softness/flexibility can be evaluated using a Gas Bearing electrodynameter, an instrument that measures skin pressure/tension properties. The viscoelastic properties of skin are related to skin moisturization. Measurement of the predetermined site in the cheek region may be achieved by attaching a probe to the skin surface with a double sided tape. Approximately 3.5gm of force is applied parallel to the skin surface, accurately measuring the displacement of the skin. The flexibility of the skin can then be calculated and expressed as DSR (dynamic spring rate in gm/mm).

Skin surface profile analysis by profilometer/stylus method: the skin surface profile can be measured by a method using a profilometer/stylus. This involves flashing or dragging the stylus across the replica surface. The vertical displacement of the stylus can be recorded into a computer by means of a distance sensor, and after scanning the replica over a certain distance, an analysis of the skin contour can be generated as a two-dimensional surface. The scan may be repeated any number of times along a fixed axis to produce a simulated 3-D image of the skin. Ten random replica cross sections can be obtained using stylus technology and combined to produce an average. Values of interest include Ra, which is the arithmetic mean of all roughness (height) values calculated by integrating the profile height relative to the average profile height. Rt, which is the maximum vertical distance between the highest peak and lowest valley, and Rz, which is the average peak amplitude minus the average peak height. Values are given as values in mm. The device should be normalized by scanning a metal standard of known value before each use. The Ra value can be calculated by the following formula: raNormalized roughness; lmTransverse (scan) length; and y is the absolute value (x-axis) of the profile position relative to the mean profile height.

MELANODERMTMAnd (3) testing: in other non-limiting aspects, the skin can be treated by using a skin analog such as melandodermTMTo evaluate the efficacy of any one of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed in the specification. Melanocytes, one of the cells in the skin analog, are clearly contaminated when exposed to L-dihydroxyphenylalanine (L-DOPA), a precursor of melanin. Skin analog melandodermTMEach active ingredient disclosed in the specification, a combination of any one of the ingredients, or various bases of the composition having the combination, or a single base as a control may be used for the treatment. Alternatively, an untreated sample of a skin analog can be used as a control.

Production of silk fibroin: changes in keratinocytes due to the silk polyprotein of any of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed in the present specification can be determined. Silk polyprotein is a precursor of Natural Moisturizing Factor (NMF) in the skin. Increased NMF increased the moisture content in the skin. Biological assays to analyze silk polyprotein concentration in keratinocyte lysates are used to determine silk polyprotein production in treated and untreated keratinocytes. A non-limiting example of a biological assay that can be used to quantify silk fibroin production isSIMONTMWestern blot protocol. For each sample, Normal Human Epidermal Keratinocytes (NHEK) were incubated with calcium-containing EPI-200-Mattek EPILIFE from Life Technologies (M-EP-500-CA)TMGrowing in a growth medium. 5% CO at 37 ℃ before treatment2NHEK was grown in growth medium overnight. The NHEK was then incubated in growth medium with 1% test compound/extract or without compound/extract (negative control) for 24 to 36 hours. The NHEK may then be washed, collected and stored on ice or colder objects until lysed with lysis buffer and sonicated. Determining the sampleProtein concentration was used to normalize samples. The lysate can be stored at-80 ℃ until use in a quantitative assay.

SIMONTMWestern blot bioanalysis uses quantitative western blot immunoassay techniques that use antibodies specific for silk fibroin to quantitatively detect silk fibroin in a sample. Cell samples were lysed and normalized for protein concentration. The normalized sample and molecular weight standards can then be loaded onto and run on a denatured protein separation gel using capillary electrophoresis. Proteins within the gel were immobilized and immunodetected using primary antibodies specific for silk polyprotein. The immobilized protein can then be immunodetected with an enzyme-linked detection antibody that binds to the primary antibody. A chemiluminescent substrate solution may then be added to the immobilized protein to make the chemiluminescent coloration proportional to the amount of silk polyprotein bound in the immobilization. The chemiluminescent development is terminated at a specified time and the intensity of the chemiluminescent signal can be determined and compared to positive and negative controls.

Inhibition of hyaluronidase activity: changes in hyaluronidase activity can be determined for any of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed in the specification. Hyaluronidase is an enzyme that breaks down HA. HA is a polysaccharide that is associated with the stability of the matrix structure, and is also associated with providing turgor pressure to tissues and cells. As a non-limiting example, hyaluronic acid activity may be determined using a modified in vitro protocol derived from Sigma-Aldrich protocol # EC 3.2.1.35. Briefly, hyaluronic acid type 1-S from Sigma-Aldrich (H3506) was added to reaction wells of microplates containing test compounds or controls. Tannic acid can be used as a positive control inhibitor, no test compound is added to the enzyme for control, and wells with test compound or a positive control without hyaluronic acid can be used as a negative control for background. Wells were incubated at 37 ℃ for 10 min prior to addition of substrate (HA). Substrate was added and the reaction was incubated at 37 ℃ for 45 minutes. Then, a part of each reaction solution was transferred and gently mixed in a solution of sodium acetate and acetic acid having a pH of 3.75 to stop the part of the reaction (stopped well). After a portion of the reaction solution is added to the stopped well, both the stopped well and the reaction well should contain the same volume of solution. Both reaction wells and stopped wells were incubated at room temperature for 10 minutes. The absorbance at 600nm of the reaction wells and the stopped wells was then measured. Inhibition can be calculated using the following formula: inhibitor (or control) activity ═ (inhibitor stopped well absorbance at 600 nm-inhibitor reaction well absorbance at 600 nm); initial activity-control enzyme absorbance at 600 nm; percent inhibition ═ [ (initial activity/inhibitor activity) × 100] -100.

Activity of peroxisome proliferator-activated receptor γ (PPAR- γ): changes in PPAR-gamma activity due to any of the active ingredients, combinations of ingredients, or compositions having the combinations disclosed in the specification can be determined. PPAR-gamma is a receptor critical for sebum production. By way of non-limiting example, PPAR- γ activity can be assayed using a biological assay that analyzes the ability of a test compound or composition to inhibit ligand binding. Briefly, FluORMONE, available from Life Technologies (PV4894), can be used as a fluorescent small molecule pan-PPAR ligandTMPan-PPAR Green to determine whether the tested compound or composition is capable of inhibiting ligand binding to PPAR-gamma. The sample wells contain PPAR-gamma and a fluorescent ligand and a test compound or composition (test); a reference inhibitor; rosiglitazone (positive control); or no test compound (negative control). The wells are incubated for a set period of time to allow an opportunity for ligand binding to PPAR-gamma. The fluorescence polarization of each sample well can then be measured and compared to a negative control well to determine the percent inhibition of the test compound or composition.

Cytokine analysis: human epidermal keratinocytes were cultured to a confluency of 70% to 80%. The plate was aspirated of medium and 0.025% trypsin/EDTA was added. When the cells became confluent, the dish was gently tapped to release the cells. Cells containing trypsin/EDTA were removed from the culture dish and neutralized. Will be provided withCells were centrifuged for 5 minutes. Cell pellets were formed at 180 Xg. The supernatant was aspirated. In EPILIFETMThe pellets obtained were resuspended in culture medium (Cascade Biologics). Cells were seeded into 6-well plates at about 10% to 20% confluency. After the cells become approximately 80% confluency, the medium is aspirated and 1.0ml of EPILIFE is addedTMAnd phorbol 13-myristate 12-acetate ("PMA") (known as an inflammation inducer) and test composition dilutions were added to two replicate wells (i.e. 1.0% (100 μ l of 100X stock) and 0.1% (10 μ l of 100X stock) of test composition diluted to a final volume of 1ml of episife growth medium). The medium was gently stirred to ensure adequate mixing. In addition, 1.0ml of EPILIFE was added to control wells with or without additional PMATM. After dosing, the plates were placed at 37. + -. 1 ℃ and 5.0. + -. 1% CO2And incubated for about 5 hours. After 5 hours of incubation, all media were collected in conical tubes and frozen at-70 ℃.

For analysis, 16 hybridization cassettes were attached to 16 pieces of FAST slides with a triplicate array of 16 anti-cytokine antibodies plus experimental controls (Whatman Bioscience) and the slides placed in FAST frames (4 slides per frame) for processing. At room temperature, the arrays were blocked for 15 minutes using 70ml S & S protein array blocking buffer (Whatman Schleicher and Scheull). The blocking buffer was removed and 70ml of the respective supernatant sample was added to each array. The arrays were incubated at room temperature for 3 hours under gentle shaking. The array was washed 3 times with TBS-T. The arrays were treated with 70ml of a mixed antibody containing one biotinylated antibody corresponding to each array capture antibody. The arrays were incubated at room temperature for 1 hour with gentle shaking. The array was washed 3 times with TBS-T. Incubation was performed at room temperature for 1 hour using 70ml of a solution containing streptavidin-Cy 5 conjugate under gentle shaking. The array was washed 3 times with TBS-T, rinsed rapidly in deionized water and dried.

Slides can be imaged in a Perkin-Elmer ScanArray 4000 confocal fluorescence imaging system. Array images can be saved and analyzed using Imaging Research ArrayVision software. Briefly, spot intensity is determined by removing the background signal. Spot replicates from each sample condition can be averaged and then compared to the appropriate control.

Endothelial cell tube formation: the formation of endothelial cells involves angiogenesis and microcapillary formation. Capillary formation and vascularization can contribute to skin redness and rosacea. The ability of endothelial cells to form tubes can be determined in cell culture systems using a capillary disruption assay with preformed primary Human Umbilical Vein Endothelial Cells (HUVECs) in the presence or absence of test extracts and compounds.

Briefly, HUVECs are cultured in vitro in an extracellular matrix that stimulates tubular morphogenesis of the junctures and endothelial cells to form a capillary-like luminal structure. In many respects, these in vitro formed capillaries are similar to human capillaries. Capillary tests are based on this phenomenon and are used to evaluate potential vasculature targeting agents.

HUVEC cultures in 5% CO2Growth in cell culture at 37 ℃. The complete growth medium for HUVEC was endothelial cell basal Medium (EBM) supplemented with 2% Fetal Bovine Serum (FBS), 12. mu.g/ml bovine brain extract, 1. mu.g/ml cortisol and 1. mu.g/ml GA-1000 (gentamicin-amphotericin B). HUVEC cultures between passage 3 and passage 8 can be used for all assays.

HUVECs were pre-labeled with the fluorescent reagent Calcein AM and seeded into the extracellular matrix of 96-well culture plates coated with their entire growth medium. Approximately 4 hours after the morphogenetic process, endothelial capillaries formed. Then, as a treatment condition, a set dose of test reagent was applied to the formed capillary culture in a volume of 50 μ l. A vehicle for the test agent can be added to the no treatment control. Sutent, an FDA approved anti-angiogenic drug, may be associated with analytical performance control. At about 6 hours after the treatment, images of each well were obtained by microscopy to examine the endothelial cell tube morphology in each well and to quantify the capillary destruction activity under the treatment conditions. Each test condition can be performed in duplicate wells, including a control group.

**************

All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

31页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:新型闭塞制剂

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!

技术分类