Allowed Publications
Slot System
Featured Buckets
Featured Buckets Admin

Circadian rhythms, part 2: Can treating cutaneous conditions at different times of the day improve outcomes?

Article Type
Changed
Fri, 01/15/2021 - 09:04

We continue with a focus on circadian rhythms and implications for skin care this month, paying particular attention to research or insights pertaining to specific skin conditions, when possible, as well as clinical studies that may shed light on how to time skin care treatments.

Vectorial illustration of a clock with human hands, representing the circadian rhythm of sleep and wakefulness. In the balanced cycle, a half circle contains the night and the other one the day.
Francesca Bellini/iStock/Getty Images

It is important to remember that several studies in the last 20 years have revealed cutaneous tendencies based on the time of day. For instance, sebum production is known to be highest around noon, and pH also peaks during the day and is at its lowest at night.1-5
 

Skin aging

In 2019, Dong and associates showed that blue light at 410 nm reduces PER1 transcription in keratinocytes, indicating that epidermal cells have the capacity to directly sense light and regulate their own clock gene expression. With the introduction of blue light at night, circadian rhythm is disrupted as epidermal skin cells act as if it is daytime. The investigators also considered blue light–induced damage to skin cells at various doses and exposure times in comparison with cells that remained unexposed to light. The production of reactive oxygen species increased in the exposed cells, as did DNA impairment and the emergence of inflammatory mediators, all of which have the potential to hasten aging.6

Early this year, Dong and associates demonstrated that melatonin can dose-dependently stimulate PER1 clock gene expression in normal human dermal fibroblasts and normal human epidermal keratinocytes, and verified that the MT-1 melatonin receptor in such fibroblasts manifests a marked decline with age. The researchers concluded that the melatonin pathway contributes significantly in cutaneous aging and impairment, and that its relationship with skin circadian rhythm points to a possible role in slowing the rate of skin aging through the modulation of cutaneous melatonin receptors.7
 

Wound healing

In 2019, Walker and associates investigated the effects of dim artificial light at night on wound healing in female C57BL/6 mice, and found that those conditions prior to wounding reduced healing. They concluded that such information might warrant consideration in prescribing treatment.8

Atopic dermatitis

Vaughn and associates contended that alterations in circadian rhythm may contribute to the development of atopic dermatitis.9 A good example of the impact of circadian rhythms on cutaneous health is the nocturnal exacerbation of atopic dermatitis, particularly in children.10

Psoriasis

According to Plikus and associates, recent evidence has emerged showing that the circadian clock regulates UVB-induced DNA damage and cutaneous cancers, and it is also associated with the immune-mediated disorder psoriasis.11

Clinical studies

In 2018, Deshayes and associates conducted a clinical study to evaluate the precursors and stem cell attributes of hHF (human hair follicle keratinocytes), hEpi (human interfollicular epidermal keratinocytes), and hHFDP (hair follicle dermal papilla stem cells) in response to clock pathway changes caused by long-term deregulation of circadian rhythms. A total of 20 women participated in the study, 10 in each group (day workers were the control group and compared with shift workers). Two 3-mm fresh punch biopsies were collected from the occipital region of each participant. The investigators reported that chronic circadian rhythm deregulation influenced clock pathway protein expression and correlated with changes in hHF, hEpi, and hHFDP. They concluded that their findings represented the first data in humans suggesting that deregulation of the clock pathway modulates regenerative activity in human cutaneous and hair precursor cells.12

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Later that year, Wu and associates reported on the role of the circadian clock in the transcriptional regulation of human epidermis. Investigators sampled 20 human participants through a 24-hour period and a population of 219 people once, finding a potent circadian oscillator in human epidermis at the population level, hundreds of rhythmically expressed genes, as well as a biomarker set for human epidermis that can, with one sample, highlight circadian phase within a 3-hour time frame. The team concluded that rhythms in human epidermis persist at the population level, and that they were able to present an effective single-sample circadian biomarker.13 This is important, as Morris pointed out, because the standard practice for measuring an individual’s internal clock is to use a dim-light melatonin onset assay over the course of a day.14 In 2019, Jia and associates studied the skin surface lipid profiles of young women to evaluate and characterize circadian human facial surface lipid composition. The investigators identified significant markers of circadian rhythm, with glycerolipids most affected. They ascribed changes in skin barrier function, such as variable pH and transepidermal water loss, to alterations in triacylglycerol levels as well as free fatty acid chain lengths and content that were affected by variations in circadian rhythm.15
 

Sleep and the timing of topicals

Based on their recent review of the literature on circadian rhythm and skin, Lyons and associates argued that an understanding of circadian rhythm helps dermatologists in recommending the optimal times for patients to apply topical medications. They added that urging patients to get sufficient sleep is important because DNA repair of the skin occurs best at that time.16

Conclusions

Doctors have known for half a century that timing drug delivery to a patient’s circadian clock can enhance outcomes. Chronobiological research into how circadian rhythms work at the cellular level, and in cutaneous cells in particular, is a fascinating and expanding area of inquiry that could help dermatologists more accurately recommend timing for skin care regimens. Much more research, especially in clinical trials, is necessary to further elucidate how to best work with the skin’s natural rhythms.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on skin care technologies. Write to her at dermnews@mdedge.com.

References

1. Mehling A et al. Skin Pharmacol Physiol. 2006;19(4):182-9.

2. Latreille J et al. Skin Pharmacol Physiol. 2004 May-Jun;17(3):133-40.

3. Le Fur I et al. J Invest Dermatol. 2001 Sep;117(3):718-24.

4. Verschoore M et al. Chronobiol Int. 1993 Oct;10(5):349-59.

5. Yosipovitch G et al. J Invest Dermatol. 1998 Jan;110(1):20-3.

6. Dong K et al. Int J Cosmet Sci. 2019 Dec;41(6):558-62.

7. Dong K et al. Int J Mol Sci. 2020 Jan 3;21(1):326.

8. Walker WH II et al. Arch Dermatol Res. 2019 Sep;311(7):573-6.

9. Vaughn AR et al. Pediatr Dermatol. 2018 Jan;35(1):152-7.

10. Fishbein AB et al. J Allergy Clin Immunol. 2015 Nov;136(5):1170-7.

11. Plikus MV et al. J Biol Rhythms. 2015 Jun;30(3):163-82.

12. Deshayes N et al. Eur J Dermatol. 2018 Aug 1;28(4):467-75.

13. Wu G et al. Proc Natl Acad Sci U S A. 2018 Nov 27;115(48):12313-8.

14. Morris A. Nat Rev Endocrinol. 2018 Dec;15(1):3.

15. Jia Y et al. Exp Dermatol. 2019 Jul;28(7):858-62.

16. Lyons AB et al. J Clin Aesthet Dermatol. 2019 Sep;12(9):42-5.
 

Publications
Topics
Sections

We continue with a focus on circadian rhythms and implications for skin care this month, paying particular attention to research or insights pertaining to specific skin conditions, when possible, as well as clinical studies that may shed light on how to time skin care treatments.

Vectorial illustration of a clock with human hands, representing the circadian rhythm of sleep and wakefulness. In the balanced cycle, a half circle contains the night and the other one the day.
Francesca Bellini/iStock/Getty Images

It is important to remember that several studies in the last 20 years have revealed cutaneous tendencies based on the time of day. For instance, sebum production is known to be highest around noon, and pH also peaks during the day and is at its lowest at night.1-5
 

Skin aging

In 2019, Dong and associates showed that blue light at 410 nm reduces PER1 transcription in keratinocytes, indicating that epidermal cells have the capacity to directly sense light and regulate their own clock gene expression. With the introduction of blue light at night, circadian rhythm is disrupted as epidermal skin cells act as if it is daytime. The investigators also considered blue light–induced damage to skin cells at various doses and exposure times in comparison with cells that remained unexposed to light. The production of reactive oxygen species increased in the exposed cells, as did DNA impairment and the emergence of inflammatory mediators, all of which have the potential to hasten aging.6

Early this year, Dong and associates demonstrated that melatonin can dose-dependently stimulate PER1 clock gene expression in normal human dermal fibroblasts and normal human epidermal keratinocytes, and verified that the MT-1 melatonin receptor in such fibroblasts manifests a marked decline with age. The researchers concluded that the melatonin pathway contributes significantly in cutaneous aging and impairment, and that its relationship with skin circadian rhythm points to a possible role in slowing the rate of skin aging through the modulation of cutaneous melatonin receptors.7
 

Wound healing

In 2019, Walker and associates investigated the effects of dim artificial light at night on wound healing in female C57BL/6 mice, and found that those conditions prior to wounding reduced healing. They concluded that such information might warrant consideration in prescribing treatment.8

Atopic dermatitis

Vaughn and associates contended that alterations in circadian rhythm may contribute to the development of atopic dermatitis.9 A good example of the impact of circadian rhythms on cutaneous health is the nocturnal exacerbation of atopic dermatitis, particularly in children.10

Psoriasis

According to Plikus and associates, recent evidence has emerged showing that the circadian clock regulates UVB-induced DNA damage and cutaneous cancers, and it is also associated with the immune-mediated disorder psoriasis.11

Clinical studies

In 2018, Deshayes and associates conducted a clinical study to evaluate the precursors and stem cell attributes of hHF (human hair follicle keratinocytes), hEpi (human interfollicular epidermal keratinocytes), and hHFDP (hair follicle dermal papilla stem cells) in response to clock pathway changes caused by long-term deregulation of circadian rhythms. A total of 20 women participated in the study, 10 in each group (day workers were the control group and compared with shift workers). Two 3-mm fresh punch biopsies were collected from the occipital region of each participant. The investigators reported that chronic circadian rhythm deregulation influenced clock pathway protein expression and correlated with changes in hHF, hEpi, and hHFDP. They concluded that their findings represented the first data in humans suggesting that deregulation of the clock pathway modulates regenerative activity in human cutaneous and hair precursor cells.12

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Later that year, Wu and associates reported on the role of the circadian clock in the transcriptional regulation of human epidermis. Investigators sampled 20 human participants through a 24-hour period and a population of 219 people once, finding a potent circadian oscillator in human epidermis at the population level, hundreds of rhythmically expressed genes, as well as a biomarker set for human epidermis that can, with one sample, highlight circadian phase within a 3-hour time frame. The team concluded that rhythms in human epidermis persist at the population level, and that they were able to present an effective single-sample circadian biomarker.13 This is important, as Morris pointed out, because the standard practice for measuring an individual’s internal clock is to use a dim-light melatonin onset assay over the course of a day.14 In 2019, Jia and associates studied the skin surface lipid profiles of young women to evaluate and characterize circadian human facial surface lipid composition. The investigators identified significant markers of circadian rhythm, with glycerolipids most affected. They ascribed changes in skin barrier function, such as variable pH and transepidermal water loss, to alterations in triacylglycerol levels as well as free fatty acid chain lengths and content that were affected by variations in circadian rhythm.15
 

Sleep and the timing of topicals

Based on their recent review of the literature on circadian rhythm and skin, Lyons and associates argued that an understanding of circadian rhythm helps dermatologists in recommending the optimal times for patients to apply topical medications. They added that urging patients to get sufficient sleep is important because DNA repair of the skin occurs best at that time.16

Conclusions

Doctors have known for half a century that timing drug delivery to a patient’s circadian clock can enhance outcomes. Chronobiological research into how circadian rhythms work at the cellular level, and in cutaneous cells in particular, is a fascinating and expanding area of inquiry that could help dermatologists more accurately recommend timing for skin care regimens. Much more research, especially in clinical trials, is necessary to further elucidate how to best work with the skin’s natural rhythms.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on skin care technologies. Write to her at dermnews@mdedge.com.

References

1. Mehling A et al. Skin Pharmacol Physiol. 2006;19(4):182-9.

2. Latreille J et al. Skin Pharmacol Physiol. 2004 May-Jun;17(3):133-40.

3. Le Fur I et al. J Invest Dermatol. 2001 Sep;117(3):718-24.

4. Verschoore M et al. Chronobiol Int. 1993 Oct;10(5):349-59.

5. Yosipovitch G et al. J Invest Dermatol. 1998 Jan;110(1):20-3.

6. Dong K et al. Int J Cosmet Sci. 2019 Dec;41(6):558-62.

7. Dong K et al. Int J Mol Sci. 2020 Jan 3;21(1):326.

8. Walker WH II et al. Arch Dermatol Res. 2019 Sep;311(7):573-6.

9. Vaughn AR et al. Pediatr Dermatol. 2018 Jan;35(1):152-7.

10. Fishbein AB et al. J Allergy Clin Immunol. 2015 Nov;136(5):1170-7.

11. Plikus MV et al. J Biol Rhythms. 2015 Jun;30(3):163-82.

12. Deshayes N et al. Eur J Dermatol. 2018 Aug 1;28(4):467-75.

13. Wu G et al. Proc Natl Acad Sci U S A. 2018 Nov 27;115(48):12313-8.

14. Morris A. Nat Rev Endocrinol. 2018 Dec;15(1):3.

15. Jia Y et al. Exp Dermatol. 2019 Jul;28(7):858-62.

16. Lyons AB et al. J Clin Aesthet Dermatol. 2019 Sep;12(9):42-5.
 

We continue with a focus on circadian rhythms and implications for skin care this month, paying particular attention to research or insights pertaining to specific skin conditions, when possible, as well as clinical studies that may shed light on how to time skin care treatments.

Vectorial illustration of a clock with human hands, representing the circadian rhythm of sleep and wakefulness. In the balanced cycle, a half circle contains the night and the other one the day.
Francesca Bellini/iStock/Getty Images

It is important to remember that several studies in the last 20 years have revealed cutaneous tendencies based on the time of day. For instance, sebum production is known to be highest around noon, and pH also peaks during the day and is at its lowest at night.1-5
 

Skin aging

In 2019, Dong and associates showed that blue light at 410 nm reduces PER1 transcription in keratinocytes, indicating that epidermal cells have the capacity to directly sense light and regulate their own clock gene expression. With the introduction of blue light at night, circadian rhythm is disrupted as epidermal skin cells act as if it is daytime. The investigators also considered blue light–induced damage to skin cells at various doses and exposure times in comparison with cells that remained unexposed to light. The production of reactive oxygen species increased in the exposed cells, as did DNA impairment and the emergence of inflammatory mediators, all of which have the potential to hasten aging.6

Early this year, Dong and associates demonstrated that melatonin can dose-dependently stimulate PER1 clock gene expression in normal human dermal fibroblasts and normal human epidermal keratinocytes, and verified that the MT-1 melatonin receptor in such fibroblasts manifests a marked decline with age. The researchers concluded that the melatonin pathway contributes significantly in cutaneous aging and impairment, and that its relationship with skin circadian rhythm points to a possible role in slowing the rate of skin aging through the modulation of cutaneous melatonin receptors.7
 

Wound healing

In 2019, Walker and associates investigated the effects of dim artificial light at night on wound healing in female C57BL/6 mice, and found that those conditions prior to wounding reduced healing. They concluded that such information might warrant consideration in prescribing treatment.8

Atopic dermatitis

Vaughn and associates contended that alterations in circadian rhythm may contribute to the development of atopic dermatitis.9 A good example of the impact of circadian rhythms on cutaneous health is the nocturnal exacerbation of atopic dermatitis, particularly in children.10

Psoriasis

According to Plikus and associates, recent evidence has emerged showing that the circadian clock regulates UVB-induced DNA damage and cutaneous cancers, and it is also associated with the immune-mediated disorder psoriasis.11

Clinical studies

In 2018, Deshayes and associates conducted a clinical study to evaluate the precursors and stem cell attributes of hHF (human hair follicle keratinocytes), hEpi (human interfollicular epidermal keratinocytes), and hHFDP (hair follicle dermal papilla stem cells) in response to clock pathway changes caused by long-term deregulation of circadian rhythms. A total of 20 women participated in the study, 10 in each group (day workers were the control group and compared with shift workers). Two 3-mm fresh punch biopsies were collected from the occipital region of each participant. The investigators reported that chronic circadian rhythm deregulation influenced clock pathway protein expression and correlated with changes in hHF, hEpi, and hHFDP. They concluded that their findings represented the first data in humans suggesting that deregulation of the clock pathway modulates regenerative activity in human cutaneous and hair precursor cells.12

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Later that year, Wu and associates reported on the role of the circadian clock in the transcriptional regulation of human epidermis. Investigators sampled 20 human participants through a 24-hour period and a population of 219 people once, finding a potent circadian oscillator in human epidermis at the population level, hundreds of rhythmically expressed genes, as well as a biomarker set for human epidermis that can, with one sample, highlight circadian phase within a 3-hour time frame. The team concluded that rhythms in human epidermis persist at the population level, and that they were able to present an effective single-sample circadian biomarker.13 This is important, as Morris pointed out, because the standard practice for measuring an individual’s internal clock is to use a dim-light melatonin onset assay over the course of a day.14 In 2019, Jia and associates studied the skin surface lipid profiles of young women to evaluate and characterize circadian human facial surface lipid composition. The investigators identified significant markers of circadian rhythm, with glycerolipids most affected. They ascribed changes in skin barrier function, such as variable pH and transepidermal water loss, to alterations in triacylglycerol levels as well as free fatty acid chain lengths and content that were affected by variations in circadian rhythm.15
 

Sleep and the timing of topicals

Based on their recent review of the literature on circadian rhythm and skin, Lyons and associates argued that an understanding of circadian rhythm helps dermatologists in recommending the optimal times for patients to apply topical medications. They added that urging patients to get sufficient sleep is important because DNA repair of the skin occurs best at that time.16

Conclusions

Doctors have known for half a century that timing drug delivery to a patient’s circadian clock can enhance outcomes. Chronobiological research into how circadian rhythms work at the cellular level, and in cutaneous cells in particular, is a fascinating and expanding area of inquiry that could help dermatologists more accurately recommend timing for skin care regimens. Much more research, especially in clinical trials, is necessary to further elucidate how to best work with the skin’s natural rhythms.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on skin care technologies. Write to her at dermnews@mdedge.com.

References

1. Mehling A et al. Skin Pharmacol Physiol. 2006;19(4):182-9.

2. Latreille J et al. Skin Pharmacol Physiol. 2004 May-Jun;17(3):133-40.

3. Le Fur I et al. J Invest Dermatol. 2001 Sep;117(3):718-24.

4. Verschoore M et al. Chronobiol Int. 1993 Oct;10(5):349-59.

5. Yosipovitch G et al. J Invest Dermatol. 1998 Jan;110(1):20-3.

6. Dong K et al. Int J Cosmet Sci. 2019 Dec;41(6):558-62.

7. Dong K et al. Int J Mol Sci. 2020 Jan 3;21(1):326.

8. Walker WH II et al. Arch Dermatol Res. 2019 Sep;311(7):573-6.

9. Vaughn AR et al. Pediatr Dermatol. 2018 Jan;35(1):152-7.

10. Fishbein AB et al. J Allergy Clin Immunol. 2015 Nov;136(5):1170-7.

11. Plikus MV et al. J Biol Rhythms. 2015 Jun;30(3):163-82.

12. Deshayes N et al. Eur J Dermatol. 2018 Aug 1;28(4):467-75.

13. Wu G et al. Proc Natl Acad Sci U S A. 2018 Nov 27;115(48):12313-8.

14. Morris A. Nat Rev Endocrinol. 2018 Dec;15(1):3.

15. Jia Y et al. Exp Dermatol. 2019 Jul;28(7):858-62.

16. Lyons AB et al. J Clin Aesthet Dermatol. 2019 Sep;12(9):42-5.
 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article

Circadian rhythms: Does the time of day you use a skin care product matter?

Article Type
Changed
Thu, 12/24/2020 - 14:02

The majority of human cells, including skin and hair cells, keep their own time; that is, they manifest autonomous clocks and the genes that regulate their functioning.1 During the day, one primary function of the skin is protection; at night, repairing any damage (particularly DNA impairment) incurred during the day prevails.2-4 These activities are driven through circadian rhythms using clock genes that exist in all cutaneous cells.2 Important cutaneous functions such as blood flow, transepidermal water loss, and capacitance are affected by circadian rhythms.5 Hydration and inflammation are also among the several functions pertaining to epidermal homeostasis affected by circadian rhythms.6 In addition, some collagens and extracellular matrix proteases are diurnally regulated, and approximately 10% of the transcriptome, including the extracellular matrix, is thought to be controlled by circadian rhythms.7

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Emerging research on the circadian rhythms displayed in the skin yield implications related to skin care. Cutaneous cell migration and proliferation, wound healing, and tissue vulnerability to harm from UV exposure, oxidative stress, and protease activity, for example, are affected by circadian rhythms, Sherratt et al. noted in suggesting that chronotherapy presents promise for enhancing skin therapy.7 Indeed, recent research has led to the understanding that cutaneous aging, cellular repair, optimal timing for drug delivery to the skin, and skin cancer development are all affected by the chronobiological functioning of the skin.8

We have known for several years that certain types of products should be used at different times of the day. For instance, antioxidants should be used in the morning to protect skin from sun exposure and retinols should be used in the evening because of its induction of light sensitivity. The remainder of this column focuses on research in the last 2 decades that reinforces the notion of circadian rhythms working in the skin, and may alter how we view the timing of skin care. Next month’s column, part two on the circadian rhythms of the skin, will address recent clinical trials and the implications for timing treatments for certain cutaneous conditions.
 

Emerging data on the circadian rhythms of the skin

In 2001, Le Fur et al. studied the cutaneous circadian rhythms in the facial and forearm skin of eight healthy White women during a 48-hour period. They were able to detect such rhythms in facial sebum excretion, transepidermal water loss (TEWL) in the face and forearm, pH in the face, forearm skin temperature, and forearm capacitance using cosinor or analysis of variance methods. The investigators also observed 8- and 12-hour rhythms in TEWL in both areas, and 12 hours for forearm skin temperature. They verified that such rhythms could be measured and that they vary between skin sites. In addition, they were the first to show that ultradian and/or component rhythms can also be found in TEWL, sebum excretion, and skin temperature.9

A year later, Kawara et al. showed that mRNA of the circadian clock genes Per1, Clock, and bmal1/mop3 are expressed in normal human-cultured keratinocytes and that low-dose UVB down-regulates these genes and changes their express in keratinocyte cell cultures. They concluded that UV targeting of keratinocytes could alter circadian rhythms.10

In 2011, Spörl and colleagues characterized an in vitro functional cell autonomous circadian clock in adult human low calcium temperature keratinocytes, demonstrating that the molecular composition of the keratinocyte clock was comparable with peripheral tissue clocks. Notably, they observed that temperature acts as a robust time cue for epidermal traits, such as cholesterol homeostasis and differentiation.11

The next year, Sandu et al. investigated the kinetics of clock gene expression in epidermal and dermal cells collected from the same donor and compared their characteristics. They were able to reveal the presence of functional circadian machinery in primary cultures of fibroblasts, keratinocytes, and melanocytes, with oscillators identified in all skin cell types and thought to be involved in spurring cutaneous rhythmic functions as they exhibited discrete periods and phase relationships between clock genes.12

Three years later, Sandu et al. characterized the circadian clocks in rat skin and dermal fibroblasts. They found that skin has a self-sustaining circadian clock that experiences age-dependent alterations, and that dermal fibroblasts manifest circadian rhythms that can be modulated by endogenous (e.g., melatonin) and exogenous (e.g., temperature) influences.13

In 2019, Park et al. demonstrated that the diurnal expression of the gene TIMP3, which is thought to evince a circadian rhythm in synchronized human keratinocytes, experiences disruptions in such rhythms by UVB exposure. The inflammation that results can be blocked, they argued, by recovering the circadian expression of TIMP3 using synthetic TIMP3 peptides or bioactive natural ingredients, such as green tea extracts.6

Conclusion

Circadian rhythms and the biological clocks by which most cells, including skin and hair cells, regulate themselves represent a ripe and fascinating area of research. Applying evidence in this realm to skin care has been occurring over time and is likely to enhance our practice even more as we continue to elucidate the behavior of cutaneous cells based on the solar day. Based on this information, my recommendations are to use antioxidants and protective products in the morning, and use DNA repair enzymes, retinoids, and other repair products at night.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Dong K et al. Int J Mol Sci. 2020 Jan 3. doi: 10.3390/ijms21010326.

2. Dong K et al. Int J Cosmet Sci. 2019 Dec;41(6):558-62.

3. Lyons AB et al. J Clin Aesthet Dermatol. 2019 Sep;12(9):42-5.

4. Wu G et al. Proc Natl Acad Sci U S A. 2018 Nov 27;115(48):12313-8.

5. Vaughn AR et al. Pediatr Dermatol. 2018 Jan;35(1):152-7.

6. Park S et al. Int J Mol Sci. 2019 Feb 16. doi: 10.3390/ijms20040862.

7. Sherratt MJ et al. Matrix Biol. 2019 Nov;84:97-110.

8. Luber AJ et al. J Drugs Dermatol. 2014 Feb;13(2):130-4.

9. Le Fur I et al. J Invest Dermatol. 2001 Sep;117(3):718-24.

10. Kawara S et al. J Invest Dermatol. 2002 Dec;119(6):1220-3.

11. Spörl F et al. J Invest Dermatol. 2011 Feb;131(2):338-48.

12. Sandu C et al. Cell Mol Life Sci. 2012 Oct;69(19):3329-39.

13. Sandu C et al. Cell Mol Life Sci. 2015 Jun;72(11):2237-48.

Publications
Topics
Sections

The majority of human cells, including skin and hair cells, keep their own time; that is, they manifest autonomous clocks and the genes that regulate their functioning.1 During the day, one primary function of the skin is protection; at night, repairing any damage (particularly DNA impairment) incurred during the day prevails.2-4 These activities are driven through circadian rhythms using clock genes that exist in all cutaneous cells.2 Important cutaneous functions such as blood flow, transepidermal water loss, and capacitance are affected by circadian rhythms.5 Hydration and inflammation are also among the several functions pertaining to epidermal homeostasis affected by circadian rhythms.6 In addition, some collagens and extracellular matrix proteases are diurnally regulated, and approximately 10% of the transcriptome, including the extracellular matrix, is thought to be controlled by circadian rhythms.7

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Emerging research on the circadian rhythms displayed in the skin yield implications related to skin care. Cutaneous cell migration and proliferation, wound healing, and tissue vulnerability to harm from UV exposure, oxidative stress, and protease activity, for example, are affected by circadian rhythms, Sherratt et al. noted in suggesting that chronotherapy presents promise for enhancing skin therapy.7 Indeed, recent research has led to the understanding that cutaneous aging, cellular repair, optimal timing for drug delivery to the skin, and skin cancer development are all affected by the chronobiological functioning of the skin.8

We have known for several years that certain types of products should be used at different times of the day. For instance, antioxidants should be used in the morning to protect skin from sun exposure and retinols should be used in the evening because of its induction of light sensitivity. The remainder of this column focuses on research in the last 2 decades that reinforces the notion of circadian rhythms working in the skin, and may alter how we view the timing of skin care. Next month’s column, part two on the circadian rhythms of the skin, will address recent clinical trials and the implications for timing treatments for certain cutaneous conditions.
 

Emerging data on the circadian rhythms of the skin

In 2001, Le Fur et al. studied the cutaneous circadian rhythms in the facial and forearm skin of eight healthy White women during a 48-hour period. They were able to detect such rhythms in facial sebum excretion, transepidermal water loss (TEWL) in the face and forearm, pH in the face, forearm skin temperature, and forearm capacitance using cosinor or analysis of variance methods. The investigators also observed 8- and 12-hour rhythms in TEWL in both areas, and 12 hours for forearm skin temperature. They verified that such rhythms could be measured and that they vary between skin sites. In addition, they were the first to show that ultradian and/or component rhythms can also be found in TEWL, sebum excretion, and skin temperature.9

A year later, Kawara et al. showed that mRNA of the circadian clock genes Per1, Clock, and bmal1/mop3 are expressed in normal human-cultured keratinocytes and that low-dose UVB down-regulates these genes and changes their express in keratinocyte cell cultures. They concluded that UV targeting of keratinocytes could alter circadian rhythms.10

In 2011, Spörl and colleagues characterized an in vitro functional cell autonomous circadian clock in adult human low calcium temperature keratinocytes, demonstrating that the molecular composition of the keratinocyte clock was comparable with peripheral tissue clocks. Notably, they observed that temperature acts as a robust time cue for epidermal traits, such as cholesterol homeostasis and differentiation.11

The next year, Sandu et al. investigated the kinetics of clock gene expression in epidermal and dermal cells collected from the same donor and compared their characteristics. They were able to reveal the presence of functional circadian machinery in primary cultures of fibroblasts, keratinocytes, and melanocytes, with oscillators identified in all skin cell types and thought to be involved in spurring cutaneous rhythmic functions as they exhibited discrete periods and phase relationships between clock genes.12

Three years later, Sandu et al. characterized the circadian clocks in rat skin and dermal fibroblasts. They found that skin has a self-sustaining circadian clock that experiences age-dependent alterations, and that dermal fibroblasts manifest circadian rhythms that can be modulated by endogenous (e.g., melatonin) and exogenous (e.g., temperature) influences.13

In 2019, Park et al. demonstrated that the diurnal expression of the gene TIMP3, which is thought to evince a circadian rhythm in synchronized human keratinocytes, experiences disruptions in such rhythms by UVB exposure. The inflammation that results can be blocked, they argued, by recovering the circadian expression of TIMP3 using synthetic TIMP3 peptides or bioactive natural ingredients, such as green tea extracts.6

Conclusion

Circadian rhythms and the biological clocks by which most cells, including skin and hair cells, regulate themselves represent a ripe and fascinating area of research. Applying evidence in this realm to skin care has been occurring over time and is likely to enhance our practice even more as we continue to elucidate the behavior of cutaneous cells based on the solar day. Based on this information, my recommendations are to use antioxidants and protective products in the morning, and use DNA repair enzymes, retinoids, and other repair products at night.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Dong K et al. Int J Mol Sci. 2020 Jan 3. doi: 10.3390/ijms21010326.

2. Dong K et al. Int J Cosmet Sci. 2019 Dec;41(6):558-62.

3. Lyons AB et al. J Clin Aesthet Dermatol. 2019 Sep;12(9):42-5.

4. Wu G et al. Proc Natl Acad Sci U S A. 2018 Nov 27;115(48):12313-8.

5. Vaughn AR et al. Pediatr Dermatol. 2018 Jan;35(1):152-7.

6. Park S et al. Int J Mol Sci. 2019 Feb 16. doi: 10.3390/ijms20040862.

7. Sherratt MJ et al. Matrix Biol. 2019 Nov;84:97-110.

8. Luber AJ et al. J Drugs Dermatol. 2014 Feb;13(2):130-4.

9. Le Fur I et al. J Invest Dermatol. 2001 Sep;117(3):718-24.

10. Kawara S et al. J Invest Dermatol. 2002 Dec;119(6):1220-3.

11. Spörl F et al. J Invest Dermatol. 2011 Feb;131(2):338-48.

12. Sandu C et al. Cell Mol Life Sci. 2012 Oct;69(19):3329-39.

13. Sandu C et al. Cell Mol Life Sci. 2015 Jun;72(11):2237-48.

The majority of human cells, including skin and hair cells, keep their own time; that is, they manifest autonomous clocks and the genes that regulate their functioning.1 During the day, one primary function of the skin is protection; at night, repairing any damage (particularly DNA impairment) incurred during the day prevails.2-4 These activities are driven through circadian rhythms using clock genes that exist in all cutaneous cells.2 Important cutaneous functions such as blood flow, transepidermal water loss, and capacitance are affected by circadian rhythms.5 Hydration and inflammation are also among the several functions pertaining to epidermal homeostasis affected by circadian rhythms.6 In addition, some collagens and extracellular matrix proteases are diurnally regulated, and approximately 10% of the transcriptome, including the extracellular matrix, is thought to be controlled by circadian rhythms.7

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Emerging research on the circadian rhythms displayed in the skin yield implications related to skin care. Cutaneous cell migration and proliferation, wound healing, and tissue vulnerability to harm from UV exposure, oxidative stress, and protease activity, for example, are affected by circadian rhythms, Sherratt et al. noted in suggesting that chronotherapy presents promise for enhancing skin therapy.7 Indeed, recent research has led to the understanding that cutaneous aging, cellular repair, optimal timing for drug delivery to the skin, and skin cancer development are all affected by the chronobiological functioning of the skin.8

We have known for several years that certain types of products should be used at different times of the day. For instance, antioxidants should be used in the morning to protect skin from sun exposure and retinols should be used in the evening because of its induction of light sensitivity. The remainder of this column focuses on research in the last 2 decades that reinforces the notion of circadian rhythms working in the skin, and may alter how we view the timing of skin care. Next month’s column, part two on the circadian rhythms of the skin, will address recent clinical trials and the implications for timing treatments for certain cutaneous conditions.
 

Emerging data on the circadian rhythms of the skin

In 2001, Le Fur et al. studied the cutaneous circadian rhythms in the facial and forearm skin of eight healthy White women during a 48-hour period. They were able to detect such rhythms in facial sebum excretion, transepidermal water loss (TEWL) in the face and forearm, pH in the face, forearm skin temperature, and forearm capacitance using cosinor or analysis of variance methods. The investigators also observed 8- and 12-hour rhythms in TEWL in both areas, and 12 hours for forearm skin temperature. They verified that such rhythms could be measured and that they vary between skin sites. In addition, they were the first to show that ultradian and/or component rhythms can also be found in TEWL, sebum excretion, and skin temperature.9

A year later, Kawara et al. showed that mRNA of the circadian clock genes Per1, Clock, and bmal1/mop3 are expressed in normal human-cultured keratinocytes and that low-dose UVB down-regulates these genes and changes their express in keratinocyte cell cultures. They concluded that UV targeting of keratinocytes could alter circadian rhythms.10

In 2011, Spörl and colleagues characterized an in vitro functional cell autonomous circadian clock in adult human low calcium temperature keratinocytes, demonstrating that the molecular composition of the keratinocyte clock was comparable with peripheral tissue clocks. Notably, they observed that temperature acts as a robust time cue for epidermal traits, such as cholesterol homeostasis and differentiation.11

The next year, Sandu et al. investigated the kinetics of clock gene expression in epidermal and dermal cells collected from the same donor and compared their characteristics. They were able to reveal the presence of functional circadian machinery in primary cultures of fibroblasts, keratinocytes, and melanocytes, with oscillators identified in all skin cell types and thought to be involved in spurring cutaneous rhythmic functions as they exhibited discrete periods and phase relationships between clock genes.12

Three years later, Sandu et al. characterized the circadian clocks in rat skin and dermal fibroblasts. They found that skin has a self-sustaining circadian clock that experiences age-dependent alterations, and that dermal fibroblasts manifest circadian rhythms that can be modulated by endogenous (e.g., melatonin) and exogenous (e.g., temperature) influences.13

In 2019, Park et al. demonstrated that the diurnal expression of the gene TIMP3, which is thought to evince a circadian rhythm in synchronized human keratinocytes, experiences disruptions in such rhythms by UVB exposure. The inflammation that results can be blocked, they argued, by recovering the circadian expression of TIMP3 using synthetic TIMP3 peptides or bioactive natural ingredients, such as green tea extracts.6

Conclusion

Circadian rhythms and the biological clocks by which most cells, including skin and hair cells, regulate themselves represent a ripe and fascinating area of research. Applying evidence in this realm to skin care has been occurring over time and is likely to enhance our practice even more as we continue to elucidate the behavior of cutaneous cells based on the solar day. Based on this information, my recommendations are to use antioxidants and protective products in the morning, and use DNA repair enzymes, retinoids, and other repair products at night.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Dong K et al. Int J Mol Sci. 2020 Jan 3. doi: 10.3390/ijms21010326.

2. Dong K et al. Int J Cosmet Sci. 2019 Dec;41(6):558-62.

3. Lyons AB et al. J Clin Aesthet Dermatol. 2019 Sep;12(9):42-5.

4. Wu G et al. Proc Natl Acad Sci U S A. 2018 Nov 27;115(48):12313-8.

5. Vaughn AR et al. Pediatr Dermatol. 2018 Jan;35(1):152-7.

6. Park S et al. Int J Mol Sci. 2019 Feb 16. doi: 10.3390/ijms20040862.

7. Sherratt MJ et al. Matrix Biol. 2019 Nov;84:97-110.

8. Luber AJ et al. J Drugs Dermatol. 2014 Feb;13(2):130-4.

9. Le Fur I et al. J Invest Dermatol. 2001 Sep;117(3):718-24.

10. Kawara S et al. J Invest Dermatol. 2002 Dec;119(6):1220-3.

11. Spörl F et al. J Invest Dermatol. 2011 Feb;131(2):338-48.

12. Sandu C et al. Cell Mol Life Sci. 2012 Oct;69(19):3329-39.

13. Sandu C et al. Cell Mol Life Sci. 2015 Jun;72(11):2237-48.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article

Dermatologists and the history of skin care and beauty devices: Part 4

Article Type
Changed
Thu, 11/19/2020 - 10:12

In this series on the role dermatologists have played in the history of skin care, I have covered dermatologists who developed cosmeceutical ingredients, dermatologists who consulted for the skin care industry, and those who developed a novel and successful skin care line. In this column, part 4 of the series, I will continue to discuss the role that dermatologists have played in developing skin care products and devices used in the skin care and beauty industry.
 

Dermatologists and Stiefel Laboratories

The Stiefel Medicinal Soap Company, founded in 1847, later became Stiefel Laboratories and was sold to GlaxoSmithKline in 2009. Stiefel Laboratories made many contributions over the years to the field of dermatology as chronicled in the excellent book, Skin Saga” written by Charles Stiefel and published in 2018. The company was first known for soaps and groundbreaking products, such as “Freckle Soap” that sped epidermal turnover, resulting in a more even toned complexion.

Courtesy of Dr. Leslie Baumann
The Stiefel Medicinal Soap Company was first known for soaps and groundbreaking products, such as 'Freckle Soap' that sped epidermal turnover, resulting in a more even toned complexion.


Many dermatologists were involved in developing products and providing advice to the company. Herman Sharlit, MD, in New York, had the idea for a moisturizing soap (Oilatum), a detergent soap (Acne Aid detergent soap), and a coal tar soap (Polytar). Eugene Farber, MD, who was professor and chairman of the department of dermatology at Stanford (Calif.) University, consulted for Stiefel Laboratories and helped them identify and develop many products over the years.1 Stiefel Labs came out with the first facial scrub called Brasivol, an abrasive cream with aluminum oxide particles – the predecessor to modern day microdermabrasion. This facial scrub was conceived by dermatologist Rose Saperstein, MD, Los Angeles, who published a report2 on this in 1960 and also received a patent for it in 1963.3 Brasivol became the company’s first million dollar product.1

Stiefel Laboratories worked with many dermatologists to help them develop their ideas. They included Cleveland White, MD, who patented a highly absorbent foot and body powder known as Zeasorb powder. William Pace, MD, was a Canadian dermatologist who patented an acne treatment containing benzoyl peroxide and sulfur that Stiefel Labs marketed as Sulfoxyl Lotion. Dr. Pace is lovingly referred to as “the father of benzoyl peroxide” because his idea led Stiefel Labs to develop more benzoyl peroxide products. Benzoyl peroxide remains the most popular OTC ingredient to treat acne.

Comedone extractors

Many dermatologists have developed ways to extract comedones. There are publications on using paper clips,4,5safety pins,6 and medicine droppers,7 but some dermatologists have developed special comedone extractors, which include the following: Jay Schamberg, MD, developed a comedone extractor with a loop at each end. He disapproved of cutting a comedone, so did not include a needle or scalpel in his extractor.8

  • Leonard Savitt, MD,9 attached a scalpel to one end of the Schamberg extractor.
  • Alan Shalita, MD, developed a comedone extractor with a large, keyhole-shaped extracting orifice that made the tool easier to clean.10

The Saalfield comedone extractor combines a fixed pointed blade at one end and a small spoon-shaped expressor foot at the other end. (However, I have not been able to determine if Saalfield was a dermatologist.)
 

 

 

Dermatologist who developed methods for lesion excisions

Robert Segal, MD, a dermatologist at the University of Arizona, Tucson, invented the Dermablade. Although this is technically not a beauty device, I am including it because it has made the removal of unsightly moles and lesions much easier. He holds six patents on this device.

Dermatologists developed dermabrasion and microneedling

Ernst Kromayer, MD,11 a dermatologist in Germany, first described microneedling in 1905 when he mounted dental burrs on motor-driven flexible cord equipment to treat scars. Abner Kurtin, MD, a New York dermatologist, learned about Dr. Kromayer’s technique and modified it using stainless wireless brushes. Dr. Kurtin is known as the “father of dermabrasion.” His work was noted by Nobel Laureate Alexis Carrel, MD, who moved to New York City and began using the technique. Dr. Carrel’s protege, New York dermatologist, Norman Orentreich, MD, began using hypodermic needles instead of wire brushes. Microneedling has gained much popularity over the last decade and has been combined with platelet rich plasma injections.

Dermatologist-developed injection to shrink fat

Adam Rotunda, MD, was a dermatology resident at the University of California, Los Angeles, when he and his professor Michael Kolodney, MD, PhD, had the idea to develop deoxycholate as an injectable to reduce fat deposits. They filed a patent in 2004, conducted clinical trials, and it worked! In 2009, the patent for deoxycholic acid (ATX-10), marketed as Kybella, was granted. The rights to the drug were purchased by Aestherx, which later became Kythera Biopharmaceuticals. Kybella received Food and Drug Administration approval in 2015, and 6 months later, Kythera was acquired by Allergan.

Development of FDA-approved drugs to improve skin appearance

In 2004, dermatologists Stuart Shanler, MD, and Andrew Ondo, MD, filed a patent for the use of topical oxymetazoline for the treatment of the erythema of rosacea. They published their observations in 2007, noting that oxymetazoline improved facial flushing and erythema.11 Dr. Shanler then teamed up with dermatologist Neal Walker, MD, to form a start-up pharmaceutical company, Vicept Therapeutics, and took this compound through phase 2 clinical trials, while Dr. Shanler filed additional patents on oxymetazoline compositions and their uses. Once they successfully demonstrated the efficacy of topical oxymetazoline for rosacea, Allergan acquired the rights of the drug, successfully completed the phase 3 clinical trials, and Rhofade was approved by the FDA in 2017. It is the only topical drug invented and developed by a dermatologist to receive FDA approval since tretinoin (Renova) was developed by Albert Kligman, MD, and approved by the FDA for the improvement in appearance of fine wrinkling, mottled hyperpigmentation and roughness associated with photodamage in 1992.

The development of lasers

The last dermatologist I will discuss in this history series is R. Rox Anderson, MD, professor of dermatology at Harvard University, and director of the Wellman Center for Photomedicine at Massachusetts General Hospital, Boston. It is impossible to list all his contributions in such a limited space. It would take a book. Building on efforts pioneered by Leon Goldman, MD, Dr. Anderson and his associates pioneered the use of lasers in dermatology and invented the idea of photothermolysis when they filed a patent on using light to remove hair in 1995.Dieter Manstein, MD, PhD,Dr. Anderson and others filed many patents that led to devices such as hair removal lasers, resurfacing lasers, and Fraxel lasers. They also made discoveries related to using cold to shrink fat. One of their inventions is known as CoolSculpting. They were so influential in the development of cosmetic dermatology that it is hard to imagine the field without their contributions.

This concludes my four-part series on the history of dermatologists’ role in the development of the skin care industry. I hope I have not forgotten anyone; if I did, I apologize. I have asked for ideas on Dermchat, Facebook and LinkedIn. Feel free to reach out if I missed one of your contributions. I will be giving lectures on this topic in the future and would be happy to include anyone I missed.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann


As the year 2020 ends, I want to say, Happy 50th Anniversary Dermatology News! I hope you enjoyed this historical series in honor of this anniversary.
 

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions Inc., a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Stiefel, CW. (n.d.). Skin Saga: How a Tiny Family Soap Business Evolved Over Six Generations Into the #1 Dermatology Company in the World. United States: Smart Business Network.

2. Saperstein, RB. Arch Dermatol. 1960 Apr;81:601.

3. Saperstein, RB, and Stiefel, WK (1963). U.S. Patent No. 3,092,111. Washington, DC: U.S. Patent and Trademark Office.

4. George DE et al. J Am Acad Dermatol. 2006 Feb;54(2):326.

5. Cvancara JL, Meffert JJ. J Am Acad Dermatol. 1999 Mar;40(3):477-8.

6. Mukhtar M., Sharma R. Int J Dermatol. 2004 Dec;43(12):967-8.

7. Shellow, H. JAMA. 1951;147(18):1777.

8. Wright CS. Arch Dermatol. 1961;84(3):515.

9. Savitt LE. Arch Dermatol. 1961 Apr;83:660-1.

10. Shalita AR, Harris H. Arch Dermatol. 1972 May;105(5):759-60.

11. Shanler SD, Ondo AL. Arch Dermatol. 2007 Nov;143(11):1369-71.

Publications
Topics
Sections

In this series on the role dermatologists have played in the history of skin care, I have covered dermatologists who developed cosmeceutical ingredients, dermatologists who consulted for the skin care industry, and those who developed a novel and successful skin care line. In this column, part 4 of the series, I will continue to discuss the role that dermatologists have played in developing skin care products and devices used in the skin care and beauty industry.
 

Dermatologists and Stiefel Laboratories

The Stiefel Medicinal Soap Company, founded in 1847, later became Stiefel Laboratories and was sold to GlaxoSmithKline in 2009. Stiefel Laboratories made many contributions over the years to the field of dermatology as chronicled in the excellent book, Skin Saga” written by Charles Stiefel and published in 2018. The company was first known for soaps and groundbreaking products, such as “Freckle Soap” that sped epidermal turnover, resulting in a more even toned complexion.

Courtesy of Dr. Leslie Baumann
The Stiefel Medicinal Soap Company was first known for soaps and groundbreaking products, such as 'Freckle Soap' that sped epidermal turnover, resulting in a more even toned complexion.


Many dermatologists were involved in developing products and providing advice to the company. Herman Sharlit, MD, in New York, had the idea for a moisturizing soap (Oilatum), a detergent soap (Acne Aid detergent soap), and a coal tar soap (Polytar). Eugene Farber, MD, who was professor and chairman of the department of dermatology at Stanford (Calif.) University, consulted for Stiefel Laboratories and helped them identify and develop many products over the years.1 Stiefel Labs came out with the first facial scrub called Brasivol, an abrasive cream with aluminum oxide particles – the predecessor to modern day microdermabrasion. This facial scrub was conceived by dermatologist Rose Saperstein, MD, Los Angeles, who published a report2 on this in 1960 and also received a patent for it in 1963.3 Brasivol became the company’s first million dollar product.1

Stiefel Laboratories worked with many dermatologists to help them develop their ideas. They included Cleveland White, MD, who patented a highly absorbent foot and body powder known as Zeasorb powder. William Pace, MD, was a Canadian dermatologist who patented an acne treatment containing benzoyl peroxide and sulfur that Stiefel Labs marketed as Sulfoxyl Lotion. Dr. Pace is lovingly referred to as “the father of benzoyl peroxide” because his idea led Stiefel Labs to develop more benzoyl peroxide products. Benzoyl peroxide remains the most popular OTC ingredient to treat acne.

Comedone extractors

Many dermatologists have developed ways to extract comedones. There are publications on using paper clips,4,5safety pins,6 and medicine droppers,7 but some dermatologists have developed special comedone extractors, which include the following: Jay Schamberg, MD, developed a comedone extractor with a loop at each end. He disapproved of cutting a comedone, so did not include a needle or scalpel in his extractor.8

  • Leonard Savitt, MD,9 attached a scalpel to one end of the Schamberg extractor.
  • Alan Shalita, MD, developed a comedone extractor with a large, keyhole-shaped extracting orifice that made the tool easier to clean.10

The Saalfield comedone extractor combines a fixed pointed blade at one end and a small spoon-shaped expressor foot at the other end. (However, I have not been able to determine if Saalfield was a dermatologist.)
 

 

 

Dermatologist who developed methods for lesion excisions

Robert Segal, MD, a dermatologist at the University of Arizona, Tucson, invented the Dermablade. Although this is technically not a beauty device, I am including it because it has made the removal of unsightly moles and lesions much easier. He holds six patents on this device.

Dermatologists developed dermabrasion and microneedling

Ernst Kromayer, MD,11 a dermatologist in Germany, first described microneedling in 1905 when he mounted dental burrs on motor-driven flexible cord equipment to treat scars. Abner Kurtin, MD, a New York dermatologist, learned about Dr. Kromayer’s technique and modified it using stainless wireless brushes. Dr. Kurtin is known as the “father of dermabrasion.” His work was noted by Nobel Laureate Alexis Carrel, MD, who moved to New York City and began using the technique. Dr. Carrel’s protege, New York dermatologist, Norman Orentreich, MD, began using hypodermic needles instead of wire brushes. Microneedling has gained much popularity over the last decade and has been combined with platelet rich plasma injections.

Dermatologist-developed injection to shrink fat

Adam Rotunda, MD, was a dermatology resident at the University of California, Los Angeles, when he and his professor Michael Kolodney, MD, PhD, had the idea to develop deoxycholate as an injectable to reduce fat deposits. They filed a patent in 2004, conducted clinical trials, and it worked! In 2009, the patent for deoxycholic acid (ATX-10), marketed as Kybella, was granted. The rights to the drug were purchased by Aestherx, which later became Kythera Biopharmaceuticals. Kybella received Food and Drug Administration approval in 2015, and 6 months later, Kythera was acquired by Allergan.

Development of FDA-approved drugs to improve skin appearance

In 2004, dermatologists Stuart Shanler, MD, and Andrew Ondo, MD, filed a patent for the use of topical oxymetazoline for the treatment of the erythema of rosacea. They published their observations in 2007, noting that oxymetazoline improved facial flushing and erythema.11 Dr. Shanler then teamed up with dermatologist Neal Walker, MD, to form a start-up pharmaceutical company, Vicept Therapeutics, and took this compound through phase 2 clinical trials, while Dr. Shanler filed additional patents on oxymetazoline compositions and their uses. Once they successfully demonstrated the efficacy of topical oxymetazoline for rosacea, Allergan acquired the rights of the drug, successfully completed the phase 3 clinical trials, and Rhofade was approved by the FDA in 2017. It is the only topical drug invented and developed by a dermatologist to receive FDA approval since tretinoin (Renova) was developed by Albert Kligman, MD, and approved by the FDA for the improvement in appearance of fine wrinkling, mottled hyperpigmentation and roughness associated with photodamage in 1992.

The development of lasers

The last dermatologist I will discuss in this history series is R. Rox Anderson, MD, professor of dermatology at Harvard University, and director of the Wellman Center for Photomedicine at Massachusetts General Hospital, Boston. It is impossible to list all his contributions in such a limited space. It would take a book. Building on efforts pioneered by Leon Goldman, MD, Dr. Anderson and his associates pioneered the use of lasers in dermatology and invented the idea of photothermolysis when they filed a patent on using light to remove hair in 1995.Dieter Manstein, MD, PhD,Dr. Anderson and others filed many patents that led to devices such as hair removal lasers, resurfacing lasers, and Fraxel lasers. They also made discoveries related to using cold to shrink fat. One of their inventions is known as CoolSculpting. They were so influential in the development of cosmetic dermatology that it is hard to imagine the field without their contributions.

This concludes my four-part series on the history of dermatologists’ role in the development of the skin care industry. I hope I have not forgotten anyone; if I did, I apologize. I have asked for ideas on Dermchat, Facebook and LinkedIn. Feel free to reach out if I missed one of your contributions. I will be giving lectures on this topic in the future and would be happy to include anyone I missed.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann


As the year 2020 ends, I want to say, Happy 50th Anniversary Dermatology News! I hope you enjoyed this historical series in honor of this anniversary.
 

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions Inc., a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Stiefel, CW. (n.d.). Skin Saga: How a Tiny Family Soap Business Evolved Over Six Generations Into the #1 Dermatology Company in the World. United States: Smart Business Network.

2. Saperstein, RB. Arch Dermatol. 1960 Apr;81:601.

3. Saperstein, RB, and Stiefel, WK (1963). U.S. Patent No. 3,092,111. Washington, DC: U.S. Patent and Trademark Office.

4. George DE et al. J Am Acad Dermatol. 2006 Feb;54(2):326.

5. Cvancara JL, Meffert JJ. J Am Acad Dermatol. 1999 Mar;40(3):477-8.

6. Mukhtar M., Sharma R. Int J Dermatol. 2004 Dec;43(12):967-8.

7. Shellow, H. JAMA. 1951;147(18):1777.

8. Wright CS. Arch Dermatol. 1961;84(3):515.

9. Savitt LE. Arch Dermatol. 1961 Apr;83:660-1.

10. Shalita AR, Harris H. Arch Dermatol. 1972 May;105(5):759-60.

11. Shanler SD, Ondo AL. Arch Dermatol. 2007 Nov;143(11):1369-71.

In this series on the role dermatologists have played in the history of skin care, I have covered dermatologists who developed cosmeceutical ingredients, dermatologists who consulted for the skin care industry, and those who developed a novel and successful skin care line. In this column, part 4 of the series, I will continue to discuss the role that dermatologists have played in developing skin care products and devices used in the skin care and beauty industry.
 

Dermatologists and Stiefel Laboratories

The Stiefel Medicinal Soap Company, founded in 1847, later became Stiefel Laboratories and was sold to GlaxoSmithKline in 2009. Stiefel Laboratories made many contributions over the years to the field of dermatology as chronicled in the excellent book, Skin Saga” written by Charles Stiefel and published in 2018. The company was first known for soaps and groundbreaking products, such as “Freckle Soap” that sped epidermal turnover, resulting in a more even toned complexion.

Courtesy of Dr. Leslie Baumann
The Stiefel Medicinal Soap Company was first known for soaps and groundbreaking products, such as 'Freckle Soap' that sped epidermal turnover, resulting in a more even toned complexion.


Many dermatologists were involved in developing products and providing advice to the company. Herman Sharlit, MD, in New York, had the idea for a moisturizing soap (Oilatum), a detergent soap (Acne Aid detergent soap), and a coal tar soap (Polytar). Eugene Farber, MD, who was professor and chairman of the department of dermatology at Stanford (Calif.) University, consulted for Stiefel Laboratories and helped them identify and develop many products over the years.1 Stiefel Labs came out with the first facial scrub called Brasivol, an abrasive cream with aluminum oxide particles – the predecessor to modern day microdermabrasion. This facial scrub was conceived by dermatologist Rose Saperstein, MD, Los Angeles, who published a report2 on this in 1960 and also received a patent for it in 1963.3 Brasivol became the company’s first million dollar product.1

Stiefel Laboratories worked with many dermatologists to help them develop their ideas. They included Cleveland White, MD, who patented a highly absorbent foot and body powder known as Zeasorb powder. William Pace, MD, was a Canadian dermatologist who patented an acne treatment containing benzoyl peroxide and sulfur that Stiefel Labs marketed as Sulfoxyl Lotion. Dr. Pace is lovingly referred to as “the father of benzoyl peroxide” because his idea led Stiefel Labs to develop more benzoyl peroxide products. Benzoyl peroxide remains the most popular OTC ingredient to treat acne.

Comedone extractors

Many dermatologists have developed ways to extract comedones. There are publications on using paper clips,4,5safety pins,6 and medicine droppers,7 but some dermatologists have developed special comedone extractors, which include the following: Jay Schamberg, MD, developed a comedone extractor with a loop at each end. He disapproved of cutting a comedone, so did not include a needle or scalpel in his extractor.8

  • Leonard Savitt, MD,9 attached a scalpel to one end of the Schamberg extractor.
  • Alan Shalita, MD, developed a comedone extractor with a large, keyhole-shaped extracting orifice that made the tool easier to clean.10

The Saalfield comedone extractor combines a fixed pointed blade at one end and a small spoon-shaped expressor foot at the other end. (However, I have not been able to determine if Saalfield was a dermatologist.)
 

 

 

Dermatologist who developed methods for lesion excisions

Robert Segal, MD, a dermatologist at the University of Arizona, Tucson, invented the Dermablade. Although this is technically not a beauty device, I am including it because it has made the removal of unsightly moles and lesions much easier. He holds six patents on this device.

Dermatologists developed dermabrasion and microneedling

Ernst Kromayer, MD,11 a dermatologist in Germany, first described microneedling in 1905 when he mounted dental burrs on motor-driven flexible cord equipment to treat scars. Abner Kurtin, MD, a New York dermatologist, learned about Dr. Kromayer’s technique and modified it using stainless wireless brushes. Dr. Kurtin is known as the “father of dermabrasion.” His work was noted by Nobel Laureate Alexis Carrel, MD, who moved to New York City and began using the technique. Dr. Carrel’s protege, New York dermatologist, Norman Orentreich, MD, began using hypodermic needles instead of wire brushes. Microneedling has gained much popularity over the last decade and has been combined with platelet rich plasma injections.

Dermatologist-developed injection to shrink fat

Adam Rotunda, MD, was a dermatology resident at the University of California, Los Angeles, when he and his professor Michael Kolodney, MD, PhD, had the idea to develop deoxycholate as an injectable to reduce fat deposits. They filed a patent in 2004, conducted clinical trials, and it worked! In 2009, the patent for deoxycholic acid (ATX-10), marketed as Kybella, was granted. The rights to the drug were purchased by Aestherx, which later became Kythera Biopharmaceuticals. Kybella received Food and Drug Administration approval in 2015, and 6 months later, Kythera was acquired by Allergan.

Development of FDA-approved drugs to improve skin appearance

In 2004, dermatologists Stuart Shanler, MD, and Andrew Ondo, MD, filed a patent for the use of topical oxymetazoline for the treatment of the erythema of rosacea. They published their observations in 2007, noting that oxymetazoline improved facial flushing and erythema.11 Dr. Shanler then teamed up with dermatologist Neal Walker, MD, to form a start-up pharmaceutical company, Vicept Therapeutics, and took this compound through phase 2 clinical trials, while Dr. Shanler filed additional patents on oxymetazoline compositions and their uses. Once they successfully demonstrated the efficacy of topical oxymetazoline for rosacea, Allergan acquired the rights of the drug, successfully completed the phase 3 clinical trials, and Rhofade was approved by the FDA in 2017. It is the only topical drug invented and developed by a dermatologist to receive FDA approval since tretinoin (Renova) was developed by Albert Kligman, MD, and approved by the FDA for the improvement in appearance of fine wrinkling, mottled hyperpigmentation and roughness associated with photodamage in 1992.

The development of lasers

The last dermatologist I will discuss in this history series is R. Rox Anderson, MD, professor of dermatology at Harvard University, and director of the Wellman Center for Photomedicine at Massachusetts General Hospital, Boston. It is impossible to list all his contributions in such a limited space. It would take a book. Building on efforts pioneered by Leon Goldman, MD, Dr. Anderson and his associates pioneered the use of lasers in dermatology and invented the idea of photothermolysis when they filed a patent on using light to remove hair in 1995.Dieter Manstein, MD, PhD,Dr. Anderson and others filed many patents that led to devices such as hair removal lasers, resurfacing lasers, and Fraxel lasers. They also made discoveries related to using cold to shrink fat. One of their inventions is known as CoolSculpting. They were so influential in the development of cosmetic dermatology that it is hard to imagine the field without their contributions.

This concludes my four-part series on the history of dermatologists’ role in the development of the skin care industry. I hope I have not forgotten anyone; if I did, I apologize. I have asked for ideas on Dermchat, Facebook and LinkedIn. Feel free to reach out if I missed one of your contributions. I will be giving lectures on this topic in the future and would be happy to include anyone I missed.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann


As the year 2020 ends, I want to say, Happy 50th Anniversary Dermatology News! I hope you enjoyed this historical series in honor of this anniversary.
 

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions Inc., a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Stiefel, CW. (n.d.). Skin Saga: How a Tiny Family Soap Business Evolved Over Six Generations Into the #1 Dermatology Company in the World. United States: Smart Business Network.

2. Saperstein, RB. Arch Dermatol. 1960 Apr;81:601.

3. Saperstein, RB, and Stiefel, WK (1963). U.S. Patent No. 3,092,111. Washington, DC: U.S. Patent and Trademark Office.

4. George DE et al. J Am Acad Dermatol. 2006 Feb;54(2):326.

5. Cvancara JL, Meffert JJ. J Am Acad Dermatol. 1999 Mar;40(3):477-8.

6. Mukhtar M., Sharma R. Int J Dermatol. 2004 Dec;43(12):967-8.

7. Shellow, H. JAMA. 1951;147(18):1777.

8. Wright CS. Arch Dermatol. 1961;84(3):515.

9. Savitt LE. Arch Dermatol. 1961 Apr;83:660-1.

10. Shalita AR, Harris H. Arch Dermatol. 1972 May;105(5):759-60.

11. Shanler SD, Ondo AL. Arch Dermatol. 2007 Nov;143(11):1369-71.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article

Dermatologists’ role in the development of the skin care industry

Article Type
Changed
Thu, 10/22/2020 - 16:25

This is the third in a series of columns discussing the important roles that dermatologists have played in the skin care industry. This section will discuss those dermatologists who worked behind the scenes with the cosmetic industry, rather than developing their own skin care lines.
 

Norman Orentreich, MD

Dr. Orentreich was a successful New York City dermatologist and the first to perform hair transplants. This new technique brought him fame and notoriety and arguably made him the first “celebrity dermatologist.” (He was also a member of the original advisory board of Dermatology News, at that time Skin & Allergy News, in January 1970.) Dr. Orentreich was a seminal figure in the trend to link the cosmetic industry and dermatology. In August 1967, Vogue magazine1 published an article on him, titled “Can Great Skin be Created?” This popular article caught the attention of Leonard Lauder, of Estée Lauder, who recruited Dr. Orentreich to help create the skin care line Clinique. Clinique was intended to be a brand with a medical look that promoted its products as “allergy tested,” with packaging that has an antiseptic look and beauty counter salespeople wearing white coats.

An interview with Dr. Norman Orentreich in this 1967 issue of Vogue led to the creation of Clinique.

Dr. Orentreich’s input into the development of a skin type–based skin care line was fundamental to the development of this brand. The four-question questionnaire with an iconic plastic lever that customers slide left or right instantly provided them with an assessment of their skin type at the beauty counter, with one of four skin types: Very Dry to Dry Skin (Skin Type 1), Dry Combination (Skin Type 2), Combination Oily (Skin Type 3), and Oily (Skin Type 4).

Although this skin-typing system was not scientifically accurate (there is no scientific definition of combination skin), it was reminiscent of the system developed by cosmetic company tycoon Helena Rubinstein in the 1940s that classified people into four skin types: oily, dry, combination, and sensitive. Clinique became a blockbuster skin care brand and was one of the first developed by a dermatologist – although Dr. Orentreich did not put his name on it.

In 1972, Dr. Orentreich filed a patent2 for an exfoliating pad for the skin that later became known as the “Buf-Puf.” I heard years ago that he got the idea from the machines used to buff the floors in the hospital. The buffing pad had a hole in the center where the machine attached. Dr. Orentreich purportedly thought “I wonder what they do with the cut-out centers?” He looked into this, and subsequently used the centers to create the Buf-Puf. I cannot find a reference for this, but I love this story and hope it’s true. If any readers have any knowledge of this, please let me know, so I can amend my story if it is incorrect.
 

Almay

Almay, an amalgamation of the founders’ names, Alfred and Fanny May Woititz, was the first hypoallergenic brand, established in 1931, and the first to provide hypoallergenic cosmetics, long before Clinique. In addition, the company was the first skin care brand to become available by prescription only (as it was initially), fully disclose all individual ingredients in its products (well before this became mandatory in 1976), provide totally fragrance-free products, develop a hypoallergenic fragrance – and provide patch tests and other materials to physicians to identify contact allergens.

Over 90 years, the company was also the first among skin care brands to do the following:

  • Provide custom formulations to individuals proven to be allergic to a specific ingredient, through their physicians.
  • Perform a full range of premarket safety testing on all products for allergy and irritation, and test all its products for comedogenicity.
  • Formulate cosmetics for use around the eye area (eye shadows and eyeliners) specifically for contact lens wearers.
  • Formulate hypoallergenic regimens for specific skin types in the mass market.
  • Provide a specific cosmetic regimen for acne-prone women, including a silicone-based makeup and active ingredients for treatment in cosmetics and skin care.

I recently interviewed Stanley Levy, MD, who was one of the consultants to Almay, and practices in Chapel Hill, N.C., where he has an academic niche related to skin care formulation and safety. He told me how Almay provided patch test materials to dermatologists to help identify contact dermatitis to cosmetic ingredients, and described Almay’s relationship with the dermatology field as follows: “From the outset, Almay was linked to dermatology. In 1930, a chemist and pharmacist in New York City, Al Woititz, was looking to compound cosmetics for his wife suffering from cosmetic allergies, Fannie May. He enlisted the counsel of the preeminent dermatologic expert in contact dermatitis at the time, Dr. Marion Sulzberger, to suggest ingredients to avoid. [Dr. Sulzberger was also a member of the original Dermatology News editorial advisory board.] Soon, dermatologists around New York City were recommending these formulations. This led to a product line free of the known allergens and a fledgling company trademarked as Almay. For the past 90 years, [the company] has kept a close relationship with dermatologists, well before that was the norm.”

The Almay research overseen by Dr. Levy and others contributed greatly to our understanding of the allergenicity of skin care.
 

Albert Kligman, MD

The turning point for the interface of dermatology with the cosmetic industry was the shift from a safety-based approach (hypoallergenic and noncomedogenic) to an emphasis on efficacy claims in the 1980s. Part of the impetus for this was the Dr. Kligman’s observation that retinoids could improve photoaging.

Dr. Kligman, a well-known dermatologist at the University of Pennsylvania, Philadelphia, showed that retinoids were an effective treatment for acne. For more about this, listen to my interview on the Dermatology Weekly podcast, with James Leyden, MD, about his work at the University of Pennsylvania with Dr. Kligman on the development of oral and topical retinoids. During Dr. Kligman’s research on acne, he noticed that wrinkles improved after treatment with tretinoin, and in 1986, he and Dr. Leyden (and several other authors) published the first article about tretinoin’s use for photoaged skin.3 This led to a double-blind study4 conducted by John J. Voorhees, MD, University of Michigan, Ann Arbor, and coauthors that showed statistically significant improvement of photoaged skin when treated with topical tretinoin. Dr. Voorhees and his group did many more studies on retinoids5,6 and photoaging7 – so many that, at one time, he was (and maybe still is) the most widely published dermatologist in the United States. These studies showed that, not only did prescription tretinoin improve the appearance of wrinkles, but so did over-the-counter retinol.8 Retinoids remain the most efficacious prescription and cosmeceutical ingredients to treat wrinkled skin.

When studies conducted by Dr. Kligman, Dr. Voorhees, and by Barbara Gilcrest, MD, 9,10 showed that retinoids improved wrinkles, a major change in the focus in the skin care industry occurred.



During the same time period, the studies on alpha hydroxy acids by Chérie Ditre, MD, Eugene Van Scott, MD, and colleages11,12; and studies by Sheldon Pinnell, MD, on Vitamin C (see part 1 of this series) all demonstrated the efficacy of cosmetic ingredients on photoaged skin. This triggered a major change in how skin care products were marketed, with an efficacy approach rather than a safety approach.

With the shift from safety (hypoallergenic and noncomedogenic issues) to efficacy claims in the 1980s, and as nondrug active ingredients like retinol were shown to have biologic effects, the lines between the Food and Drug Administration’s definition of a drug versus a cosmetic became blurred. In 1984, Dr. Kligman suggested a new classification for the ingredients that fell in the middle, proposing the term “cosmeceutical” and thus, the concept of a cosmeceutical was introduced. To this day, cosmeceutical is not an official definition and the FDA has yet to deal with it as a quasi-drug category. FDA regulations as to what constitutes a drug versus a cosmetic date back to the 1938 Food, Drug and Cosmetic Act.

Once marketing focused on efficacy, many companies made outrageous claims. During the second half of the 1980s, the FDA issued some warning letters to some companies in an effort to control these claims.

Now efficacy claims abound and we, as dermatologists, should be the experts who back up these claims with scientific data. As the cosmeceutical market has evolved and grown, consumers are bewildered by the myriad of active ingredients being promoted and the number of products in the marketplace. As dermatologic innovation has led to more efficacious active ingredients, our patients look to us as knowledgeable and credible sources of information and for recommendations about the best skin care routines for their skin issues. This is all reflected in the fact that physician-dispensed skin care is becoming the fastest growing segment in this market. It is incumbent upon dermatologists to be knowledgeable and conversant about skin care products and skin care routines, and is particularly true for those of us who sell skin care products in our offices.

Dr. Leslie Baumann

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Burt’s Bees, Evolus, Galderma, and Revance. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Vogue Magazine, 1967 Aug 15. “Can Great Skin be Created?”

2. https://patents.google.com/patent/US3910284.

3. Kligman AM et al. J Am Acad Dermatol. 1986 Oct;15(4 Pt 2):836-59.

4. Weiss JS et al. JAMA. 1988 Jan 22-29;259(4):527-32.

5. Goldfarb MT et al. J Am Acad Dermatol. 1989 Sep;21(3 Pt 2):645-50.

6. Ellis CN et al. J Am Acad Dermatol. 1990 Oct;23(4 Pt 1):629-37.

7. Kang S; Voorhees JJ. J Am Acad Dermatol. 1998 Aug;39(2 Pt 3):S55-61.

8. Kafi R et al. Arch Dermatol. 2007 May;143(5):606-12.

9. Gilchrest BA. J Am Acad Dermatol. 1989 Sep;21(3 Pt 2):610-3.

10. Bhawan J et al. Arch Dermatol. 1991 May;127(5):666-72.

11. Griffin TD et al. J Am Acad Dermatol. 1996 Feb;34(2 Pt 1):196-203.

12. Ditre CM et al. J Am Acad Dermatol. 1996 Feb;34(2 Pt 1):187-95.
 

Publications
Topics
Sections

This is the third in a series of columns discussing the important roles that dermatologists have played in the skin care industry. This section will discuss those dermatologists who worked behind the scenes with the cosmetic industry, rather than developing their own skin care lines.
 

Norman Orentreich, MD

Dr. Orentreich was a successful New York City dermatologist and the first to perform hair transplants. This new technique brought him fame and notoriety and arguably made him the first “celebrity dermatologist.” (He was also a member of the original advisory board of Dermatology News, at that time Skin & Allergy News, in January 1970.) Dr. Orentreich was a seminal figure in the trend to link the cosmetic industry and dermatology. In August 1967, Vogue magazine1 published an article on him, titled “Can Great Skin be Created?” This popular article caught the attention of Leonard Lauder, of Estée Lauder, who recruited Dr. Orentreich to help create the skin care line Clinique. Clinique was intended to be a brand with a medical look that promoted its products as “allergy tested,” with packaging that has an antiseptic look and beauty counter salespeople wearing white coats.

An interview with Dr. Norman Orentreich in this 1967 issue of Vogue led to the creation of Clinique.

Dr. Orentreich’s input into the development of a skin type–based skin care line was fundamental to the development of this brand. The four-question questionnaire with an iconic plastic lever that customers slide left or right instantly provided them with an assessment of their skin type at the beauty counter, with one of four skin types: Very Dry to Dry Skin (Skin Type 1), Dry Combination (Skin Type 2), Combination Oily (Skin Type 3), and Oily (Skin Type 4).

Although this skin-typing system was not scientifically accurate (there is no scientific definition of combination skin), it was reminiscent of the system developed by cosmetic company tycoon Helena Rubinstein in the 1940s that classified people into four skin types: oily, dry, combination, and sensitive. Clinique became a blockbuster skin care brand and was one of the first developed by a dermatologist – although Dr. Orentreich did not put his name on it.

In 1972, Dr. Orentreich filed a patent2 for an exfoliating pad for the skin that later became known as the “Buf-Puf.” I heard years ago that he got the idea from the machines used to buff the floors in the hospital. The buffing pad had a hole in the center where the machine attached. Dr. Orentreich purportedly thought “I wonder what they do with the cut-out centers?” He looked into this, and subsequently used the centers to create the Buf-Puf. I cannot find a reference for this, but I love this story and hope it’s true. If any readers have any knowledge of this, please let me know, so I can amend my story if it is incorrect.
 

Almay

Almay, an amalgamation of the founders’ names, Alfred and Fanny May Woititz, was the first hypoallergenic brand, established in 1931, and the first to provide hypoallergenic cosmetics, long before Clinique. In addition, the company was the first skin care brand to become available by prescription only (as it was initially), fully disclose all individual ingredients in its products (well before this became mandatory in 1976), provide totally fragrance-free products, develop a hypoallergenic fragrance – and provide patch tests and other materials to physicians to identify contact allergens.

Over 90 years, the company was also the first among skin care brands to do the following:

  • Provide custom formulations to individuals proven to be allergic to a specific ingredient, through their physicians.
  • Perform a full range of premarket safety testing on all products for allergy and irritation, and test all its products for comedogenicity.
  • Formulate cosmetics for use around the eye area (eye shadows and eyeliners) specifically for contact lens wearers.
  • Formulate hypoallergenic regimens for specific skin types in the mass market.
  • Provide a specific cosmetic regimen for acne-prone women, including a silicone-based makeup and active ingredients for treatment in cosmetics and skin care.

I recently interviewed Stanley Levy, MD, who was one of the consultants to Almay, and practices in Chapel Hill, N.C., where he has an academic niche related to skin care formulation and safety. He told me how Almay provided patch test materials to dermatologists to help identify contact dermatitis to cosmetic ingredients, and described Almay’s relationship with the dermatology field as follows: “From the outset, Almay was linked to dermatology. In 1930, a chemist and pharmacist in New York City, Al Woititz, was looking to compound cosmetics for his wife suffering from cosmetic allergies, Fannie May. He enlisted the counsel of the preeminent dermatologic expert in contact dermatitis at the time, Dr. Marion Sulzberger, to suggest ingredients to avoid. [Dr. Sulzberger was also a member of the original Dermatology News editorial advisory board.] Soon, dermatologists around New York City were recommending these formulations. This led to a product line free of the known allergens and a fledgling company trademarked as Almay. For the past 90 years, [the company] has kept a close relationship with dermatologists, well before that was the norm.”

The Almay research overseen by Dr. Levy and others contributed greatly to our understanding of the allergenicity of skin care.
 

Albert Kligman, MD

The turning point for the interface of dermatology with the cosmetic industry was the shift from a safety-based approach (hypoallergenic and noncomedogenic) to an emphasis on efficacy claims in the 1980s. Part of the impetus for this was the Dr. Kligman’s observation that retinoids could improve photoaging.

Dr. Kligman, a well-known dermatologist at the University of Pennsylvania, Philadelphia, showed that retinoids were an effective treatment for acne. For more about this, listen to my interview on the Dermatology Weekly podcast, with James Leyden, MD, about his work at the University of Pennsylvania with Dr. Kligman on the development of oral and topical retinoids. During Dr. Kligman’s research on acne, he noticed that wrinkles improved after treatment with tretinoin, and in 1986, he and Dr. Leyden (and several other authors) published the first article about tretinoin’s use for photoaged skin.3 This led to a double-blind study4 conducted by John J. Voorhees, MD, University of Michigan, Ann Arbor, and coauthors that showed statistically significant improvement of photoaged skin when treated with topical tretinoin. Dr. Voorhees and his group did many more studies on retinoids5,6 and photoaging7 – so many that, at one time, he was (and maybe still is) the most widely published dermatologist in the United States. These studies showed that, not only did prescription tretinoin improve the appearance of wrinkles, but so did over-the-counter retinol.8 Retinoids remain the most efficacious prescription and cosmeceutical ingredients to treat wrinkled skin.

When studies conducted by Dr. Kligman, Dr. Voorhees, and by Barbara Gilcrest, MD, 9,10 showed that retinoids improved wrinkles, a major change in the focus in the skin care industry occurred.



During the same time period, the studies on alpha hydroxy acids by Chérie Ditre, MD, Eugene Van Scott, MD, and colleages11,12; and studies by Sheldon Pinnell, MD, on Vitamin C (see part 1 of this series) all demonstrated the efficacy of cosmetic ingredients on photoaged skin. This triggered a major change in how skin care products were marketed, with an efficacy approach rather than a safety approach.

With the shift from safety (hypoallergenic and noncomedogenic issues) to efficacy claims in the 1980s, and as nondrug active ingredients like retinol were shown to have biologic effects, the lines between the Food and Drug Administration’s definition of a drug versus a cosmetic became blurred. In 1984, Dr. Kligman suggested a new classification for the ingredients that fell in the middle, proposing the term “cosmeceutical” and thus, the concept of a cosmeceutical was introduced. To this day, cosmeceutical is not an official definition and the FDA has yet to deal with it as a quasi-drug category. FDA regulations as to what constitutes a drug versus a cosmetic date back to the 1938 Food, Drug and Cosmetic Act.

Once marketing focused on efficacy, many companies made outrageous claims. During the second half of the 1980s, the FDA issued some warning letters to some companies in an effort to control these claims.

Now efficacy claims abound and we, as dermatologists, should be the experts who back up these claims with scientific data. As the cosmeceutical market has evolved and grown, consumers are bewildered by the myriad of active ingredients being promoted and the number of products in the marketplace. As dermatologic innovation has led to more efficacious active ingredients, our patients look to us as knowledgeable and credible sources of information and for recommendations about the best skin care routines for their skin issues. This is all reflected in the fact that physician-dispensed skin care is becoming the fastest growing segment in this market. It is incumbent upon dermatologists to be knowledgeable and conversant about skin care products and skin care routines, and is particularly true for those of us who sell skin care products in our offices.

Dr. Leslie Baumann

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Burt’s Bees, Evolus, Galderma, and Revance. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Vogue Magazine, 1967 Aug 15. “Can Great Skin be Created?”

2. https://patents.google.com/patent/US3910284.

3. Kligman AM et al. J Am Acad Dermatol. 1986 Oct;15(4 Pt 2):836-59.

4. Weiss JS et al. JAMA. 1988 Jan 22-29;259(4):527-32.

5. Goldfarb MT et al. J Am Acad Dermatol. 1989 Sep;21(3 Pt 2):645-50.

6. Ellis CN et al. J Am Acad Dermatol. 1990 Oct;23(4 Pt 1):629-37.

7. Kang S; Voorhees JJ. J Am Acad Dermatol. 1998 Aug;39(2 Pt 3):S55-61.

8. Kafi R et al. Arch Dermatol. 2007 May;143(5):606-12.

9. Gilchrest BA. J Am Acad Dermatol. 1989 Sep;21(3 Pt 2):610-3.

10. Bhawan J et al. Arch Dermatol. 1991 May;127(5):666-72.

11. Griffin TD et al. J Am Acad Dermatol. 1996 Feb;34(2 Pt 1):196-203.

12. Ditre CM et al. J Am Acad Dermatol. 1996 Feb;34(2 Pt 1):187-95.
 

This is the third in a series of columns discussing the important roles that dermatologists have played in the skin care industry. This section will discuss those dermatologists who worked behind the scenes with the cosmetic industry, rather than developing their own skin care lines.
 

Norman Orentreich, MD

Dr. Orentreich was a successful New York City dermatologist and the first to perform hair transplants. This new technique brought him fame and notoriety and arguably made him the first “celebrity dermatologist.” (He was also a member of the original advisory board of Dermatology News, at that time Skin & Allergy News, in January 1970.) Dr. Orentreich was a seminal figure in the trend to link the cosmetic industry and dermatology. In August 1967, Vogue magazine1 published an article on him, titled “Can Great Skin be Created?” This popular article caught the attention of Leonard Lauder, of Estée Lauder, who recruited Dr. Orentreich to help create the skin care line Clinique. Clinique was intended to be a brand with a medical look that promoted its products as “allergy tested,” with packaging that has an antiseptic look and beauty counter salespeople wearing white coats.

An interview with Dr. Norman Orentreich in this 1967 issue of Vogue led to the creation of Clinique.

Dr. Orentreich’s input into the development of a skin type–based skin care line was fundamental to the development of this brand. The four-question questionnaire with an iconic plastic lever that customers slide left or right instantly provided them with an assessment of their skin type at the beauty counter, with one of four skin types: Very Dry to Dry Skin (Skin Type 1), Dry Combination (Skin Type 2), Combination Oily (Skin Type 3), and Oily (Skin Type 4).

Although this skin-typing system was not scientifically accurate (there is no scientific definition of combination skin), it was reminiscent of the system developed by cosmetic company tycoon Helena Rubinstein in the 1940s that classified people into four skin types: oily, dry, combination, and sensitive. Clinique became a blockbuster skin care brand and was one of the first developed by a dermatologist – although Dr. Orentreich did not put his name on it.

In 1972, Dr. Orentreich filed a patent2 for an exfoliating pad for the skin that later became known as the “Buf-Puf.” I heard years ago that he got the idea from the machines used to buff the floors in the hospital. The buffing pad had a hole in the center where the machine attached. Dr. Orentreich purportedly thought “I wonder what they do with the cut-out centers?” He looked into this, and subsequently used the centers to create the Buf-Puf. I cannot find a reference for this, but I love this story and hope it’s true. If any readers have any knowledge of this, please let me know, so I can amend my story if it is incorrect.
 

Almay

Almay, an amalgamation of the founders’ names, Alfred and Fanny May Woititz, was the first hypoallergenic brand, established in 1931, and the first to provide hypoallergenic cosmetics, long before Clinique. In addition, the company was the first skin care brand to become available by prescription only (as it was initially), fully disclose all individual ingredients in its products (well before this became mandatory in 1976), provide totally fragrance-free products, develop a hypoallergenic fragrance – and provide patch tests and other materials to physicians to identify contact allergens.

Over 90 years, the company was also the first among skin care brands to do the following:

  • Provide custom formulations to individuals proven to be allergic to a specific ingredient, through their physicians.
  • Perform a full range of premarket safety testing on all products for allergy and irritation, and test all its products for comedogenicity.
  • Formulate cosmetics for use around the eye area (eye shadows and eyeliners) specifically for contact lens wearers.
  • Formulate hypoallergenic regimens for specific skin types in the mass market.
  • Provide a specific cosmetic regimen for acne-prone women, including a silicone-based makeup and active ingredients for treatment in cosmetics and skin care.

I recently interviewed Stanley Levy, MD, who was one of the consultants to Almay, and practices in Chapel Hill, N.C., where he has an academic niche related to skin care formulation and safety. He told me how Almay provided patch test materials to dermatologists to help identify contact dermatitis to cosmetic ingredients, and described Almay’s relationship with the dermatology field as follows: “From the outset, Almay was linked to dermatology. In 1930, a chemist and pharmacist in New York City, Al Woititz, was looking to compound cosmetics for his wife suffering from cosmetic allergies, Fannie May. He enlisted the counsel of the preeminent dermatologic expert in contact dermatitis at the time, Dr. Marion Sulzberger, to suggest ingredients to avoid. [Dr. Sulzberger was also a member of the original Dermatology News editorial advisory board.] Soon, dermatologists around New York City were recommending these formulations. This led to a product line free of the known allergens and a fledgling company trademarked as Almay. For the past 90 years, [the company] has kept a close relationship with dermatologists, well before that was the norm.”

The Almay research overseen by Dr. Levy and others contributed greatly to our understanding of the allergenicity of skin care.
 

Albert Kligman, MD

The turning point for the interface of dermatology with the cosmetic industry was the shift from a safety-based approach (hypoallergenic and noncomedogenic) to an emphasis on efficacy claims in the 1980s. Part of the impetus for this was the Dr. Kligman’s observation that retinoids could improve photoaging.

Dr. Kligman, a well-known dermatologist at the University of Pennsylvania, Philadelphia, showed that retinoids were an effective treatment for acne. For more about this, listen to my interview on the Dermatology Weekly podcast, with James Leyden, MD, about his work at the University of Pennsylvania with Dr. Kligman on the development of oral and topical retinoids. During Dr. Kligman’s research on acne, he noticed that wrinkles improved after treatment with tretinoin, and in 1986, he and Dr. Leyden (and several other authors) published the first article about tretinoin’s use for photoaged skin.3 This led to a double-blind study4 conducted by John J. Voorhees, MD, University of Michigan, Ann Arbor, and coauthors that showed statistically significant improvement of photoaged skin when treated with topical tretinoin. Dr. Voorhees and his group did many more studies on retinoids5,6 and photoaging7 – so many that, at one time, he was (and maybe still is) the most widely published dermatologist in the United States. These studies showed that, not only did prescription tretinoin improve the appearance of wrinkles, but so did over-the-counter retinol.8 Retinoids remain the most efficacious prescription and cosmeceutical ingredients to treat wrinkled skin.

When studies conducted by Dr. Kligman, Dr. Voorhees, and by Barbara Gilcrest, MD, 9,10 showed that retinoids improved wrinkles, a major change in the focus in the skin care industry occurred.



During the same time period, the studies on alpha hydroxy acids by Chérie Ditre, MD, Eugene Van Scott, MD, and colleages11,12; and studies by Sheldon Pinnell, MD, on Vitamin C (see part 1 of this series) all demonstrated the efficacy of cosmetic ingredients on photoaged skin. This triggered a major change in how skin care products were marketed, with an efficacy approach rather than a safety approach.

With the shift from safety (hypoallergenic and noncomedogenic issues) to efficacy claims in the 1980s, and as nondrug active ingredients like retinol were shown to have biologic effects, the lines between the Food and Drug Administration’s definition of a drug versus a cosmetic became blurred. In 1984, Dr. Kligman suggested a new classification for the ingredients that fell in the middle, proposing the term “cosmeceutical” and thus, the concept of a cosmeceutical was introduced. To this day, cosmeceutical is not an official definition and the FDA has yet to deal with it as a quasi-drug category. FDA regulations as to what constitutes a drug versus a cosmetic date back to the 1938 Food, Drug and Cosmetic Act.

Once marketing focused on efficacy, many companies made outrageous claims. During the second half of the 1980s, the FDA issued some warning letters to some companies in an effort to control these claims.

Now efficacy claims abound and we, as dermatologists, should be the experts who back up these claims with scientific data. As the cosmeceutical market has evolved and grown, consumers are bewildered by the myriad of active ingredients being promoted and the number of products in the marketplace. As dermatologic innovation has led to more efficacious active ingredients, our patients look to us as knowledgeable and credible sources of information and for recommendations about the best skin care routines for their skin issues. This is all reflected in the fact that physician-dispensed skin care is becoming the fastest growing segment in this market. It is incumbent upon dermatologists to be knowledgeable and conversant about skin care products and skin care routines, and is particularly true for those of us who sell skin care products in our offices.

Dr. Leslie Baumann

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Burt’s Bees, Evolus, Galderma, and Revance. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Vogue Magazine, 1967 Aug 15. “Can Great Skin be Created?”

2. https://patents.google.com/patent/US3910284.

3. Kligman AM et al. J Am Acad Dermatol. 1986 Oct;15(4 Pt 2):836-59.

4. Weiss JS et al. JAMA. 1988 Jan 22-29;259(4):527-32.

5. Goldfarb MT et al. J Am Acad Dermatol. 1989 Sep;21(3 Pt 2):645-50.

6. Ellis CN et al. J Am Acad Dermatol. 1990 Oct;23(4 Pt 1):629-37.

7. Kang S; Voorhees JJ. J Am Acad Dermatol. 1998 Aug;39(2 Pt 3):S55-61.

8. Kafi R et al. Arch Dermatol. 2007 May;143(5):606-12.

9. Gilchrest BA. J Am Acad Dermatol. 1989 Sep;21(3 Pt 2):610-3.

10. Bhawan J et al. Arch Dermatol. 1991 May;127(5):666-72.

11. Griffin TD et al. J Am Acad Dermatol. 1996 Feb;34(2 Pt 1):196-203.

12. Ditre CM et al. J Am Acad Dermatol. 1996 Feb;34(2 Pt 1):187-95.
 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article

More on the history of dermatologists and skin care

Article Type
Changed
Fri, 09/25/2020 - 09:53

In this historical portrait, I am focusing mainly on dermatologists who have developed a skin care line, and I am loosely following a chronological order. Thanks to all the readers of Part 1 (The interesting history of dermatologist-developed skin care), who gave me interesting feedback on my Facebook and LinkedIn accounts. There was so much interest that I will write Part 3 next month. Feel free to DM me your ideas on LinkedIn.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

The history of dermatologist-developed skin care continues as more dermatologists become interested in developing a skin care line or retailing skin care in their medical practice. A report in July 2020 showed that physician-dispensed skin care is the largest growing segment of the skin care business with a projected compound annual growth rate of 9.9% from 2020 to 2027. I have not seen national sales numbers since this July report, but we have noticed a large increase in online sales for the doctors using my Skin Type Solutions System. This is most likely because, in a national crisis, the self-care and beauty business segments often see growth. So as you can see, dermatologist-dispensed skin care is becoming a major player in national skin care sales. Let’s get back to the story of how this came to be the case.

Peter Elias, MD. Dr. Elias is professor in the department of dermatology at the University of California, San Francisco. In 1996, Dr. Elias published a landmark paper in the Journal of Investigative Dermatology demonstrating that a 1:1:1 ratio of ceramides, fatty acids, and cholesterol is required to repair a damaged skin barrier. He filed multiple patents for using these lipids in moisturizers as early as 1992. His lipid research has stood the test of time, and this paper is still frequently cited. Dr. Elias has authored over 500 peer reviewed articles on the skin barrier, has edited or coauthored three books on skin barrier science, and developed EpiCeram, a product that utilizes ceramide, the fatty acid linoleic acid, and cholesterol. EpiCeram is the only barrier repair moisturizer approved by the Food and Drug Administration and is available by prescription only.



Kathy Fields, MD, and Katie Rodan, MD. Dr. Fields and Dr. Rodan met at Stanford (Calif.) University. In the 1980s, these entrepreneurial dermatologists realized that patients did not understand the role of preventing acne rather than just treating it. As dermatologists, they knew that a consistent daily routine to prevent acne was much more effective than waiting for an outbreak and spot-treating lesions. They took an already available OTC medication – benzoyl peroxide – and educated consumers through infomercials that they needed to stay ahead of acne instead of waiting for a breakout. Using infomercials to sell skin care, selling skin care kits, and educating patients about the need to prevent acne rather than spot treat it was very unusual at the time. As we all know, reeducating your patients is a huge challenge. Dr. Fields and Dr. Rodan changed consumers thinking in a genius way that continues to resonate today by choosing a brand name to make their point: Proactiv. Their simple 3-step acne kit encouraged patients to be proactive about their acne and encouraged compliance. (Patients love exact skin care steps as demonstrated again by the success of the skin care line from plastic surgeon Suzan Obagi, MD, which became available around 1988).

 

 

Dr. Fields and Dr. Rodan first offered Proactiv to Neutrogena, which turned it down. This early disappointment did not deter them and ended up benefiting them because this gave them the idea to do infomercials. Guthy Renker agreed to market and distribute the product, and the first Proactiv infomercial appeared on TV in 1995. It quickly became popular and is still one of the best selling skin care lines of all time. It’s important to note that the “overnight success” of Proactiv took at least a decade of effort.

White bottles on cloudscape.


Dr. Rodan and Dr. Fields started a new skin care line called Rodan and Fields in 2002, which was sold in department stores. This was at a time when department stores were losing market share of the skin care business, and Dr. Rodan and Dr. Fields wisely relaunched in 2007 using a direct sales model similar to Mary Kay and Avon. Their ability to encourage and motivate their team is apparent in the enthusiasm seen in their sales consultants.

Heather Woolery Lloyd, MD. Dr. Woolery Lloyd got her medical degree at the University of Miami where she also completed her dermatology residency. Her interest in skin of color led to her appointment as director of Ethnic Skin Care at the University of Miami, the country’s first cosmetic ethnic skin care department at a major university. She spent years lecturing around the world on skin of color issues and performing clinical trials before she developed the “Specific Beauty” skin care line for melanin rich skin types. Specific Beauty was acquired by Guthy Renker and is available online. It is the most popular dermatologist developed skin care line for skin of color.

In the late 1980s and early 1990s, other dermatologists threw their hats into the ring and came out with skin care lines – some successful and some not. Unfortunately, many skin care lines at that time were based on “pseudoscience” and exaggerated claims, which fueled the fire of those who felt dermatologists should steer clear of these entrepreneurial pursuits. A debate about the ethics of doctors retailing skin care began, and the controversy led to this 2010 statement by the American Medical Association: “In-office sale of health-related products by physicians presents a financial conflict of interest, risks placing undue pressure on the patient, and threatens to erode patient trust and undermine the primary obligation of physicians to serve the interests of their patients before their own.”

Many dermatologists dropped out of the AMA as a result because they felt the organization was no longer representing dermatologist’s interests. After all, we know skin care science better than anyone, and many dermatologists were insulted by the suggestion that we would place our personal financial gain over the best interests of our patients.

This is a perfect example of how the actions of a few unscrupulous dermatologists can affect the entire specialty. I like to focus on the ethical entrepreneurial dermatologists who made great contributions to the skin care industry based on science, efficacy, and patient education and encourage the ethical among us to provide this science-based information to our patients to protect them from pseudoscience-based opportunists. It is obvious that I believe that dermatologists have a responsibility to provide medical advice on skin care to their patients. If not us, who will do it as ethically as we will? But I plead with those of you out there who are promoting foolish stem cell–containing creams and other impossible technologies to remember that you are hurting the credibility of our entire dermatology profession.

In my next column, I will discuss dermatologists who have played a significant role behind the scenes in the development of the skin care industry.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.  

 

References


Castanedo-Tardan MP, Baumann L. Clin Dermatol. Jul-Aug 2009;27(4):355-8.

Gormley DE. Arch Dermatol. 1999 Jul;135(7):765-6.

Miller RC. Arch Dermatol. 1999 Mar;135(3):255-6.

Virtual Mentor. 2010;12(12):925-7.
 

Publications
Topics
Sections

In this historical portrait, I am focusing mainly on dermatologists who have developed a skin care line, and I am loosely following a chronological order. Thanks to all the readers of Part 1 (The interesting history of dermatologist-developed skin care), who gave me interesting feedback on my Facebook and LinkedIn accounts. There was so much interest that I will write Part 3 next month. Feel free to DM me your ideas on LinkedIn.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

The history of dermatologist-developed skin care continues as more dermatologists become interested in developing a skin care line or retailing skin care in their medical practice. A report in July 2020 showed that physician-dispensed skin care is the largest growing segment of the skin care business with a projected compound annual growth rate of 9.9% from 2020 to 2027. I have not seen national sales numbers since this July report, but we have noticed a large increase in online sales for the doctors using my Skin Type Solutions System. This is most likely because, in a national crisis, the self-care and beauty business segments often see growth. So as you can see, dermatologist-dispensed skin care is becoming a major player in national skin care sales. Let’s get back to the story of how this came to be the case.

Peter Elias, MD. Dr. Elias is professor in the department of dermatology at the University of California, San Francisco. In 1996, Dr. Elias published a landmark paper in the Journal of Investigative Dermatology demonstrating that a 1:1:1 ratio of ceramides, fatty acids, and cholesterol is required to repair a damaged skin barrier. He filed multiple patents for using these lipids in moisturizers as early as 1992. His lipid research has stood the test of time, and this paper is still frequently cited. Dr. Elias has authored over 500 peer reviewed articles on the skin barrier, has edited or coauthored three books on skin barrier science, and developed EpiCeram, a product that utilizes ceramide, the fatty acid linoleic acid, and cholesterol. EpiCeram is the only barrier repair moisturizer approved by the Food and Drug Administration and is available by prescription only.



Kathy Fields, MD, and Katie Rodan, MD. Dr. Fields and Dr. Rodan met at Stanford (Calif.) University. In the 1980s, these entrepreneurial dermatologists realized that patients did not understand the role of preventing acne rather than just treating it. As dermatologists, they knew that a consistent daily routine to prevent acne was much more effective than waiting for an outbreak and spot-treating lesions. They took an already available OTC medication – benzoyl peroxide – and educated consumers through infomercials that they needed to stay ahead of acne instead of waiting for a breakout. Using infomercials to sell skin care, selling skin care kits, and educating patients about the need to prevent acne rather than spot treat it was very unusual at the time. As we all know, reeducating your patients is a huge challenge. Dr. Fields and Dr. Rodan changed consumers thinking in a genius way that continues to resonate today by choosing a brand name to make their point: Proactiv. Their simple 3-step acne kit encouraged patients to be proactive about their acne and encouraged compliance. (Patients love exact skin care steps as demonstrated again by the success of the skin care line from plastic surgeon Suzan Obagi, MD, which became available around 1988).

 

 

Dr. Fields and Dr. Rodan first offered Proactiv to Neutrogena, which turned it down. This early disappointment did not deter them and ended up benefiting them because this gave them the idea to do infomercials. Guthy Renker agreed to market and distribute the product, and the first Proactiv infomercial appeared on TV in 1995. It quickly became popular and is still one of the best selling skin care lines of all time. It’s important to note that the “overnight success” of Proactiv took at least a decade of effort.

White bottles on cloudscape.


Dr. Rodan and Dr. Fields started a new skin care line called Rodan and Fields in 2002, which was sold in department stores. This was at a time when department stores were losing market share of the skin care business, and Dr. Rodan and Dr. Fields wisely relaunched in 2007 using a direct sales model similar to Mary Kay and Avon. Their ability to encourage and motivate their team is apparent in the enthusiasm seen in their sales consultants.

Heather Woolery Lloyd, MD. Dr. Woolery Lloyd got her medical degree at the University of Miami where she also completed her dermatology residency. Her interest in skin of color led to her appointment as director of Ethnic Skin Care at the University of Miami, the country’s first cosmetic ethnic skin care department at a major university. She spent years lecturing around the world on skin of color issues and performing clinical trials before she developed the “Specific Beauty” skin care line for melanin rich skin types. Specific Beauty was acquired by Guthy Renker and is available online. It is the most popular dermatologist developed skin care line for skin of color.

In the late 1980s and early 1990s, other dermatologists threw their hats into the ring and came out with skin care lines – some successful and some not. Unfortunately, many skin care lines at that time were based on “pseudoscience” and exaggerated claims, which fueled the fire of those who felt dermatologists should steer clear of these entrepreneurial pursuits. A debate about the ethics of doctors retailing skin care began, and the controversy led to this 2010 statement by the American Medical Association: “In-office sale of health-related products by physicians presents a financial conflict of interest, risks placing undue pressure on the patient, and threatens to erode patient trust and undermine the primary obligation of physicians to serve the interests of their patients before their own.”

Many dermatologists dropped out of the AMA as a result because they felt the organization was no longer representing dermatologist’s interests. After all, we know skin care science better than anyone, and many dermatologists were insulted by the suggestion that we would place our personal financial gain over the best interests of our patients.

This is a perfect example of how the actions of a few unscrupulous dermatologists can affect the entire specialty. I like to focus on the ethical entrepreneurial dermatologists who made great contributions to the skin care industry based on science, efficacy, and patient education and encourage the ethical among us to provide this science-based information to our patients to protect them from pseudoscience-based opportunists. It is obvious that I believe that dermatologists have a responsibility to provide medical advice on skin care to their patients. If not us, who will do it as ethically as we will? But I plead with those of you out there who are promoting foolish stem cell–containing creams and other impossible technologies to remember that you are hurting the credibility of our entire dermatology profession.

In my next column, I will discuss dermatologists who have played a significant role behind the scenes in the development of the skin care industry.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.  

 

References


Castanedo-Tardan MP, Baumann L. Clin Dermatol. Jul-Aug 2009;27(4):355-8.

Gormley DE. Arch Dermatol. 1999 Jul;135(7):765-6.

Miller RC. Arch Dermatol. 1999 Mar;135(3):255-6.

Virtual Mentor. 2010;12(12):925-7.
 

In this historical portrait, I am focusing mainly on dermatologists who have developed a skin care line, and I am loosely following a chronological order. Thanks to all the readers of Part 1 (The interesting history of dermatologist-developed skin care), who gave me interesting feedback on my Facebook and LinkedIn accounts. There was so much interest that I will write Part 3 next month. Feel free to DM me your ideas on LinkedIn.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

The history of dermatologist-developed skin care continues as more dermatologists become interested in developing a skin care line or retailing skin care in their medical practice. A report in July 2020 showed that physician-dispensed skin care is the largest growing segment of the skin care business with a projected compound annual growth rate of 9.9% from 2020 to 2027. I have not seen national sales numbers since this July report, but we have noticed a large increase in online sales for the doctors using my Skin Type Solutions System. This is most likely because, in a national crisis, the self-care and beauty business segments often see growth. So as you can see, dermatologist-dispensed skin care is becoming a major player in national skin care sales. Let’s get back to the story of how this came to be the case.

Peter Elias, MD. Dr. Elias is professor in the department of dermatology at the University of California, San Francisco. In 1996, Dr. Elias published a landmark paper in the Journal of Investigative Dermatology demonstrating that a 1:1:1 ratio of ceramides, fatty acids, and cholesterol is required to repair a damaged skin barrier. He filed multiple patents for using these lipids in moisturizers as early as 1992. His lipid research has stood the test of time, and this paper is still frequently cited. Dr. Elias has authored over 500 peer reviewed articles on the skin barrier, has edited or coauthored three books on skin barrier science, and developed EpiCeram, a product that utilizes ceramide, the fatty acid linoleic acid, and cholesterol. EpiCeram is the only barrier repair moisturizer approved by the Food and Drug Administration and is available by prescription only.



Kathy Fields, MD, and Katie Rodan, MD. Dr. Fields and Dr. Rodan met at Stanford (Calif.) University. In the 1980s, these entrepreneurial dermatologists realized that patients did not understand the role of preventing acne rather than just treating it. As dermatologists, they knew that a consistent daily routine to prevent acne was much more effective than waiting for an outbreak and spot-treating lesions. They took an already available OTC medication – benzoyl peroxide – and educated consumers through infomercials that they needed to stay ahead of acne instead of waiting for a breakout. Using infomercials to sell skin care, selling skin care kits, and educating patients about the need to prevent acne rather than spot treat it was very unusual at the time. As we all know, reeducating your patients is a huge challenge. Dr. Fields and Dr. Rodan changed consumers thinking in a genius way that continues to resonate today by choosing a brand name to make their point: Proactiv. Their simple 3-step acne kit encouraged patients to be proactive about their acne and encouraged compliance. (Patients love exact skin care steps as demonstrated again by the success of the skin care line from plastic surgeon Suzan Obagi, MD, which became available around 1988).

 

 

Dr. Fields and Dr. Rodan first offered Proactiv to Neutrogena, which turned it down. This early disappointment did not deter them and ended up benefiting them because this gave them the idea to do infomercials. Guthy Renker agreed to market and distribute the product, and the first Proactiv infomercial appeared on TV in 1995. It quickly became popular and is still one of the best selling skin care lines of all time. It’s important to note that the “overnight success” of Proactiv took at least a decade of effort.

White bottles on cloudscape.


Dr. Rodan and Dr. Fields started a new skin care line called Rodan and Fields in 2002, which was sold in department stores. This was at a time when department stores were losing market share of the skin care business, and Dr. Rodan and Dr. Fields wisely relaunched in 2007 using a direct sales model similar to Mary Kay and Avon. Their ability to encourage and motivate their team is apparent in the enthusiasm seen in their sales consultants.

Heather Woolery Lloyd, MD. Dr. Woolery Lloyd got her medical degree at the University of Miami where she also completed her dermatology residency. Her interest in skin of color led to her appointment as director of Ethnic Skin Care at the University of Miami, the country’s first cosmetic ethnic skin care department at a major university. She spent years lecturing around the world on skin of color issues and performing clinical trials before she developed the “Specific Beauty” skin care line for melanin rich skin types. Specific Beauty was acquired by Guthy Renker and is available online. It is the most popular dermatologist developed skin care line for skin of color.

In the late 1980s and early 1990s, other dermatologists threw their hats into the ring and came out with skin care lines – some successful and some not. Unfortunately, many skin care lines at that time were based on “pseudoscience” and exaggerated claims, which fueled the fire of those who felt dermatologists should steer clear of these entrepreneurial pursuits. A debate about the ethics of doctors retailing skin care began, and the controversy led to this 2010 statement by the American Medical Association: “In-office sale of health-related products by physicians presents a financial conflict of interest, risks placing undue pressure on the patient, and threatens to erode patient trust and undermine the primary obligation of physicians to serve the interests of their patients before their own.”

Many dermatologists dropped out of the AMA as a result because they felt the organization was no longer representing dermatologist’s interests. After all, we know skin care science better than anyone, and many dermatologists were insulted by the suggestion that we would place our personal financial gain over the best interests of our patients.

This is a perfect example of how the actions of a few unscrupulous dermatologists can affect the entire specialty. I like to focus on the ethical entrepreneurial dermatologists who made great contributions to the skin care industry based on science, efficacy, and patient education and encourage the ethical among us to provide this science-based information to our patients to protect them from pseudoscience-based opportunists. It is obvious that I believe that dermatologists have a responsibility to provide medical advice on skin care to their patients. If not us, who will do it as ethically as we will? But I plead with those of you out there who are promoting foolish stem cell–containing creams and other impossible technologies to remember that you are hurting the credibility of our entire dermatology profession.

In my next column, I will discuss dermatologists who have played a significant role behind the scenes in the development of the skin care industry.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Galderma, Revance, Evolus, and Burt’s Bees. She is the CEO of Skin Type Solutions, a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.  

 

References


Castanedo-Tardan MP, Baumann L. Clin Dermatol. Jul-Aug 2009;27(4):355-8.

Gormley DE. Arch Dermatol. 1999 Jul;135(7):765-6.

Miller RC. Arch Dermatol. 1999 Mar;135(3):255-6.

Virtual Mentor. 2010;12(12):925-7.
 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article

The interesting history of dermatologist-developed skin care

Article Type
Changed
Tue, 06/01/2021 - 17:54

Those of you who have visited my dermatology practice in Miami know that the art in my office is dedicated to the history of the skin care industry. I collect vintage ads, and vintage skin care and personal care products, and biographies of anyone involved in skin care. I can’t get enough of the history of cosmetics, and I have written this historical column in honor of the 50th anniversary of Dermatology News.

Dr. Leslie S. Baumann in her office, in front of a Camay soap ad from the 1920s.
Courtesy Dr. Leslie S. Baumann
Dr. Leslie S. Baumann stands in her office, in front of a Camay soap ad from the 1920s.

The first doctor to market his own cosmetic product, Erasmus Wilson, MD, faced scrutiny from his colleagues. Although he had contributed much to the field of dermatology, he was criticized by other dermatologists when he promoted a hair wash. The next doctor in my story, William Pusey, MD, was criticized for helping the company that manufactured Camay soap because he allowed his name to be used in Camay advertisements. The scrutiny that these two well-respected dermatologists endured from their colleagues deterred dermatologists from entering the skin care business for decades. The professional jealousy from dermatologic colleagues left the skin care field wide open for imposters, charlatans, and nondermatologists who had no concern for efficacy and patient outcomes to flourish. This is the story of a group of brilliant entrepreneurial dermatologists and one chiropractor who misrepresented himself as a dermatologist and how they influenced skin care as we know it.

Erasmus Wilson, MD1 (1809-1884): In 1840, Erasmus Wilson2 was a physician in London who chose to specialize in dermatology at a time when that specialization was frowned upon. He was a subeditor for The Lancet and wrote several books on dermatology including “Diseases of the Skin – A Practical and Theoretical Treatise,” “Portraits of the Diseases of the Skin,” and “Student’s Book on Diseases of the Skin.” He was the first professor of dermatology in the College of Surgeons and by 1869, was the leading English-speaking dermatologist in the world. He contributed much to dermatology, including his pioneering characterizations of Demodex mites, lichen planus, exfoliative dermatitis, neurotic excoriations, and roseola. Dr. Wilson was knighted in 1881 for his good works and notable generosity. (He was known for giving his poor patients money for food, endowing chairs in dermatology, and donating a famous obelisk in London).

Courtesy of Dr. Leslie S. Baumann
An advertisement in the September 1929 Ladies' Home Journal says that Camay soap has the 'unanimous approval' of '72 of the most eminent dermatologists in America.'


In 1854, Dr. Wilson wrote a book for laypeople called “Healthy Skin: A Popular Treatise on the Skin and Hair, Their Preservation and Management,” in which he advocated cleanliness and bathing, which led to the popularity of Turkish baths and bathing resorts in Europe. Despite his undeniable contributions to dermatology, he was widely criticized by his colleagues for promoting a “Hair Wash” and a turtle oil soap. I cannot find any information about whether or not he developed the hair wash and turtle soap himself, but it seems that he earned income from sales of these two products, even though he was said to have donated it all to charities.

William A. Pusey MD (1865-1940): Dr. Pusey was the first chairman of dermatology at the University of Illinois College of Medicine, Chicago. He published several books, including “Care of the Skin and Hair,” “Syphilis as a Modern Problem,” “The Principles and Practices of Dermatology,” and “History of Dermatology” among others. He is best known for his work in developing the use of x-rays (roentgen rays) and phototherapy in dermatology, and in 1907, he was the first dermatologist to describe the use of solid carbon dioxide to treat skin lesions. He was president of the American Dermatological Association in 1910, president of the Chicago Medical Society in 1918, editor of the Archives of Dermatology in 1920, and president of the American Medical Association in 1924.

Courtesy of Dr. Leslie S. Baumann


In the early 1920s, skin care companies were beginning to advertise their products using endorsements from celebrities and socialites, and were making misleading claims. Dr. Pusey wanted to work with these companies to help them perform evidence-based trials so they could make scientifically correct claims. Proctor & Gamble asked Dr. Pusey to advise them on how they could advertise honestly about their new soap, “Camay.” In Dr. Pusey’s words,3 “they (Proctor & Gamble) wanted to give the public authoritative advice about the use of soap and water. They suggested that I get a group of dermatologists of my selection to examine the soap and prepare instructions for bathing and the use of soap, and, if they found this soap was of high quality, to certify to that effect.” The research was performed as he suggested, and he allowed his name to be used in the Camay soap ads from 1926 to 1929. He said that he allowed them to use his name hoping to promote the need for evidence-based research, in contrast to the skin care products endorsed by socialites and celebrities that were flooding the market around that time.

Herbert Rattner, MD, at Northwestern University, Chicago, was his friend and one of the many dermatologists who criticized Dr. Pusey for allowing his name to be used in the Camay ads. Dr. Pusey’s reply to the criticism (according to Dr. Rattner) was that Proctor & Gamble was “proposing to do what the medical profession always is criticizing commercial concerns for not doing, namely, coming to physicians for information on medical matters. Could the profession hope to have any influence with business concerns if it was always eager to criticize bad commercial practices but never willing to support good ones?”3

While Dr. Pusey felt his reasons for adding his name to the Camay ads and research were justified, many of his friends stated that in hindsight, he regretted the action because of the negative response of his colleagues. It was years before dermatologists began providing input again into the skin care industry. During that time, radio, television and print ads were rampant with misleading claims – which led the way for a dermatologic imposter to make a fortune on skin care.

John Woodbury (1851-1909): John Woodbury, a chiropractor, never went to medical school, but that did not stop him from claiming he was a dermatologist and cosmetic surgeon. In 1889, he opened the John H. Woodbury Dermatological Institute in New York City, and over the next few years, opened Woodbury Dermatological Institutes in at least 5 states and employed 25 “physicians” who were not licensed to practice medicine. He came out with face soaps, tonics, and cold creams and spent a fortune on advertising these products and his institutes. In 1901, he sold his “Woodbury Soap” to the Andrew Jergens Company for $212,500 and 10% in royalties.

Multiple lawsuits occurred from 1898 to 1907 because he continued using the Woodbury name on his own products, despite having sold the “Woodbury” trademark to Jergens. He was sued for practicing medicine without a medical license and claiming to be a dermatologist when he was not. He lost most of these lawsuits, including one in 1907 in which the court ruled that corporations may not employ unlicensed professionals to practice medicine. In 1909, John Woodbury committed suicide. The Woodbury Soap company flourished in the 1930s and 1940s, as part of Jergens, until the brand was discontinued in 1970 when Jergens was acquired by American Brands.

1916 Ladies' Home Journal vol 33#9
A 1916 advertisement in Ladies' Home Journal: John Woodbury sold the Woodbury trademark to Jergens in 1901.


The next dermatologists to come along did not make the same mistakes as those of their predecessors. They all made scientific discoveries through their basic science research in the laboratory, filed patents, formed skin care companies, perfected the formulations, and conducted research trials of the final product. Their marketing focused on science and efficacy and only rarely used their names and images in advertising, allowing them to maintain their reputations in the dermatology field.

Eugene Van Scott, MD (1922-present): Dermatologist Dr. Van Scott and dermatopharmacologist Ruey Yu, PhD, filed a method patent in the early 1970s on the effectiveness of alpha hydroxy acids to treat ichthyosis. They invented the abbreviation “AHA” and have continued their work on organic acids to this day. They now have more than 125 patents, which they have licensed to 60 companies in the cosmetics and pharmaceutical industries.

In 1988, 14 years after their initial publication, they founded the company they named Polystrata, which grew into today’s NeoStrata.4 Over the years, they had to defend their patents because many personal care companies used their technologies without licensing them. In 2007, they won a $41 million settlement in a patent infringement suit against Mary Kay filed in March 2005. They have both been very philanthropic in the dermatology world5 and are highly respected in the field. Among many other honors, Dr. Van Scott was named a Master Dermatologist by the American Academy of Dermatology in 1998 and received the Dermatology Foundation’s Distinguished Service Medallion in 2004.

Sheldon Pinnell, MD (1937-2013): After Dr. Pinnell completed his dermatology residency at Harvard Medical School, he spent 2 years studying collagen chemistry at the Max Planck Institute in Munich, Germany. In 1973, he returned to Duke University where he had earned his undergraduate degree before attending Yale University. He remained at Duke for the duration of his career and was professor and chief of dermatology there for many years. Early in his career, he focused on the role of vitamin C in collagen biosynthesis and discovered some of the mechanisms by which sun exposure causes photoaging. He described the use of the first (and most popular) topically applied L-ascorbic acid (vitamin C) to prevent and treat skin aging.

Dr. Pinnell’s many discoveries include showing that the addition of ascorbic acid to fibroblast cultures increases collagen production and that topically applied L-ascorbic acid penetrates into the skin best at a pH of 2-2.5. Dr. Pinnell changed the way the world uses topical antioxidants today; he was widely respected and was a member of the American Dermatological Association and an honorary member of the Society of Investigative Dermatology. He published more than 200 scientific articles and held 10 patents. He started the skin care company Skinceuticals, based on his antioxidant technologies. It was acquired by L’Oreal in 2005.

Richard Fitzpatrick, MD (1944-2014): The dermatologist affectionately known as “Fitz” is credited with being the first to use lasers for skin resurfacing. He went to medical school at Emory University and did his dermatology residency at the University of California, Los Angeles. He authored more than 130 publications and was one of the first doctors to specialize in cosmetic dermatology. He realized that fibroblast cell cultures used to produce the collagen filler CosmoPlast (no longer on the market) generated many growth factors that could rejuvenate the skin, and in 1999, he launched the skin care brand SkinMedica. In 2000, he received a patent for fibroblast-derived growth factors used topically for antiaging – a formula he called Tissue Nutrient Solution. In 2001, the popular product TNS Recovery Complex was launched based on the patented growth factor technology. It is still the most popular growth factor technology on the market.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

What can we learn from these pioneers? I have had several interesting discussions about this topic with Leonard Hoenig, MD, section editor for Reflections on Dermatology: Past, Present, and Future, in Clinics in Dermatology. (Dr. Hoenig told me the interesting story that Listerine mouthwash was named in honor of Joseph Lister but accounts vary as to whether he gave permission to do so. This makes Dr. Lister the most famous physician to endorse a personal care product.) When Dr. Hoenig and I discussed the ethics of dermatologists creating a skin care line or retailing skin care in their medical practice, he stated my sentiments perfectly: “We should rely on professional, ethical, and legal guidelines to help us do what is right. Most importantly, we should have the best interests of our patients at heart when recommending any treatments.”

Dermatologists have unique knowledge, experience, and perspective on treating the skin with topical agents and have the true desire to improve skin health. If we do not discover, research, patent, and develop efficacious skin care products, someone else will do it – and I do not think they will do it as well as a dermatologist can.
 

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan and Burt’s Bees. She is the CEO of Skin Type Solutions Inc., a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Everett MA. Int J Dermatol. 1978 May;17(4):345-52.

2. Moxon RK. N Engl J Med. 1976 Apr 1;294(14):762-4.

3. Rattner H. Arch Derm Syphilol. 1937;35(1):25-66.

4. Neostrata: More than Hope, by Elaine Strauss, U.S. 1 Newspaper, Feb. 24, 1999.

5. Two legends in the field of dermatology provide $1 million gift to Temple University school of medicine’s department of dermatology, Temple University, June 5, 2015.

Publications
Topics
Sections

Those of you who have visited my dermatology practice in Miami know that the art in my office is dedicated to the history of the skin care industry. I collect vintage ads, and vintage skin care and personal care products, and biographies of anyone involved in skin care. I can’t get enough of the history of cosmetics, and I have written this historical column in honor of the 50th anniversary of Dermatology News.

Dr. Leslie S. Baumann in her office, in front of a Camay soap ad from the 1920s.
Courtesy Dr. Leslie S. Baumann
Dr. Leslie S. Baumann stands in her office, in front of a Camay soap ad from the 1920s.

The first doctor to market his own cosmetic product, Erasmus Wilson, MD, faced scrutiny from his colleagues. Although he had contributed much to the field of dermatology, he was criticized by other dermatologists when he promoted a hair wash. The next doctor in my story, William Pusey, MD, was criticized for helping the company that manufactured Camay soap because he allowed his name to be used in Camay advertisements. The scrutiny that these two well-respected dermatologists endured from their colleagues deterred dermatologists from entering the skin care business for decades. The professional jealousy from dermatologic colleagues left the skin care field wide open for imposters, charlatans, and nondermatologists who had no concern for efficacy and patient outcomes to flourish. This is the story of a group of brilliant entrepreneurial dermatologists and one chiropractor who misrepresented himself as a dermatologist and how they influenced skin care as we know it.

Erasmus Wilson, MD1 (1809-1884): In 1840, Erasmus Wilson2 was a physician in London who chose to specialize in dermatology at a time when that specialization was frowned upon. He was a subeditor for The Lancet and wrote several books on dermatology including “Diseases of the Skin – A Practical and Theoretical Treatise,” “Portraits of the Diseases of the Skin,” and “Student’s Book on Diseases of the Skin.” He was the first professor of dermatology in the College of Surgeons and by 1869, was the leading English-speaking dermatologist in the world. He contributed much to dermatology, including his pioneering characterizations of Demodex mites, lichen planus, exfoliative dermatitis, neurotic excoriations, and roseola. Dr. Wilson was knighted in 1881 for his good works and notable generosity. (He was known for giving his poor patients money for food, endowing chairs in dermatology, and donating a famous obelisk in London).

Courtesy of Dr. Leslie S. Baumann
An advertisement in the September 1929 Ladies' Home Journal says that Camay soap has the 'unanimous approval' of '72 of the most eminent dermatologists in America.'


In 1854, Dr. Wilson wrote a book for laypeople called “Healthy Skin: A Popular Treatise on the Skin and Hair, Their Preservation and Management,” in which he advocated cleanliness and bathing, which led to the popularity of Turkish baths and bathing resorts in Europe. Despite his undeniable contributions to dermatology, he was widely criticized by his colleagues for promoting a “Hair Wash” and a turtle oil soap. I cannot find any information about whether or not he developed the hair wash and turtle soap himself, but it seems that he earned income from sales of these two products, even though he was said to have donated it all to charities.

William A. Pusey MD (1865-1940): Dr. Pusey was the first chairman of dermatology at the University of Illinois College of Medicine, Chicago. He published several books, including “Care of the Skin and Hair,” “Syphilis as a Modern Problem,” “The Principles and Practices of Dermatology,” and “History of Dermatology” among others. He is best known for his work in developing the use of x-rays (roentgen rays) and phototherapy in dermatology, and in 1907, he was the first dermatologist to describe the use of solid carbon dioxide to treat skin lesions. He was president of the American Dermatological Association in 1910, president of the Chicago Medical Society in 1918, editor of the Archives of Dermatology in 1920, and president of the American Medical Association in 1924.

Courtesy of Dr. Leslie S. Baumann


In the early 1920s, skin care companies were beginning to advertise their products using endorsements from celebrities and socialites, and were making misleading claims. Dr. Pusey wanted to work with these companies to help them perform evidence-based trials so they could make scientifically correct claims. Proctor & Gamble asked Dr. Pusey to advise them on how they could advertise honestly about their new soap, “Camay.” In Dr. Pusey’s words,3 “they (Proctor & Gamble) wanted to give the public authoritative advice about the use of soap and water. They suggested that I get a group of dermatologists of my selection to examine the soap and prepare instructions for bathing and the use of soap, and, if they found this soap was of high quality, to certify to that effect.” The research was performed as he suggested, and he allowed his name to be used in the Camay soap ads from 1926 to 1929. He said that he allowed them to use his name hoping to promote the need for evidence-based research, in contrast to the skin care products endorsed by socialites and celebrities that were flooding the market around that time.

Herbert Rattner, MD, at Northwestern University, Chicago, was his friend and one of the many dermatologists who criticized Dr. Pusey for allowing his name to be used in the Camay ads. Dr. Pusey’s reply to the criticism (according to Dr. Rattner) was that Proctor & Gamble was “proposing to do what the medical profession always is criticizing commercial concerns for not doing, namely, coming to physicians for information on medical matters. Could the profession hope to have any influence with business concerns if it was always eager to criticize bad commercial practices but never willing to support good ones?”3

While Dr. Pusey felt his reasons for adding his name to the Camay ads and research were justified, many of his friends stated that in hindsight, he regretted the action because of the negative response of his colleagues. It was years before dermatologists began providing input again into the skin care industry. During that time, radio, television and print ads were rampant with misleading claims – which led the way for a dermatologic imposter to make a fortune on skin care.

John Woodbury (1851-1909): John Woodbury, a chiropractor, never went to medical school, but that did not stop him from claiming he was a dermatologist and cosmetic surgeon. In 1889, he opened the John H. Woodbury Dermatological Institute in New York City, and over the next few years, opened Woodbury Dermatological Institutes in at least 5 states and employed 25 “physicians” who were not licensed to practice medicine. He came out with face soaps, tonics, and cold creams and spent a fortune on advertising these products and his institutes. In 1901, he sold his “Woodbury Soap” to the Andrew Jergens Company for $212,500 and 10% in royalties.

Multiple lawsuits occurred from 1898 to 1907 because he continued using the Woodbury name on his own products, despite having sold the “Woodbury” trademark to Jergens. He was sued for practicing medicine without a medical license and claiming to be a dermatologist when he was not. He lost most of these lawsuits, including one in 1907 in which the court ruled that corporations may not employ unlicensed professionals to practice medicine. In 1909, John Woodbury committed suicide. The Woodbury Soap company flourished in the 1930s and 1940s, as part of Jergens, until the brand was discontinued in 1970 when Jergens was acquired by American Brands.

1916 Ladies' Home Journal vol 33#9
A 1916 advertisement in Ladies' Home Journal: John Woodbury sold the Woodbury trademark to Jergens in 1901.


The next dermatologists to come along did not make the same mistakes as those of their predecessors. They all made scientific discoveries through their basic science research in the laboratory, filed patents, formed skin care companies, perfected the formulations, and conducted research trials of the final product. Their marketing focused on science and efficacy and only rarely used their names and images in advertising, allowing them to maintain their reputations in the dermatology field.

Eugene Van Scott, MD (1922-present): Dermatologist Dr. Van Scott and dermatopharmacologist Ruey Yu, PhD, filed a method patent in the early 1970s on the effectiveness of alpha hydroxy acids to treat ichthyosis. They invented the abbreviation “AHA” and have continued their work on organic acids to this day. They now have more than 125 patents, which they have licensed to 60 companies in the cosmetics and pharmaceutical industries.

In 1988, 14 years after their initial publication, they founded the company they named Polystrata, which grew into today’s NeoStrata.4 Over the years, they had to defend their patents because many personal care companies used their technologies without licensing them. In 2007, they won a $41 million settlement in a patent infringement suit against Mary Kay filed in March 2005. They have both been very philanthropic in the dermatology world5 and are highly respected in the field. Among many other honors, Dr. Van Scott was named a Master Dermatologist by the American Academy of Dermatology in 1998 and received the Dermatology Foundation’s Distinguished Service Medallion in 2004.

Sheldon Pinnell, MD (1937-2013): After Dr. Pinnell completed his dermatology residency at Harvard Medical School, he spent 2 years studying collagen chemistry at the Max Planck Institute in Munich, Germany. In 1973, he returned to Duke University where he had earned his undergraduate degree before attending Yale University. He remained at Duke for the duration of his career and was professor and chief of dermatology there for many years. Early in his career, he focused on the role of vitamin C in collagen biosynthesis and discovered some of the mechanisms by which sun exposure causes photoaging. He described the use of the first (and most popular) topically applied L-ascorbic acid (vitamin C) to prevent and treat skin aging.

Dr. Pinnell’s many discoveries include showing that the addition of ascorbic acid to fibroblast cultures increases collagen production and that topically applied L-ascorbic acid penetrates into the skin best at a pH of 2-2.5. Dr. Pinnell changed the way the world uses topical antioxidants today; he was widely respected and was a member of the American Dermatological Association and an honorary member of the Society of Investigative Dermatology. He published more than 200 scientific articles and held 10 patents. He started the skin care company Skinceuticals, based on his antioxidant technologies. It was acquired by L’Oreal in 2005.

Richard Fitzpatrick, MD (1944-2014): The dermatologist affectionately known as “Fitz” is credited with being the first to use lasers for skin resurfacing. He went to medical school at Emory University and did his dermatology residency at the University of California, Los Angeles. He authored more than 130 publications and was one of the first doctors to specialize in cosmetic dermatology. He realized that fibroblast cell cultures used to produce the collagen filler CosmoPlast (no longer on the market) generated many growth factors that could rejuvenate the skin, and in 1999, he launched the skin care brand SkinMedica. In 2000, he received a patent for fibroblast-derived growth factors used topically for antiaging – a formula he called Tissue Nutrient Solution. In 2001, the popular product TNS Recovery Complex was launched based on the patented growth factor technology. It is still the most popular growth factor technology on the market.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

What can we learn from these pioneers? I have had several interesting discussions about this topic with Leonard Hoenig, MD, section editor for Reflections on Dermatology: Past, Present, and Future, in Clinics in Dermatology. (Dr. Hoenig told me the interesting story that Listerine mouthwash was named in honor of Joseph Lister but accounts vary as to whether he gave permission to do so. This makes Dr. Lister the most famous physician to endorse a personal care product.) When Dr. Hoenig and I discussed the ethics of dermatologists creating a skin care line or retailing skin care in their medical practice, he stated my sentiments perfectly: “We should rely on professional, ethical, and legal guidelines to help us do what is right. Most importantly, we should have the best interests of our patients at heart when recommending any treatments.”

Dermatologists have unique knowledge, experience, and perspective on treating the skin with topical agents and have the true desire to improve skin health. If we do not discover, research, patent, and develop efficacious skin care products, someone else will do it – and I do not think they will do it as well as a dermatologist can.
 

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan and Burt’s Bees. She is the CEO of Skin Type Solutions Inc., a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Everett MA. Int J Dermatol. 1978 May;17(4):345-52.

2. Moxon RK. N Engl J Med. 1976 Apr 1;294(14):762-4.

3. Rattner H. Arch Derm Syphilol. 1937;35(1):25-66.

4. Neostrata: More than Hope, by Elaine Strauss, U.S. 1 Newspaper, Feb. 24, 1999.

5. Two legends in the field of dermatology provide $1 million gift to Temple University school of medicine’s department of dermatology, Temple University, June 5, 2015.

Those of you who have visited my dermatology practice in Miami know that the art in my office is dedicated to the history of the skin care industry. I collect vintage ads, and vintage skin care and personal care products, and biographies of anyone involved in skin care. I can’t get enough of the history of cosmetics, and I have written this historical column in honor of the 50th anniversary of Dermatology News.

Dr. Leslie S. Baumann in her office, in front of a Camay soap ad from the 1920s.
Courtesy Dr. Leslie S. Baumann
Dr. Leslie S. Baumann stands in her office, in front of a Camay soap ad from the 1920s.

The first doctor to market his own cosmetic product, Erasmus Wilson, MD, faced scrutiny from his colleagues. Although he had contributed much to the field of dermatology, he was criticized by other dermatologists when he promoted a hair wash. The next doctor in my story, William Pusey, MD, was criticized for helping the company that manufactured Camay soap because he allowed his name to be used in Camay advertisements. The scrutiny that these two well-respected dermatologists endured from their colleagues deterred dermatologists from entering the skin care business for decades. The professional jealousy from dermatologic colleagues left the skin care field wide open for imposters, charlatans, and nondermatologists who had no concern for efficacy and patient outcomes to flourish. This is the story of a group of brilliant entrepreneurial dermatologists and one chiropractor who misrepresented himself as a dermatologist and how they influenced skin care as we know it.

Erasmus Wilson, MD1 (1809-1884): In 1840, Erasmus Wilson2 was a physician in London who chose to specialize in dermatology at a time when that specialization was frowned upon. He was a subeditor for The Lancet and wrote several books on dermatology including “Diseases of the Skin – A Practical and Theoretical Treatise,” “Portraits of the Diseases of the Skin,” and “Student’s Book on Diseases of the Skin.” He was the first professor of dermatology in the College of Surgeons and by 1869, was the leading English-speaking dermatologist in the world. He contributed much to dermatology, including his pioneering characterizations of Demodex mites, lichen planus, exfoliative dermatitis, neurotic excoriations, and roseola. Dr. Wilson was knighted in 1881 for his good works and notable generosity. (He was known for giving his poor patients money for food, endowing chairs in dermatology, and donating a famous obelisk in London).

Courtesy of Dr. Leslie S. Baumann
An advertisement in the September 1929 Ladies' Home Journal says that Camay soap has the 'unanimous approval' of '72 of the most eminent dermatologists in America.'


In 1854, Dr. Wilson wrote a book for laypeople called “Healthy Skin: A Popular Treatise on the Skin and Hair, Their Preservation and Management,” in which he advocated cleanliness and bathing, which led to the popularity of Turkish baths and bathing resorts in Europe. Despite his undeniable contributions to dermatology, he was widely criticized by his colleagues for promoting a “Hair Wash” and a turtle oil soap. I cannot find any information about whether or not he developed the hair wash and turtle soap himself, but it seems that he earned income from sales of these two products, even though he was said to have donated it all to charities.

William A. Pusey MD (1865-1940): Dr. Pusey was the first chairman of dermatology at the University of Illinois College of Medicine, Chicago. He published several books, including “Care of the Skin and Hair,” “Syphilis as a Modern Problem,” “The Principles and Practices of Dermatology,” and “History of Dermatology” among others. He is best known for his work in developing the use of x-rays (roentgen rays) and phototherapy in dermatology, and in 1907, he was the first dermatologist to describe the use of solid carbon dioxide to treat skin lesions. He was president of the American Dermatological Association in 1910, president of the Chicago Medical Society in 1918, editor of the Archives of Dermatology in 1920, and president of the American Medical Association in 1924.

Courtesy of Dr. Leslie S. Baumann


In the early 1920s, skin care companies were beginning to advertise their products using endorsements from celebrities and socialites, and were making misleading claims. Dr. Pusey wanted to work with these companies to help them perform evidence-based trials so they could make scientifically correct claims. Proctor & Gamble asked Dr. Pusey to advise them on how they could advertise honestly about their new soap, “Camay.” In Dr. Pusey’s words,3 “they (Proctor & Gamble) wanted to give the public authoritative advice about the use of soap and water. They suggested that I get a group of dermatologists of my selection to examine the soap and prepare instructions for bathing and the use of soap, and, if they found this soap was of high quality, to certify to that effect.” The research was performed as he suggested, and he allowed his name to be used in the Camay soap ads from 1926 to 1929. He said that he allowed them to use his name hoping to promote the need for evidence-based research, in contrast to the skin care products endorsed by socialites and celebrities that were flooding the market around that time.

Herbert Rattner, MD, at Northwestern University, Chicago, was his friend and one of the many dermatologists who criticized Dr. Pusey for allowing his name to be used in the Camay ads. Dr. Pusey’s reply to the criticism (according to Dr. Rattner) was that Proctor & Gamble was “proposing to do what the medical profession always is criticizing commercial concerns for not doing, namely, coming to physicians for information on medical matters. Could the profession hope to have any influence with business concerns if it was always eager to criticize bad commercial practices but never willing to support good ones?”3

While Dr. Pusey felt his reasons for adding his name to the Camay ads and research were justified, many of his friends stated that in hindsight, he regretted the action because of the negative response of his colleagues. It was years before dermatologists began providing input again into the skin care industry. During that time, radio, television and print ads were rampant with misleading claims – which led the way for a dermatologic imposter to make a fortune on skin care.

John Woodbury (1851-1909): John Woodbury, a chiropractor, never went to medical school, but that did not stop him from claiming he was a dermatologist and cosmetic surgeon. In 1889, he opened the John H. Woodbury Dermatological Institute in New York City, and over the next few years, opened Woodbury Dermatological Institutes in at least 5 states and employed 25 “physicians” who were not licensed to practice medicine. He came out with face soaps, tonics, and cold creams and spent a fortune on advertising these products and his institutes. In 1901, he sold his “Woodbury Soap” to the Andrew Jergens Company for $212,500 and 10% in royalties.

Multiple lawsuits occurred from 1898 to 1907 because he continued using the Woodbury name on his own products, despite having sold the “Woodbury” trademark to Jergens. He was sued for practicing medicine without a medical license and claiming to be a dermatologist when he was not. He lost most of these lawsuits, including one in 1907 in which the court ruled that corporations may not employ unlicensed professionals to practice medicine. In 1909, John Woodbury committed suicide. The Woodbury Soap company flourished in the 1930s and 1940s, as part of Jergens, until the brand was discontinued in 1970 when Jergens was acquired by American Brands.

1916 Ladies' Home Journal vol 33#9
A 1916 advertisement in Ladies' Home Journal: John Woodbury sold the Woodbury trademark to Jergens in 1901.


The next dermatologists to come along did not make the same mistakes as those of their predecessors. They all made scientific discoveries through their basic science research in the laboratory, filed patents, formed skin care companies, perfected the formulations, and conducted research trials of the final product. Their marketing focused on science and efficacy and only rarely used their names and images in advertising, allowing them to maintain their reputations in the dermatology field.

Eugene Van Scott, MD (1922-present): Dermatologist Dr. Van Scott and dermatopharmacologist Ruey Yu, PhD, filed a method patent in the early 1970s on the effectiveness of alpha hydroxy acids to treat ichthyosis. They invented the abbreviation “AHA” and have continued their work on organic acids to this day. They now have more than 125 patents, which they have licensed to 60 companies in the cosmetics and pharmaceutical industries.

In 1988, 14 years after their initial publication, they founded the company they named Polystrata, which grew into today’s NeoStrata.4 Over the years, they had to defend their patents because many personal care companies used their technologies without licensing them. In 2007, they won a $41 million settlement in a patent infringement suit against Mary Kay filed in March 2005. They have both been very philanthropic in the dermatology world5 and are highly respected in the field. Among many other honors, Dr. Van Scott was named a Master Dermatologist by the American Academy of Dermatology in 1998 and received the Dermatology Foundation’s Distinguished Service Medallion in 2004.

Sheldon Pinnell, MD (1937-2013): After Dr. Pinnell completed his dermatology residency at Harvard Medical School, he spent 2 years studying collagen chemistry at the Max Planck Institute in Munich, Germany. In 1973, he returned to Duke University where he had earned his undergraduate degree before attending Yale University. He remained at Duke for the duration of his career and was professor and chief of dermatology there for many years. Early in his career, he focused on the role of vitamin C in collagen biosynthesis and discovered some of the mechanisms by which sun exposure causes photoaging. He described the use of the first (and most popular) topically applied L-ascorbic acid (vitamin C) to prevent and treat skin aging.

Dr. Pinnell’s many discoveries include showing that the addition of ascorbic acid to fibroblast cultures increases collagen production and that topically applied L-ascorbic acid penetrates into the skin best at a pH of 2-2.5. Dr. Pinnell changed the way the world uses topical antioxidants today; he was widely respected and was a member of the American Dermatological Association and an honorary member of the Society of Investigative Dermatology. He published more than 200 scientific articles and held 10 patents. He started the skin care company Skinceuticals, based on his antioxidant technologies. It was acquired by L’Oreal in 2005.

Richard Fitzpatrick, MD (1944-2014): The dermatologist affectionately known as “Fitz” is credited with being the first to use lasers for skin resurfacing. He went to medical school at Emory University and did his dermatology residency at the University of California, Los Angeles. He authored more than 130 publications and was one of the first doctors to specialize in cosmetic dermatology. He realized that fibroblast cell cultures used to produce the collagen filler CosmoPlast (no longer on the market) generated many growth factors that could rejuvenate the skin, and in 1999, he launched the skin care brand SkinMedica. In 2000, he received a patent for fibroblast-derived growth factors used topically for antiaging – a formula he called Tissue Nutrient Solution. In 2001, the popular product TNS Recovery Complex was launched based on the patented growth factor technology. It is still the most popular growth factor technology on the market.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

What can we learn from these pioneers? I have had several interesting discussions about this topic with Leonard Hoenig, MD, section editor for Reflections on Dermatology: Past, Present, and Future, in Clinics in Dermatology. (Dr. Hoenig told me the interesting story that Listerine mouthwash was named in honor of Joseph Lister but accounts vary as to whether he gave permission to do so. This makes Dr. Lister the most famous physician to endorse a personal care product.) When Dr. Hoenig and I discussed the ethics of dermatologists creating a skin care line or retailing skin care in their medical practice, he stated my sentiments perfectly: “We should rely on professional, ethical, and legal guidelines to help us do what is right. Most importantly, we should have the best interests of our patients at heart when recommending any treatments.”

Dermatologists have unique knowledge, experience, and perspective on treating the skin with topical agents and have the true desire to improve skin health. If we do not discover, research, patent, and develop efficacious skin care products, someone else will do it – and I do not think they will do it as well as a dermatologist can.
 

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan and Burt’s Bees. She is the CEO of Skin Type Solutions Inc., a company that independently tests skin care products and makes recommendations to physicians on which skin care technologies are best. Write to her at dermnews@mdedge.com.

References

1. Everett MA. Int J Dermatol. 1978 May;17(4):345-52.

2. Moxon RK. N Engl J Med. 1976 Apr 1;294(14):762-4.

3. Rattner H. Arch Derm Syphilol. 1937;35(1):25-66.

4. Neostrata: More than Hope, by Elaine Strauss, U.S. 1 Newspaper, Feb. 24, 1999.

5. Two legends in the field of dermatology provide $1 million gift to Temple University school of medicine’s department of dermatology, Temple University, June 5, 2015.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Is bufexamac worth the risk?

Article Type
Changed
Wed, 07/15/2020 - 09:15

Bufexamac, a nonsteroidal anti-inflammatory drug agent used cutaneously and rectally, is well known globally as an initiator of allergic contact dermatitis. In fact, it has been removed from the European market (except Switzerland) for inducing allergic reactions, and is also banned in Japan, New Zealand, and the United States (where it was never approved).1 This column will primarily discuss recent findings in human trials and weigh in on the issue.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Antioxidant activity

In addition to its known anti-inflammatory activity, bufexamac has been found to exert antioxidant effects. In 2003, Trommer and Neubert demonstrated that bufexamac displayed antioxidant activity in lipid models and HaCaT keratinocytes, as measured through mass spectrometry.2 In a 2005 in vitro study of the impact of 47 drugs, plant extracts and ingredients, and polysaccharides on lipid peroxidation engendered by UV irradiation, Trommer and Neubert found that bufexamac was among the drugs shown to exhibit antioxidant activity.3

Minor allergen? Worth using?

In a 2009 study on the prevalence and risk factors for allergic contact dermatitis to topical atopic dermatitis (AD) treatments, Mailhol et al. patch tested 641 children with AD using seven then-common ingredients (chlorhexidine, hexamidine, budesonide, tixocortol pivalate, bufexamac, sodium fusidate and with the current emollient used by the child). Bufexamac was identified as an allergen in only 2.5% of the 41 positive patch tests.4

To ban or not to ban

In 2012, the European Medicines Agency’s Committee for Medicinal Products for Human Use recommended that the marketing of formulations containing bufexamac be disallowed throughout the European Union because of a tendency toward inducing severe allergic contact dermatitis.5

Given its continuing use in Australia for the local treatment of several dermatoses, Pan and Nixon, in 2012, retrospectively reviewed patch-test data at the Skin and Cancer Foundation Inc. and found 19 cases of positive reactions to bufexamac (5% petrolatum) from 451 people patch tested. In 13 of 19 patients (68%), the reaction to bufexamac was considered to be associated with the identified dermatitis. The authors concluded that allergic contact dermatitis from bufexamac exposure is underreported in the English-language literature and cautioned that physicians should consider bufexamac allergy in patients who have a history of exposure.5

Bufexamac remained available over the counter in topical formulations in Australia as of early 2019. In response, Harris et al. presented several cases of patients who experienced severe skin eruptions after using such preparations in support of their advocacy to the Therapeutic Goods Administration in Australia to ban its use.6

In the middle of that year, Wong et al. reported on the hospitalization of a 41-year-old administrative worker who applied a first aid cream containing bufexamac (5%), lignocaine (1%), and chlorhexidine (0.1%) to a superficial right foot abrasion and who developed facial edema and widespread polymorphic eruptions 2 hours later. The authors suggested that this case reinforced the need to remove bufexamac from the markets where it remains because of the tendency to provoke severe allergic contact dermatoses and lack of efficacy.1

Conclusion

Bufexamac offers the somewhat rare opportunity for advocacy. That is to say, I think there is sufficient evidence to justify the removal of this potent allergen from the market in Australia, Switzerland, and other countries where it may be available.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems LLC. She had no relevant disclosures. Write to her at dermnews@mdedge.com.

References

1. Wong GN et al. Contact Dermatitis. 2019 Jun;80(6):395-7.

2. Trommer H et al. J Pharm Pharmacol. 2003 Oct;55(10):1379-88.

3. Trommer H, Neubert RH. J Pharm Pharm Sci. 2005 Sep 15;8(3):494-506.

4. Mailhol C et al. Allergy. 2009 May;64(5):801-6.

5. Pan Y, Nixon R. Australas J Dermatol. 2012 Aug;53(3):207-10.

6. Harris AG et al. Australas J Dermatol. 2019 Feb;60(1):53-6.

Publications
Topics
Sections

Bufexamac, a nonsteroidal anti-inflammatory drug agent used cutaneously and rectally, is well known globally as an initiator of allergic contact dermatitis. In fact, it has been removed from the European market (except Switzerland) for inducing allergic reactions, and is also banned in Japan, New Zealand, and the United States (where it was never approved).1 This column will primarily discuss recent findings in human trials and weigh in on the issue.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Antioxidant activity

In addition to its known anti-inflammatory activity, bufexamac has been found to exert antioxidant effects. In 2003, Trommer and Neubert demonstrated that bufexamac displayed antioxidant activity in lipid models and HaCaT keratinocytes, as measured through mass spectrometry.2 In a 2005 in vitro study of the impact of 47 drugs, plant extracts and ingredients, and polysaccharides on lipid peroxidation engendered by UV irradiation, Trommer and Neubert found that bufexamac was among the drugs shown to exhibit antioxidant activity.3

Minor allergen? Worth using?

In a 2009 study on the prevalence and risk factors for allergic contact dermatitis to topical atopic dermatitis (AD) treatments, Mailhol et al. patch tested 641 children with AD using seven then-common ingredients (chlorhexidine, hexamidine, budesonide, tixocortol pivalate, bufexamac, sodium fusidate and with the current emollient used by the child). Bufexamac was identified as an allergen in only 2.5% of the 41 positive patch tests.4

To ban or not to ban

In 2012, the European Medicines Agency’s Committee for Medicinal Products for Human Use recommended that the marketing of formulations containing bufexamac be disallowed throughout the European Union because of a tendency toward inducing severe allergic contact dermatitis.5

Given its continuing use in Australia for the local treatment of several dermatoses, Pan and Nixon, in 2012, retrospectively reviewed patch-test data at the Skin and Cancer Foundation Inc. and found 19 cases of positive reactions to bufexamac (5% petrolatum) from 451 people patch tested. In 13 of 19 patients (68%), the reaction to bufexamac was considered to be associated with the identified dermatitis. The authors concluded that allergic contact dermatitis from bufexamac exposure is underreported in the English-language literature and cautioned that physicians should consider bufexamac allergy in patients who have a history of exposure.5

Bufexamac remained available over the counter in topical formulations in Australia as of early 2019. In response, Harris et al. presented several cases of patients who experienced severe skin eruptions after using such preparations in support of their advocacy to the Therapeutic Goods Administration in Australia to ban its use.6

In the middle of that year, Wong et al. reported on the hospitalization of a 41-year-old administrative worker who applied a first aid cream containing bufexamac (5%), lignocaine (1%), and chlorhexidine (0.1%) to a superficial right foot abrasion and who developed facial edema and widespread polymorphic eruptions 2 hours later. The authors suggested that this case reinforced the need to remove bufexamac from the markets where it remains because of the tendency to provoke severe allergic contact dermatoses and lack of efficacy.1

Conclusion

Bufexamac offers the somewhat rare opportunity for advocacy. That is to say, I think there is sufficient evidence to justify the removal of this potent allergen from the market in Australia, Switzerland, and other countries where it may be available.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems LLC. She had no relevant disclosures. Write to her at dermnews@mdedge.com.

References

1. Wong GN et al. Contact Dermatitis. 2019 Jun;80(6):395-7.

2. Trommer H et al. J Pharm Pharmacol. 2003 Oct;55(10):1379-88.

3. Trommer H, Neubert RH. J Pharm Pharm Sci. 2005 Sep 15;8(3):494-506.

4. Mailhol C et al. Allergy. 2009 May;64(5):801-6.

5. Pan Y, Nixon R. Australas J Dermatol. 2012 Aug;53(3):207-10.

6. Harris AG et al. Australas J Dermatol. 2019 Feb;60(1):53-6.

Bufexamac, a nonsteroidal anti-inflammatory drug agent used cutaneously and rectally, is well known globally as an initiator of allergic contact dermatitis. In fact, it has been removed from the European market (except Switzerland) for inducing allergic reactions, and is also banned in Japan, New Zealand, and the United States (where it was never approved).1 This column will primarily discuss recent findings in human trials and weigh in on the issue.

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Antioxidant activity

In addition to its known anti-inflammatory activity, bufexamac has been found to exert antioxidant effects. In 2003, Trommer and Neubert demonstrated that bufexamac displayed antioxidant activity in lipid models and HaCaT keratinocytes, as measured through mass spectrometry.2 In a 2005 in vitro study of the impact of 47 drugs, plant extracts and ingredients, and polysaccharides on lipid peroxidation engendered by UV irradiation, Trommer and Neubert found that bufexamac was among the drugs shown to exhibit antioxidant activity.3

Minor allergen? Worth using?

In a 2009 study on the prevalence and risk factors for allergic contact dermatitis to topical atopic dermatitis (AD) treatments, Mailhol et al. patch tested 641 children with AD using seven then-common ingredients (chlorhexidine, hexamidine, budesonide, tixocortol pivalate, bufexamac, sodium fusidate and with the current emollient used by the child). Bufexamac was identified as an allergen in only 2.5% of the 41 positive patch tests.4

To ban or not to ban

In 2012, the European Medicines Agency’s Committee for Medicinal Products for Human Use recommended that the marketing of formulations containing bufexamac be disallowed throughout the European Union because of a tendency toward inducing severe allergic contact dermatitis.5

Given its continuing use in Australia for the local treatment of several dermatoses, Pan and Nixon, in 2012, retrospectively reviewed patch-test data at the Skin and Cancer Foundation Inc. and found 19 cases of positive reactions to bufexamac (5% petrolatum) from 451 people patch tested. In 13 of 19 patients (68%), the reaction to bufexamac was considered to be associated with the identified dermatitis. The authors concluded that allergic contact dermatitis from bufexamac exposure is underreported in the English-language literature and cautioned that physicians should consider bufexamac allergy in patients who have a history of exposure.5

Bufexamac remained available over the counter in topical formulations in Australia as of early 2019. In response, Harris et al. presented several cases of patients who experienced severe skin eruptions after using such preparations in support of their advocacy to the Therapeutic Goods Administration in Australia to ban its use.6

In the middle of that year, Wong et al. reported on the hospitalization of a 41-year-old administrative worker who applied a first aid cream containing bufexamac (5%), lignocaine (1%), and chlorhexidine (0.1%) to a superficial right foot abrasion and who developed facial edema and widespread polymorphic eruptions 2 hours later. The authors suggested that this case reinforced the need to remove bufexamac from the markets where it remains because of the tendency to provoke severe allergic contact dermatoses and lack of efficacy.1

Conclusion

Bufexamac offers the somewhat rare opportunity for advocacy. That is to say, I think there is sufficient evidence to justify the removal of this potent allergen from the market in Australia, Switzerland, and other countries where it may be available.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann has written two textbooks and a New York Times Best Sellers book for consumers. Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems LLC. She had no relevant disclosures. Write to her at dermnews@mdedge.com.

References

1. Wong GN et al. Contact Dermatitis. 2019 Jun;80(6):395-7.

2. Trommer H et al. J Pharm Pharmacol. 2003 Oct;55(10):1379-88.

3. Trommer H, Neubert RH. J Pharm Pharm Sci. 2005 Sep 15;8(3):494-506.

4. Mailhol C et al. Allergy. 2009 May;64(5):801-6.

5. Pan Y, Nixon R. Australas J Dermatol. 2012 Aug;53(3):207-10.

6. Harris AG et al. Australas J Dermatol. 2019 Feb;60(1):53-6.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article

Snail mucus in skin care

Article Type
Changed
Mon, 06/15/2020 - 09:16

Although it is not glamorous, snail mucus is a popular ingredient in skin care products. Its use dates back to ancient Greece, where Hippocrates reportedly applied crushed snails to treat skin inflammations.1 The modern consideration of using snail secretions in skin care arose serendipitously in the 1990s when Chilean farmers observed accelerated healing of their skin lesions without scarring after handling snails.1

Garden Snail on green leaf
valentinrussanov/E+

Today, snail mucin is among the increasingly wide array of bioactive ingredients undergoing scientific validation and inclusion in the burgeoning Korean cosmeceutical market.2,3 In fact, a variety of Korean cosmeceuticals incorporate the mucus derived from Achatina fulica (African giant land snail) and Cryptomphalus (Helix) aspersa (common brown garden snail) based on their demonstrated antimicrobial and skin regenerative activity.1,3,4 The antioxidant properties also attributed to snail mucus are thought to originate in constituents such as glycosaminoglycans, as well as growth factors, and may justify the use of these ingredients in novel cosmeceuticals.5 The focus of this discussion is recent research into the novel use of this animal-derived product for dermatologic purposes.
 

Antioxidant activity, skin rejuvenation, and wound healing

In 2008, Brieva et al. reported on a screen for natural products yielding a molecular basis for the secretions of the mollusk Cryptomphalus aspersa, which displays skin-regenerative activity. Specifically, they found that the secretion exerts antioxidant superoxide dismutase and glutathione S-transferase, and spurred fibroblast proliferation and extracellular matrix assembly while regulating metalloproteinase function. The researchers concluded that such activities may support wound regeneration.5

Four years later, Cruz et al. found that secretions of C. aspersa promote in vitro cell proliferation and migration by localizing beta-catenin to the nuclei of human fibroblasts and keratinocytes, augment phosphorylated focal adhesion kinase, and thereby enhance cell survival. The investigators concluded that snail secretions may therefore impart regenerative and wound healing activity.3,6

Antimicrobial properties

In 2015, Pitt et al. investigated the antimicrobial properties of the mucus of the brown garden snail C. or H. aspersa, which had a reputation for exhibiting skin regeneration capabilities. Their results revealed that snail mucus displayed a strong antibacterial effect against multiple strains of Pseudomonas aeruginosa and a weak effect against Staphylococcus aureus.4

Indications for the use of snail mucin

Radiation-induced dermatitis and burns represented the first indication for the initial use of snail mucin as a cutaneous therapy.7 Experimental and clinical studies have since been performed to assess its applicability to treat acute radiation dermatitis, atopic dermatitis, partial-thickness burns, and photoaging.8-11

A 2017 in vitro investigation by Ellijimi et al. revealed that snail mucin displayed antimelanogenic and antitumoral activity against human melanoma cells, suggesting another possible application of this product.12

Human studies on photoaging

In a 2009 study by Tsoutsos et al. of an open, moist burn management protocol in deep partial-thickness facial burns, a cream containing H. aspersa secretions was identified to be an effective treatment option. For 14 days or until full epithelialization, 27 adult patients were treated with snail extract cream twice daily. Comparisons were made to 16 patients treated with moist exposure burn ointment. Visual analog scale pain scores were significantly lower in the group that received the H. aspersa cream, compared with the moist exposure burn group. The researchers concluded that the H. aspersa cream is a safe, effective, and natural option for treating partial-thickness burns in adults that acts by facilitating debris removal and accelerating reepithelialization.10

Also that year, Tribo-Boixareu et al. treated 15 patients with chronic photodamage with secretions of C. aspersa over a 3-month period, yielding significant amelioration in the clinical and histologic markers of photoaging.11

Four years later, a double-blind, split-face, randomized, controlled clinical study conducted by Fabi et al. over 12 weeks demonstrated that the topical application of an antiphotoaging formulation containing C. aspersa mucus diminished periocular and fine facial rhytides and enhanced skin texture within 8 weeks of treatment initiation.7

Snail eggs and photoaging

In 2015, Espada et al. determined in vitro that an extract derived from C. aspersa eggs could reorganize the cytoskeleton of keratinocytes and fibroblasts, as well as trigger the synthesis of the extracellular proteins collagen and fibronectin. They also found that gene expression declined in age-related genes including p53 and b-Gal. The researchers concluded that C. aspersa egg extract has the potential to reduce the signs of photoaging.3,13

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Antiaging cosmeceuticals

In a 2017 assessment of the antiaging and skin-whitening activity of the nine most popular ingredients in the South Korean skin care product market, Quay et al. considered industry profit data from Euromonitor and conducted a comprehensive literature search. They identified licorice, niacinamide, green tea, soy, beta-glucan, snail mucus, ginkgo biloba, ginseng, and pomegranate as the nine most popular ingredients, with the first four associated with the most supportive data. They found a paucity of cogent evidence on the use of the other ingredients in antiaging and skin-whitening formulations.14

Conclusion

The use of snail mucin to treat skin dates back at least to the time of Hippocrates. Recent research suggests reasons for optimism, and further investigation, as this ingredient appears to have potential across various cutaneous conditions. As is often the case, though, much more research is necessary to ascertain what enduring benefits may be derived from the use of snail mucin. Nevertheless, this product has been available on the market for the last 20 years and is associated with anecdotal reports of efficacy.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann wrote two textbooks: “Cosmetic Dermatology: Principles and Practice” (New York: McGraw-Hill, 2002), and “Cosmeceuticals and Cosmetic Ingredients” (New York: McGraw-Hill, 2014), and a New York Times Best Sellers book for consumers, “The Skin Type Solution” (New York: Bantam Dell, 2006). Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems LLC. Write to her at dermnews@mdedge.com. She has no relevant disclosures.

References

1. Liu L et al. Snails and skin care – an uncovered combination. JAMA Dermatol. 2017 Jul 1;153(7):650.

2. Nguyen JK et al. J Cosmet Dermatol. 2020 Feb 26. doi: 10.1111/jocd.13344.

3. Juhász ML et al. J Cosmet Dermatol. 2018;17(3):305-12.

4. Pitt SJ et al. Br J Biomed Sci. 2015;72(4):174-81.

5. Brieva A et al. Skin Pharmacol Physiol. 2008;21(1):15-22.

6. Cruz MC et al. Int J Cosmet Sci. 2012 Apr;34(2):183-9.

7. Fabi SG et al. J Drugs Dermatol. 2013; Apr;12(4):453-7.

8. Ledo E et al. Radioproteccion. 1999;23(7):34-8.

9. Oh M-Jet al. J Korean Med Ophthalmol Otolaryngol Dermatol. 2010; Dec,23(3):138-53.

10. Tsoutsos D et al. J Dermatolog Treat. 2009;20(4):219-22.

11. Tribo-Boixareu MJ et al. Cosmet Dermatol. 2009;22(5):247-52.

12. Ellijimi C et al. Biomed Pharmacother. 2018 May;101:871-80.

13. Espada J et al. Int J Cosmet Sci. 2015 Feb;37(1):41-55.

14. Quay ER et al. J Drugs Dermatol. 2017 Apr 1;16(4):358-63.
 

Publications
Topics
Sections

Although it is not glamorous, snail mucus is a popular ingredient in skin care products. Its use dates back to ancient Greece, where Hippocrates reportedly applied crushed snails to treat skin inflammations.1 The modern consideration of using snail secretions in skin care arose serendipitously in the 1990s when Chilean farmers observed accelerated healing of their skin lesions without scarring after handling snails.1

Garden Snail on green leaf
valentinrussanov/E+

Today, snail mucin is among the increasingly wide array of bioactive ingredients undergoing scientific validation and inclusion in the burgeoning Korean cosmeceutical market.2,3 In fact, a variety of Korean cosmeceuticals incorporate the mucus derived from Achatina fulica (African giant land snail) and Cryptomphalus (Helix) aspersa (common brown garden snail) based on their demonstrated antimicrobial and skin regenerative activity.1,3,4 The antioxidant properties also attributed to snail mucus are thought to originate in constituents such as glycosaminoglycans, as well as growth factors, and may justify the use of these ingredients in novel cosmeceuticals.5 The focus of this discussion is recent research into the novel use of this animal-derived product for dermatologic purposes.
 

Antioxidant activity, skin rejuvenation, and wound healing

In 2008, Brieva et al. reported on a screen for natural products yielding a molecular basis for the secretions of the mollusk Cryptomphalus aspersa, which displays skin-regenerative activity. Specifically, they found that the secretion exerts antioxidant superoxide dismutase and glutathione S-transferase, and spurred fibroblast proliferation and extracellular matrix assembly while regulating metalloproteinase function. The researchers concluded that such activities may support wound regeneration.5

Four years later, Cruz et al. found that secretions of C. aspersa promote in vitro cell proliferation and migration by localizing beta-catenin to the nuclei of human fibroblasts and keratinocytes, augment phosphorylated focal adhesion kinase, and thereby enhance cell survival. The investigators concluded that snail secretions may therefore impart regenerative and wound healing activity.3,6

Antimicrobial properties

In 2015, Pitt et al. investigated the antimicrobial properties of the mucus of the brown garden snail C. or H. aspersa, which had a reputation for exhibiting skin regeneration capabilities. Their results revealed that snail mucus displayed a strong antibacterial effect against multiple strains of Pseudomonas aeruginosa and a weak effect against Staphylococcus aureus.4

Indications for the use of snail mucin

Radiation-induced dermatitis and burns represented the first indication for the initial use of snail mucin as a cutaneous therapy.7 Experimental and clinical studies have since been performed to assess its applicability to treat acute radiation dermatitis, atopic dermatitis, partial-thickness burns, and photoaging.8-11

A 2017 in vitro investigation by Ellijimi et al. revealed that snail mucin displayed antimelanogenic and antitumoral activity against human melanoma cells, suggesting another possible application of this product.12

Human studies on photoaging

In a 2009 study by Tsoutsos et al. of an open, moist burn management protocol in deep partial-thickness facial burns, a cream containing H. aspersa secretions was identified to be an effective treatment option. For 14 days or until full epithelialization, 27 adult patients were treated with snail extract cream twice daily. Comparisons were made to 16 patients treated with moist exposure burn ointment. Visual analog scale pain scores were significantly lower in the group that received the H. aspersa cream, compared with the moist exposure burn group. The researchers concluded that the H. aspersa cream is a safe, effective, and natural option for treating partial-thickness burns in adults that acts by facilitating debris removal and accelerating reepithelialization.10

Also that year, Tribo-Boixareu et al. treated 15 patients with chronic photodamage with secretions of C. aspersa over a 3-month period, yielding significant amelioration in the clinical and histologic markers of photoaging.11

Four years later, a double-blind, split-face, randomized, controlled clinical study conducted by Fabi et al. over 12 weeks demonstrated that the topical application of an antiphotoaging formulation containing C. aspersa mucus diminished periocular and fine facial rhytides and enhanced skin texture within 8 weeks of treatment initiation.7

Snail eggs and photoaging

In 2015, Espada et al. determined in vitro that an extract derived from C. aspersa eggs could reorganize the cytoskeleton of keratinocytes and fibroblasts, as well as trigger the synthesis of the extracellular proteins collagen and fibronectin. They also found that gene expression declined in age-related genes including p53 and b-Gal. The researchers concluded that C. aspersa egg extract has the potential to reduce the signs of photoaging.3,13

Dr. Leslie S. Baumann, a dermatologist, researcher, author, and entrepreneur who practices in Miami.
Dr. Leslie S. Baumann

Antiaging cosmeceuticals

In a 2017 assessment of the antiaging and skin-whitening activity of the nine most popular ingredients in the South Korean skin care product market, Quay et al. considered industry profit data from Euromonitor and conducted a comprehensive literature search. They identified licorice, niacinamide, green tea, soy, beta-glucan, snail mucus, ginkgo biloba, ginseng, and pomegranate as the nine most popular ingredients, with the first four associated with the most supportive data. They found a paucity of cogent evidence on the use of the other ingredients in antiaging and skin-whitening formulations.14

Conclusion

The use of snail mucin to treat skin dates back at least to the time of Hippocrates. Recent research suggests reasons for optimism, and further investigation, as this ingredient appears to have potential across various cutaneous conditions. As is often the case, though, much more research is necessary to ascertain what enduring benefits may be derived from the use of snail mucin. Nevertheless, this product has been available on the market for the last 20 years and is associated with anecdotal reports of efficacy.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann wrote two textbooks: “Cosmetic Dermatology: Principles and Practice” (New York: McGraw-Hill, 2002), and “Cosmeceuticals and Cosmetic Ingredients” (New York: McGraw-Hill, 2014), and a New York Times Best Sellers book for consumers, “The Skin Type Solution” (New York: Bantam Dell, 2006). Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems LLC. Write to her at dermnews@mdedge.com. She has no relevant disclosures.

References

1. Liu L et al. Snails and skin care – an uncovered combination. JAMA Dermatol. 2017 Jul 1;153(7):650.

2. Nguyen JK et al. J Cosmet Dermatol. 2020 Feb 26. doi: 10.1111/jocd.13344.

3. Juhász ML et al. J Cosmet Dermatol. 2018;17(3):305-12.

4. Pitt SJ et al. Br J Biomed Sci. 2015;72(4):174-81.

5. Brieva A et al. Skin Pharmacol Physiol. 2008;21(1):15-22.

6. Cruz MC et al. Int J Cosmet Sci. 2012 Apr;34(2):183-9.

7. Fabi SG et al. J Drugs Dermatol. 2013; Apr;12(4):453-7.

8. Ledo E et al. Radioproteccion. 1999;23(7):34-8.

9. Oh M-Jet al. J Korean Med Ophthalmol Otolaryngol Dermatol. 2010; Dec,23(3):138-53.

10. Tsoutsos D et al. J Dermatolog Treat. 2009;20(4):219-22.

11. Tribo-Boixareu MJ et al. Cosmet Dermatol. 2009;22(5):247-52.

12. Ellijimi C et al. Biomed Pharmacother. 2018 May;101:871-80.

13. Espada J et al. Int J Cosmet Sci. 2015 Feb;37(1):41-55.

14. Quay ER et al. J Drugs Dermatol. 2017 Apr 1;16(4):358-63.
 

Although it is not glamorous, snail mucus is a popular ingredient in skin care products. Its use dates back to ancient Greece, where Hippocrates reportedly applied crushed snails to treat skin inflammations.1 The modern consideration of using snail secretions in skin care arose serendipitously in the 1990s when Chilean farmers observed accelerated healing of their skin lesions without scarring after handling snails.1

Garden Snail on green leaf
valentinrussanov/E+

Today, snail mucin is among the increasingly wide array of bioactive ingredients undergoing scientific validation and inclusion in the burgeoning Korean cosmeceutical market.2,3 In fact, a variety of Korean cosmeceuticals incorporate the mucus derived from Achatina fulica (African giant land snail) and Cryptomphalus (Helix) aspersa (common brown garden snail) based on their demonstrated antimicrobial and skin regenerative activity.1,3,4 The antioxidant properties also attributed to snail mucus are thought to originate in constituents such as glycosaminoglycans, as well as growth factors, and may justify the use of these ingredients in novel cosmeceuticals.5 The focus of this discussion is recent research into the novel use of this animal-derived product for dermatologic purposes.
 

Antioxidant activity, skin rejuvenation, and wound healing

In 2008, Brieva et al. reported on a screen for natural products yielding a molecular basis for the secretions of the mollusk Cryptomphalus aspersa, which displays skin-regenerative activity. Specifically, they found that the secretion exerts antioxidant superoxide dismutase and glutathione S-transferase, and spurred fibroblast proliferation and extracellular matrix assembly while regulating metalloproteinase function. The researchers concluded that such activities may support wound regeneration.5

Four years later, Cruz et al. found that secretions of C. aspersa promote in vitro cell proliferation and migration by localizing beta-catenin to the nuclei of human fibroblasts and keratinocytes, augment phosphorylated focal adhesion kinase, and thereby enhance cell survival. The investigators concluded that snail secretions may therefore impart regenerative and wound healing activity.3,6

Antimicrobial properties

In 2015, Pitt et al. investigated the antimicrobial properties of the mucus of the brown garden snail C. or H. aspersa, which had a reputation for exhibiting skin regeneration capabilities. Their results revealed that snail mucus displayed a strong antibacterial effect against multiple strains of Pseudomonas aeruginosa and a weak effect against Staphylococcus aureus.4

Indications for the use of snail mucin

Radiation-induced dermatitis and burns represented the first indication for the initial use of snail mucin as a cutaneous therapy.7 Experimental and clinical studies have since been performed to assess its applicability to treat acute radiation dermatitis, atopic dermatitis, partial-thickness burns, and photoaging.8-11

A 2017 in vitro investigation by Ellijimi et al. revealed that snail mucin displayed antimelanogenic and antitumoral activity against human melanoma cells, suggesting another possible application of this product.12

Human studies on photoaging

In a 2009 study by Tsoutsos et al. of an open, moist burn management protocol in deep partial-thickness facial burns, a cream containing H. aspersa secretions was identified to be an effective treatment option. For 14 days or until full epithelialization, 27 adult patients were treated with snail extract cream twice daily. Comparisons were made to 16 patients treated with moist exposure burn ointment. Visual analog scale pain scores were significantly lower in the group that received the H. aspersa cream, compared with the moist exposure burn group. The researchers concluded that the H. aspersa cream is a safe, effective, and natural option for treating partial-thickness burns in adults that acts by facilitating debris removal and accelerating reepithelialization.10

Also that year, Tribo-Boixareu et al. treated 15 patients with chronic photodamage with secretions of C. aspersa over a 3-month period, yielding significant amelioration in the clinical and histologic markers of photoaging.11

Four years later, a double-blind, split-face, randomized, controlled clinical study conducted by Fabi et al. over 12 weeks demonstrated that the topical application of an antiphotoaging formulation containing C. aspersa mucus diminished periocular and fine facial rhytides and enhanced skin texture within 8 weeks of treatment initiation.7

Snail eggs and photoaging

In 2015, Espada et al. determined in vitro that an extract derived from C. aspersa eggs could reorganize the cytoskeleton of keratinocytes and fibroblasts, as well as trigger the synthesis of the extracellular proteins collagen and fibronectin. They also found that gene expression declined in age-related genes including p53 and b-Gal. The researchers concluded that C. aspersa egg extract has the potential to reduce the signs of photoaging.3,13

Dr. Leslie S. Baumann

Antiaging cosmeceuticals

In a 2017 assessment of the antiaging and skin-whitening activity of the nine most popular ingredients in the South Korean skin care product market, Quay et al. considered industry profit data from Euromonitor and conducted a comprehensive literature search. They identified licorice, niacinamide, green tea, soy, beta-glucan, snail mucus, ginkgo biloba, ginseng, and pomegranate as the nine most popular ingredients, with the first four associated with the most supportive data. They found a paucity of cogent evidence on the use of the other ingredients in antiaging and skin-whitening formulations.14

Conclusion

The use of snail mucin to treat skin dates back at least to the time of Hippocrates. Recent research suggests reasons for optimism, and further investigation, as this ingredient appears to have potential across various cutaneous conditions. As is often the case, though, much more research is necessary to ascertain what enduring benefits may be derived from the use of snail mucin. Nevertheless, this product has been available on the market for the last 20 years and is associated with anecdotal reports of efficacy.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann wrote two textbooks: “Cosmetic Dermatology: Principles and Practice” (New York: McGraw-Hill, 2002), and “Cosmeceuticals and Cosmetic Ingredients” (New York: McGraw-Hill, 2014), and a New York Times Best Sellers book for consumers, “The Skin Type Solution” (New York: Bantam Dell, 2006). Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems LLC. Write to her at dermnews@mdedge.com. She has no relevant disclosures.

References

1. Liu L et al. Snails and skin care – an uncovered combination. JAMA Dermatol. 2017 Jul 1;153(7):650.

2. Nguyen JK et al. J Cosmet Dermatol. 2020 Feb 26. doi: 10.1111/jocd.13344.

3. Juhász ML et al. J Cosmet Dermatol. 2018;17(3):305-12.

4. Pitt SJ et al. Br J Biomed Sci. 2015;72(4):174-81.

5. Brieva A et al. Skin Pharmacol Physiol. 2008;21(1):15-22.

6. Cruz MC et al. Int J Cosmet Sci. 2012 Apr;34(2):183-9.

7. Fabi SG et al. J Drugs Dermatol. 2013; Apr;12(4):453-7.

8. Ledo E et al. Radioproteccion. 1999;23(7):34-8.

9. Oh M-Jet al. J Korean Med Ophthalmol Otolaryngol Dermatol. 2010; Dec,23(3):138-53.

10. Tsoutsos D et al. J Dermatolog Treat. 2009;20(4):219-22.

11. Tribo-Boixareu MJ et al. Cosmet Dermatol. 2009;22(5):247-52.

12. Ellijimi C et al. Biomed Pharmacother. 2018 May;101:871-80.

13. Espada J et al. Int J Cosmet Sci. 2015 Feb;37(1):41-55.

14. Quay ER et al. J Drugs Dermatol. 2017 Apr 1;16(4):358-63.
 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap

Sericin, a versatile silk protein, has multiple potential roles in dermatology

Article Type
Changed
Fri, 05/15/2020 - 08:51

Inexpensively obtained as a silk industry by-product, sericin is a glycoprotein found to confer various biologic effects.1 The globular protein sericin has also long been known to exhibit antityrosinase and immunomodulatory activities.2,3 This column focuses on the wide range of emerging and potential applications of sericin in cutaneous treatments.

Protection against solar radiation and photoaging

Tasar Silkworm with cocoon
Sailesh Patnaik/CCA-SA 4.0 International

Studies in mice to evaluate the potential antioxidant and skin-protective effects of sericin by Zhaorigetu et al. in 2003 revealed that, by diminishing oxidative stress, cyclooxygenase-2 protein, and cell proliferation, sericin exerted a photoprotective effect against acute harm and tumor promotion elicited by UVB.4

Using mouse skin models, Dash et al. showed in 2008 that the silk protein sericin derived from the tropical tasar silkworm is a robust antioxidant and photoprotective agent, displaying a capacity to block UVB-induced apoptosis in irradiated (30 mJ/cm2 UVB) human keratinocytes and, as compared with the mulberry silkworm, yielding protection against oxidative stress.5,6

In 2015, Berardesca et al. conducted a randomized, double-blind, vehicle-controlled, split-face study over 8 weeks in 40 women (ages 40-70 years) to assess the antiaging effects of topically applied combination therapy including gold silk sericin, niacinamide, and signaline. The investigators observed significant improvements in stratum corneum hydration, barrier function, skin elasticity, and roughness as compared with skin treated with the control formulation. They concluded that this combination formulation featuring gold silk sericin warrants attention in the arsenal for ameliorating signs of aging female facial skin.7

A year earlier, Aramwit and Bang introduced a bacterial nanocellulose gel shown to effectively release silk sericin for facial treatment. Formulated at a pH of 4.5, the bioactive mask exhibited an ultrafine and pure fiber network structure. The authors noted that the gel was less adhesive than the commercially available paper mask, while the silk sericin product displayed greater moisture absorption capacity. In vitro cytotoxicity assessments also revealed that the product is safe for facial treatments.8

Cosmeceutical antioxidant for hyperpigmentation

In 2019, Kumar et al. demonstrated the inhibitory effect of topically applied silk sericin derived from Antheraea assamensis against UV-induced melanogenesis in mouse melanoma. They suggested that the formulation shows promise as a cosmeceutical antioxidant agent designed to address hyperpigmentation.3

The previous year, Aramwit et al. demonstrated using an in vitro model that urea-extracted sericin displays a capacity to inhibit melanogenesis by hindering tyrosinase activity, attenuating inflammation and allergic reactions, and reducing the expression of microphthalmia-associated transcription factor, a marker of melanogenesis regulation, in melanocytes and keratinocytes.2

Potential use as an adjunct psoriasis treatment

A combination of naringin (from Citrus maxima) and sericin (from Bombyx mori) was evaluated in 2019 by Deenonpoe et al. for the treatment of psoriasis. They isolated human peripheral blood mononuclear cells from 10 healthy subjects and 10 patients with psoriasis. The combination formulation was much more effective than either compound alone in significantly reducing mRNA expression and the synthesis of proinflammatory cytokines in samples from psoriasis patients. The investigators concluded that the down-regulation of proinflammatory cytokines imparted by the naringin/sericin product points toward its possible clinical use as a complementary treatment for psoriasis and other inflammation-mediated conditions.9

 

 

Uremic pruritus and burn wounds

A randomized, double-blind, placebo-controlled 6-week study in 2012 conducted by Aramwit et al. assessed the use of sericin cream versus a cream base placebo in the treatment of uremic pruritus in 50 hemodialysis patients, 47 of whom completed the study. Significant differences in the creams were identified, with hydration vastly improved in patients using the sericin cream. Significant reductions in pruritus and dyspigmentation were also observed in the treatment group, with an overall quality of life improvement noted in relation to pain score.10

A tasar silk moth caterpillar.
Gail Hampshire/Wikimedia Commons CC BY 2.0

The ensuing year, Aramwit et al. showed that silk sericin promoted wound healing in vitro and, when added to silver sulfadiazine cream and evaluated in a randomized, double-blind, standard-controlled study, demonstrated clinical efficacy in healing burn wounds.11

Wound healing

An expanding body of research suggests the role of sericin in wound healing. In 2007, Aramwit et al. found that sericin, which boasts notable hydrophilic qualities, was effective as a wound-healing agent in rats. The tested sericin cream successfully reduced wound size and wound healing time was substantially shorter than in animals treated with control formula. Treatment for 15 days yielded complete healing, no ulceration, and higher collagen levels, as determined by histologic examination, in comparison with control.12 Other studies using sericin hydrogel as well as a sericin-based nanofibrous matrix with chitosan have demonstrated success in wound healing in mice.13,14

Human studies

In 2018, Napavichayanun et al. reported on the clinical efficacy and safety of bacterial cellulose wound dressings including silk sericin and PHMB as compared with Bactigras (an antiseptic dressing) as a control in split-thickness skin graft donor-site wound treatment. In this single-blinded, randomized, controlled study of 21 patients, pain scores were significantly lower and wound quality higher in the skin treated with the sericin product. The test formulation was protected against infection without inducing adverse effects.15

Previously, a silk sericin–releasing wound dressing introduced in 2014 was found to significantly diminish pain and promote more rapid healing in patients with split-thickness skin graft donor sites as compared with treatment with the Bactigras wound dressing.16

Sericin in tissue repair and as a drug delivery carrier

Sericin is associated with antioxidant and moisturizing properties as well as a mitogenic influence on mammalian cells, with a particular impact on keratinocytes and fibroblasts that render it useful in biomaterials designed for skin tissue repair.17

Dr. Leslie S. Baumann

Wang et al. have cross-linked dialdehyde carboxymethyl cellulose with silk sericin derived from the B. mori cocoon to develop a film with impressive blood compatibility and cytocompatibility that shows potential for use as a wound dressing, artificial skin, and in tissue engineering.18

Similarly, Liang et al. have been successful in preparing a medical tissue glue incorporating a gelatin, sericin, and carboxymethyl chitosan blend solution, cross-linked with 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide. The tissue glue has been found to offer notable biocompatibility and structural traits at low cost.19

Sericin protein also evinces potential as a biocompatible, bioviable carrier for drug delivery. Suktham et al. showed that resveratrol-loaded sericin nanoparticles robustly hindered growth of colorectal adenocarcinoma cells while cytotoxic to skin fibroblasts, suggesting the viability or potential of sericin nanoparticles as bionanocarriers in a drug delivery system.20 In addition, Tao et al. found silk sericin to be effective when blended with poly(vinyl alcohol) in a hydrogel with antibacterial properties as a drug delivery carrier with potential for use as wound dressing.21

Conclusion

There is a plethora of new evidence to justify the inclusion of sericin in dermatologic research and skin care, specifically wound care. Much more research is necessary, though, to explore how the antioxidant and moisturizing activities of the protein may be harnessed to confer skin-protective effects, especially against UV damage.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann wrote two textbooks: “Cosmetic Dermatology: Principles and Practice” (New York: McGraw-Hill, 2002), and “Cosmeceuticals and Cosmetic Ingredients” (New York: McGraw-Hill, 2014), and a New York Times Best Sellers book for consumers,“The Skin Type Solution” (New York: Bantam Dell, 2006). Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems. Write to her at dermnews@mdedge.com

References

1. Lamboni L et al. Biotechnol Adv. 2015 Dec;33(8):1855-67.

2. Aramwit P et al. Biol Res. 2018 Nov 29;51(1):54.

3. Kumar JP, Mandal BB. Photochem Photobiol Sci. 2019 Oct 9:18(10):2497-508.

4. Zhaorigetu S et al. J Photochem Photobiol B. 2003 Oct 15;71(1-3):11-7.

5. Dash R et al. Mol Cell Biochem. 2008 Apr;311(1-2):111-9.

6. Dash R et al. BMB Rep. 2008 Mar 31;41(3):236-41.

7. Berardesca E et al. Int J Cosmet Sci. 2015 Dec;37(6):606-12.

8. Aramwit P, Bang N. BMC Biotechnol. 2014 Dec 9;14:104.

9. Deenonpoe R et al. BMC Complement Altern Med. 2019 Jul 10;19(1):168.

10. Aramwit P et al. BMC Nephrol. 2012 Sep 24;13:119.

11. Aramwit P et al. Arch Dermatol Res. 2013 Sep;305(7):585-94.

12. Aramwit P, Sangcakul A. Biosci Biotechnol Biochem. 2007 Oct;71(10):2473-7.

13. Qi C et al. Biomater Sci. 2018 Nov 1;6(11):2859-70.

14. Sapru S et al. Acta Biomater. 2018 Sep 15;78:137-50.

15. Napavichayanun S et al. Arch Dermatol Res. 2018 Dec;310(10):795-805.

16. Siritientong T et al. Pharm Res. 2014 Jan;31(1):104-16.

17. Lamboni L et al. Biotechnol Adv. 2015 Dec;33(8):1855-67.

18. Wang P et al. Carbohydr Polym. 2019 May 15;212:403-11.

19. Liang M et al. J Appl Biomater Funct Mater. 2018 Apr;16(2):97-106.

20. Suktham K et al. Int J Pharm. 2018 Feb 15;537(1-2):48-56.

21. Tao G et al. Mater Sci Eng C Mater Biol Appl. 2019 Aug;101:341-51.

Publications
Topics
Sections

Inexpensively obtained as a silk industry by-product, sericin is a glycoprotein found to confer various biologic effects.1 The globular protein sericin has also long been known to exhibit antityrosinase and immunomodulatory activities.2,3 This column focuses on the wide range of emerging and potential applications of sericin in cutaneous treatments.

Protection against solar radiation and photoaging

Tasar Silkworm with cocoon
Sailesh Patnaik/CCA-SA 4.0 International

Studies in mice to evaluate the potential antioxidant and skin-protective effects of sericin by Zhaorigetu et al. in 2003 revealed that, by diminishing oxidative stress, cyclooxygenase-2 protein, and cell proliferation, sericin exerted a photoprotective effect against acute harm and tumor promotion elicited by UVB.4

Using mouse skin models, Dash et al. showed in 2008 that the silk protein sericin derived from the tropical tasar silkworm is a robust antioxidant and photoprotective agent, displaying a capacity to block UVB-induced apoptosis in irradiated (30 mJ/cm2 UVB) human keratinocytes and, as compared with the mulberry silkworm, yielding protection against oxidative stress.5,6

In 2015, Berardesca et al. conducted a randomized, double-blind, vehicle-controlled, split-face study over 8 weeks in 40 women (ages 40-70 years) to assess the antiaging effects of topically applied combination therapy including gold silk sericin, niacinamide, and signaline. The investigators observed significant improvements in stratum corneum hydration, barrier function, skin elasticity, and roughness as compared with skin treated with the control formulation. They concluded that this combination formulation featuring gold silk sericin warrants attention in the arsenal for ameliorating signs of aging female facial skin.7

A year earlier, Aramwit and Bang introduced a bacterial nanocellulose gel shown to effectively release silk sericin for facial treatment. Formulated at a pH of 4.5, the bioactive mask exhibited an ultrafine and pure fiber network structure. The authors noted that the gel was less adhesive than the commercially available paper mask, while the silk sericin product displayed greater moisture absorption capacity. In vitro cytotoxicity assessments also revealed that the product is safe for facial treatments.8

Cosmeceutical antioxidant for hyperpigmentation

In 2019, Kumar et al. demonstrated the inhibitory effect of topically applied silk sericin derived from Antheraea assamensis against UV-induced melanogenesis in mouse melanoma. They suggested that the formulation shows promise as a cosmeceutical antioxidant agent designed to address hyperpigmentation.3

The previous year, Aramwit et al. demonstrated using an in vitro model that urea-extracted sericin displays a capacity to inhibit melanogenesis by hindering tyrosinase activity, attenuating inflammation and allergic reactions, and reducing the expression of microphthalmia-associated transcription factor, a marker of melanogenesis regulation, in melanocytes and keratinocytes.2

Potential use as an adjunct psoriasis treatment

A combination of naringin (from Citrus maxima) and sericin (from Bombyx mori) was evaluated in 2019 by Deenonpoe et al. for the treatment of psoriasis. They isolated human peripheral blood mononuclear cells from 10 healthy subjects and 10 patients with psoriasis. The combination formulation was much more effective than either compound alone in significantly reducing mRNA expression and the synthesis of proinflammatory cytokines in samples from psoriasis patients. The investigators concluded that the down-regulation of proinflammatory cytokines imparted by the naringin/sericin product points toward its possible clinical use as a complementary treatment for psoriasis and other inflammation-mediated conditions.9

 

 

Uremic pruritus and burn wounds

A randomized, double-blind, placebo-controlled 6-week study in 2012 conducted by Aramwit et al. assessed the use of sericin cream versus a cream base placebo in the treatment of uremic pruritus in 50 hemodialysis patients, 47 of whom completed the study. Significant differences in the creams were identified, with hydration vastly improved in patients using the sericin cream. Significant reductions in pruritus and dyspigmentation were also observed in the treatment group, with an overall quality of life improvement noted in relation to pain score.10

A tasar silk moth caterpillar.
Gail Hampshire/Wikimedia Commons CC BY 2.0

The ensuing year, Aramwit et al. showed that silk sericin promoted wound healing in vitro and, when added to silver sulfadiazine cream and evaluated in a randomized, double-blind, standard-controlled study, demonstrated clinical efficacy in healing burn wounds.11

Wound healing

An expanding body of research suggests the role of sericin in wound healing. In 2007, Aramwit et al. found that sericin, which boasts notable hydrophilic qualities, was effective as a wound-healing agent in rats. The tested sericin cream successfully reduced wound size and wound healing time was substantially shorter than in animals treated with control formula. Treatment for 15 days yielded complete healing, no ulceration, and higher collagen levels, as determined by histologic examination, in comparison with control.12 Other studies using sericin hydrogel as well as a sericin-based nanofibrous matrix with chitosan have demonstrated success in wound healing in mice.13,14

Human studies

In 2018, Napavichayanun et al. reported on the clinical efficacy and safety of bacterial cellulose wound dressings including silk sericin and PHMB as compared with Bactigras (an antiseptic dressing) as a control in split-thickness skin graft donor-site wound treatment. In this single-blinded, randomized, controlled study of 21 patients, pain scores were significantly lower and wound quality higher in the skin treated with the sericin product. The test formulation was protected against infection without inducing adverse effects.15

Previously, a silk sericin–releasing wound dressing introduced in 2014 was found to significantly diminish pain and promote more rapid healing in patients with split-thickness skin graft donor sites as compared with treatment with the Bactigras wound dressing.16

Sericin in tissue repair and as a drug delivery carrier

Sericin is associated with antioxidant and moisturizing properties as well as a mitogenic influence on mammalian cells, with a particular impact on keratinocytes and fibroblasts that render it useful in biomaterials designed for skin tissue repair.17

Dr. Leslie S. Baumann

Wang et al. have cross-linked dialdehyde carboxymethyl cellulose with silk sericin derived from the B. mori cocoon to develop a film with impressive blood compatibility and cytocompatibility that shows potential for use as a wound dressing, artificial skin, and in tissue engineering.18

Similarly, Liang et al. have been successful in preparing a medical tissue glue incorporating a gelatin, sericin, and carboxymethyl chitosan blend solution, cross-linked with 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide. The tissue glue has been found to offer notable biocompatibility and structural traits at low cost.19

Sericin protein also evinces potential as a biocompatible, bioviable carrier for drug delivery. Suktham et al. showed that resveratrol-loaded sericin nanoparticles robustly hindered growth of colorectal adenocarcinoma cells while cytotoxic to skin fibroblasts, suggesting the viability or potential of sericin nanoparticles as bionanocarriers in a drug delivery system.20 In addition, Tao et al. found silk sericin to be effective when blended with poly(vinyl alcohol) in a hydrogel with antibacterial properties as a drug delivery carrier with potential for use as wound dressing.21

Conclusion

There is a plethora of new evidence to justify the inclusion of sericin in dermatologic research and skin care, specifically wound care. Much more research is necessary, though, to explore how the antioxidant and moisturizing activities of the protein may be harnessed to confer skin-protective effects, especially against UV damage.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann wrote two textbooks: “Cosmetic Dermatology: Principles and Practice” (New York: McGraw-Hill, 2002), and “Cosmeceuticals and Cosmetic Ingredients” (New York: McGraw-Hill, 2014), and a New York Times Best Sellers book for consumers,“The Skin Type Solution” (New York: Bantam Dell, 2006). Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems. Write to her at dermnews@mdedge.com

References

1. Lamboni L et al. Biotechnol Adv. 2015 Dec;33(8):1855-67.

2. Aramwit P et al. Biol Res. 2018 Nov 29;51(1):54.

3. Kumar JP, Mandal BB. Photochem Photobiol Sci. 2019 Oct 9:18(10):2497-508.

4. Zhaorigetu S et al. J Photochem Photobiol B. 2003 Oct 15;71(1-3):11-7.

5. Dash R et al. Mol Cell Biochem. 2008 Apr;311(1-2):111-9.

6. Dash R et al. BMB Rep. 2008 Mar 31;41(3):236-41.

7. Berardesca E et al. Int J Cosmet Sci. 2015 Dec;37(6):606-12.

8. Aramwit P, Bang N. BMC Biotechnol. 2014 Dec 9;14:104.

9. Deenonpoe R et al. BMC Complement Altern Med. 2019 Jul 10;19(1):168.

10. Aramwit P et al. BMC Nephrol. 2012 Sep 24;13:119.

11. Aramwit P et al. Arch Dermatol Res. 2013 Sep;305(7):585-94.

12. Aramwit P, Sangcakul A. Biosci Biotechnol Biochem. 2007 Oct;71(10):2473-7.

13. Qi C et al. Biomater Sci. 2018 Nov 1;6(11):2859-70.

14. Sapru S et al. Acta Biomater. 2018 Sep 15;78:137-50.

15. Napavichayanun S et al. Arch Dermatol Res. 2018 Dec;310(10):795-805.

16. Siritientong T et al. Pharm Res. 2014 Jan;31(1):104-16.

17. Lamboni L et al. Biotechnol Adv. 2015 Dec;33(8):1855-67.

18. Wang P et al. Carbohydr Polym. 2019 May 15;212:403-11.

19. Liang M et al. J Appl Biomater Funct Mater. 2018 Apr;16(2):97-106.

20. Suktham K et al. Int J Pharm. 2018 Feb 15;537(1-2):48-56.

21. Tao G et al. Mater Sci Eng C Mater Biol Appl. 2019 Aug;101:341-51.

Inexpensively obtained as a silk industry by-product, sericin is a glycoprotein found to confer various biologic effects.1 The globular protein sericin has also long been known to exhibit antityrosinase and immunomodulatory activities.2,3 This column focuses on the wide range of emerging and potential applications of sericin in cutaneous treatments.

Protection against solar radiation and photoaging

Tasar Silkworm with cocoon
Sailesh Patnaik/CCA-SA 4.0 International

Studies in mice to evaluate the potential antioxidant and skin-protective effects of sericin by Zhaorigetu et al. in 2003 revealed that, by diminishing oxidative stress, cyclooxygenase-2 protein, and cell proliferation, sericin exerted a photoprotective effect against acute harm and tumor promotion elicited by UVB.4

Using mouse skin models, Dash et al. showed in 2008 that the silk protein sericin derived from the tropical tasar silkworm is a robust antioxidant and photoprotective agent, displaying a capacity to block UVB-induced apoptosis in irradiated (30 mJ/cm2 UVB) human keratinocytes and, as compared with the mulberry silkworm, yielding protection against oxidative stress.5,6

In 2015, Berardesca et al. conducted a randomized, double-blind, vehicle-controlled, split-face study over 8 weeks in 40 women (ages 40-70 years) to assess the antiaging effects of topically applied combination therapy including gold silk sericin, niacinamide, and signaline. The investigators observed significant improvements in stratum corneum hydration, barrier function, skin elasticity, and roughness as compared with skin treated with the control formulation. They concluded that this combination formulation featuring gold silk sericin warrants attention in the arsenal for ameliorating signs of aging female facial skin.7

A year earlier, Aramwit and Bang introduced a bacterial nanocellulose gel shown to effectively release silk sericin for facial treatment. Formulated at a pH of 4.5, the bioactive mask exhibited an ultrafine and pure fiber network structure. The authors noted that the gel was less adhesive than the commercially available paper mask, while the silk sericin product displayed greater moisture absorption capacity. In vitro cytotoxicity assessments also revealed that the product is safe for facial treatments.8

Cosmeceutical antioxidant for hyperpigmentation

In 2019, Kumar et al. demonstrated the inhibitory effect of topically applied silk sericin derived from Antheraea assamensis against UV-induced melanogenesis in mouse melanoma. They suggested that the formulation shows promise as a cosmeceutical antioxidant agent designed to address hyperpigmentation.3

The previous year, Aramwit et al. demonstrated using an in vitro model that urea-extracted sericin displays a capacity to inhibit melanogenesis by hindering tyrosinase activity, attenuating inflammation and allergic reactions, and reducing the expression of microphthalmia-associated transcription factor, a marker of melanogenesis regulation, in melanocytes and keratinocytes.2

Potential use as an adjunct psoriasis treatment

A combination of naringin (from Citrus maxima) and sericin (from Bombyx mori) was evaluated in 2019 by Deenonpoe et al. for the treatment of psoriasis. They isolated human peripheral blood mononuclear cells from 10 healthy subjects and 10 patients with psoriasis. The combination formulation was much more effective than either compound alone in significantly reducing mRNA expression and the synthesis of proinflammatory cytokines in samples from psoriasis patients. The investigators concluded that the down-regulation of proinflammatory cytokines imparted by the naringin/sericin product points toward its possible clinical use as a complementary treatment for psoriasis and other inflammation-mediated conditions.9

 

 

Uremic pruritus and burn wounds

A randomized, double-blind, placebo-controlled 6-week study in 2012 conducted by Aramwit et al. assessed the use of sericin cream versus a cream base placebo in the treatment of uremic pruritus in 50 hemodialysis patients, 47 of whom completed the study. Significant differences in the creams were identified, with hydration vastly improved in patients using the sericin cream. Significant reductions in pruritus and dyspigmentation were also observed in the treatment group, with an overall quality of life improvement noted in relation to pain score.10

A tasar silk moth caterpillar.
Gail Hampshire/Wikimedia Commons CC BY 2.0

The ensuing year, Aramwit et al. showed that silk sericin promoted wound healing in vitro and, when added to silver sulfadiazine cream and evaluated in a randomized, double-blind, standard-controlled study, demonstrated clinical efficacy in healing burn wounds.11

Wound healing

An expanding body of research suggests the role of sericin in wound healing. In 2007, Aramwit et al. found that sericin, which boasts notable hydrophilic qualities, was effective as a wound-healing agent in rats. The tested sericin cream successfully reduced wound size and wound healing time was substantially shorter than in animals treated with control formula. Treatment for 15 days yielded complete healing, no ulceration, and higher collagen levels, as determined by histologic examination, in comparison with control.12 Other studies using sericin hydrogel as well as a sericin-based nanofibrous matrix with chitosan have demonstrated success in wound healing in mice.13,14

Human studies

In 2018, Napavichayanun et al. reported on the clinical efficacy and safety of bacterial cellulose wound dressings including silk sericin and PHMB as compared with Bactigras (an antiseptic dressing) as a control in split-thickness skin graft donor-site wound treatment. In this single-blinded, randomized, controlled study of 21 patients, pain scores were significantly lower and wound quality higher in the skin treated with the sericin product. The test formulation was protected against infection without inducing adverse effects.15

Previously, a silk sericin–releasing wound dressing introduced in 2014 was found to significantly diminish pain and promote more rapid healing in patients with split-thickness skin graft donor sites as compared with treatment with the Bactigras wound dressing.16

Sericin in tissue repair and as a drug delivery carrier

Sericin is associated with antioxidant and moisturizing properties as well as a mitogenic influence on mammalian cells, with a particular impact on keratinocytes and fibroblasts that render it useful in biomaterials designed for skin tissue repair.17

Dr. Leslie S. Baumann

Wang et al. have cross-linked dialdehyde carboxymethyl cellulose with silk sericin derived from the B. mori cocoon to develop a film with impressive blood compatibility and cytocompatibility that shows potential for use as a wound dressing, artificial skin, and in tissue engineering.18

Similarly, Liang et al. have been successful in preparing a medical tissue glue incorporating a gelatin, sericin, and carboxymethyl chitosan blend solution, cross-linked with 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide. The tissue glue has been found to offer notable biocompatibility and structural traits at low cost.19

Sericin protein also evinces potential as a biocompatible, bioviable carrier for drug delivery. Suktham et al. showed that resveratrol-loaded sericin nanoparticles robustly hindered growth of colorectal adenocarcinoma cells while cytotoxic to skin fibroblasts, suggesting the viability or potential of sericin nanoparticles as bionanocarriers in a drug delivery system.20 In addition, Tao et al. found silk sericin to be effective when blended with poly(vinyl alcohol) in a hydrogel with antibacterial properties as a drug delivery carrier with potential for use as wound dressing.21

Conclusion

There is a plethora of new evidence to justify the inclusion of sericin in dermatologic research and skin care, specifically wound care. Much more research is necessary, though, to explore how the antioxidant and moisturizing activities of the protein may be harnessed to confer skin-protective effects, especially against UV damage.

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann wrote two textbooks: “Cosmetic Dermatology: Principles and Practice” (New York: McGraw-Hill, 2002), and “Cosmeceuticals and Cosmetic Ingredients” (New York: McGraw-Hill, 2014), and a New York Times Best Sellers book for consumers,“The Skin Type Solution” (New York: Bantam Dell, 2006). Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems. Write to her at dermnews@mdedge.com

References

1. Lamboni L et al. Biotechnol Adv. 2015 Dec;33(8):1855-67.

2. Aramwit P et al. Biol Res. 2018 Nov 29;51(1):54.

3. Kumar JP, Mandal BB. Photochem Photobiol Sci. 2019 Oct 9:18(10):2497-508.

4. Zhaorigetu S et al. J Photochem Photobiol B. 2003 Oct 15;71(1-3):11-7.

5. Dash R et al. Mol Cell Biochem. 2008 Apr;311(1-2):111-9.

6. Dash R et al. BMB Rep. 2008 Mar 31;41(3):236-41.

7. Berardesca E et al. Int J Cosmet Sci. 2015 Dec;37(6):606-12.

8. Aramwit P, Bang N. BMC Biotechnol. 2014 Dec 9;14:104.

9. Deenonpoe R et al. BMC Complement Altern Med. 2019 Jul 10;19(1):168.

10. Aramwit P et al. BMC Nephrol. 2012 Sep 24;13:119.

11. Aramwit P et al. Arch Dermatol Res. 2013 Sep;305(7):585-94.

12. Aramwit P, Sangcakul A. Biosci Biotechnol Biochem. 2007 Oct;71(10):2473-7.

13. Qi C et al. Biomater Sci. 2018 Nov 1;6(11):2859-70.

14. Sapru S et al. Acta Biomater. 2018 Sep 15;78:137-50.

15. Napavichayanun S et al. Arch Dermatol Res. 2018 Dec;310(10):795-805.

16. Siritientong T et al. Pharm Res. 2014 Jan;31(1):104-16.

17. Lamboni L et al. Biotechnol Adv. 2015 Dec;33(8):1855-67.

18. Wang P et al. Carbohydr Polym. 2019 May 15;212:403-11.

19. Liang M et al. J Appl Biomater Funct Mater. 2018 Apr;16(2):97-106.

20. Suktham K et al. Int J Pharm. 2018 Feb 15;537(1-2):48-56.

21. Tao G et al. Mater Sci Eng C Mater Biol Appl. 2019 Aug;101:341-51.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap

Mother of pearl: The power of pearl powder

Article Type
Changed
Wed, 04/15/2020 - 13:42

Pearl powder has been used for thousands of years in traditional Chinese medicine, as well as in cosmetics and as health food supplements in China and Taiwan.1,2 Because of its dense protein and mineral composition, it has been used to treat several skin and bone disorders, as well as palpitations, insomnia, and epilepsy.3,4 The pearl-farming industry itself was established in Japan and has existed for more than a century; today, pearls are cultured globally and continue to receive attention for conferring health benefits.5

White Mother Of Pearl Pinctada maxima oyster
Vladyslav Danilin/iStock/Getty Images

Calcium carbonate is the primary component of mollusk shells (roughly 95%), with the remainder an organic matrix including proteins, glycoproteins, and polysaccharides.6 Pearl powder is known to have exhibited antiaging, antioxidant, antiradiative, and tonic activities; in recent years, it has been incorporated into health foods for such properties and used in the clinical setting to treat ulcers (aphthous, gastric, and duodenal).4,7 Consisting of multiple active proteins, pearl powder is thought to be conducive to skin cell growth and effective for wound repair.4 This column focuses on recent research into the dermatologic potential of the powder derived from mother of pearl.
 

Wound healing

A decade ago, Jian-Ping et al. showed in mice that the water-soluble matrix of pearl powder (Hyriopsis cumingii) could significantly induce oral fibroblast proliferation and collagen accumulation, suppress matrix metalloproteinase-2 activity, and significantly foster TIMP-1 synthesis. The investigators concluded that the wound healing facilitated by pearl powder derives, in part, from its capacity to promote fibroblast mitosis, collagen deposition, and production of TIMP-1.8

Two years later, Lee et al. evaluated the effects of water-soluble nacre (mother of pearl) on second-degree burn wound healing in porcine skin as a proxy for human skin. They found that its application quickly led to burn-induced granulation areas filling with collagen, with normal skin appearance restored to wounded dermis and epidermis. Angiogenesis was also promoted by water-soluble nacre as was wound recovery in areas with apoptotic and necrotic cellular damage. Murine fibroblast NIH3T3 cells treated with water-soluble nacre also demonstrated augmented proliferation and collagen production. The researchers cited the restoration of angiogenesis and fibroblast activity as the primary benefits of water-soluble nacre, suggesting its potential as a wound therapy, preferable to powdered nacre due to better biocompatibility with less discomfort.9

The next year, Li et al. found that mother of pearl extract promoted cell migration of fibroblasts in cell culture, demonstrating its potential as a wound healing model.7In 2019, Chen et al. studied the effects of pearl powders of varying particle sizes to treat wounds in vitro and in vivo. They found that micro- and nanosized pearl powders augmented proliferation and migration of skin cells and shortened wound closure time. All powders also improved the biomechanical strength of healed skin, enhanced collagen formation and deposition, and expanded cutaneous angiogenesis, with nanoscale pearl powder displaying greatest efficiency.4

Skin tone and atopic dermatitis

In 2000, Lopez et al. implanted powdered nacre (mother of pearl derived from Pinctada maxima), which can promote and regulate bone-forming cells, into rat dermis to evaluate its effects on skin fibroblasts. They noted that the implant yielded well-vascularized tissue and improved extracellular matrix production, synthesis of substances involved in cellular adhesion and communication, and tissue regeneration (such as collagen types I and III). The investigators concluded that the powdered nacre contributed to the conditions necessary for improved skin tone and proper physiologic functioning of the skin.10

Rousseau et al. extracted lipids from the nacre of the oyster P. margaritifera to test on artificially dehydrated skin explants with the intention of developing new treatments for atopic dermatitis. The researchers determined that the lipids spurred a reconstitution of the intercellular material of the stratum corneum, concluding that new products to treat atopic dermatitis might be based on the signaling activity of nacre lipids.11

Antifibrotic and anti-inflammatory activity

A 2015 study by Yang et al. showed that a room-temperature superextraction system to yield the main active constituents of pearl was successful in enhancing their anti-inflammatory and antiapoptotic activity in human keratinocyte cells (HaCaT) exposed to low-dose UVB. The investigators combined pearl extract and poly (gamma-glutamic acid) hydrogels and observed reductions in inflammation and apoptosis of HaCaT cells. They concluded that a marketed pearl extract may be able to suppress radiation dermatitis present in keratinocytes.12

Dr. Leslie S. Baumann

Two years later, Latire et al. used human dermal fibroblasts in primary culture to assess the potential biological activities of the matrix macromolecular components extracted from the shells of two edible mollusks (the blue mussel Mytilus edulis and the Pacific oyster Crassostrea gigas). The investigators found that both extracts influenced metabolic functions of the cells and reduced type I collagen levels, with an associated rise in matrix metalloproteinase-1 activity. Given their findings implying the effectiveness of the extracts in facilitating the catabolic pathway of human dermal fibroblasts, the authors suggest that these shell matrices present the potential for use in treating fibrosis, especially for scleroderma.6

Antioxidant and antiaging activity

Shao et al. demonstrated 10 years ago that pearl powder provides a moisturizing effect on the skin, with ultramicro pearl powder delivering a more robust moisturizing result than water-soluble pearl powder. These two types of pearl powder, along with another one tested (ultranano pearl powder), also significantly diminished the activation of tyrosinase and free radicals. Water-soluble pearl powder did not perform as well as the other two formulations in free radical scavenging. The investigators suggested that their results support the use of pearl powder to combat aging and enhance beauty, and could be used in the clinical setting.13

In 2017, Yang et al. reported on the in vitro antihemolytic and antioxidant activity of pearl powder in shielding human erythrocytes against 2,2’-azobis(2-amidinopropane) dihydrochloride–induced oxidative damage to membrane proteins/lipids. The researchers contend that the strong antioxidant qualities of pearl powder could be applied to prevent or protect against various diseases resulting from free radical damage.2

Human trials: Antioxidant, antiaging, skin appearance

Chiu et al. studied the antioxidant activity of various pearl powder extracts in a randomized, placebo-controlled trial in 2018. They also investigated the life span–prolonging effects of the powders using wild-type Caenorhabditis elegans. Twenty healthy middle-aged subjects were separated into two groups (experimental and placebo), with 3 g of pearl powder administered in capsules to the former and 3 g of placebo to the latter over 8 weeks. Blood samples taken at the beginning and every 2 weeks during the trial and in the 10th week revealed maximum antioxidant activity of the pearl powder and prolongation of C. elegans lifespan by 18.87%. Subjects using pearl powder demonstrated significant increases in total antioxidant capacity, thiols, glutathione, and enzymic antioxidant activity, along with notably inhibited lipid peroxidation products. The investigators concluded that pearl powder extract acted as a potent antioxidant and its use may be warranted to treat degenerative conditions related to aging.3

A recent study of the perception of blue light on Korean women’s faces using blue pearl pigment revealed that the pigment does indeed elicit the perception of the blue-light effect, notably transparency and gloss, which is particularly valued in Korea.14

Conclusion

The use of mother of pearl and pearl powder in traditional Chinese medicine and as a cosmetic and food additive has a rich and lengthy history. Contemporary research clearly suggests interesting avenues for further investigation and some promising results. Much more research is necessary, though, to delineate the potential roles of pearl powder in the skin care arsenal.
 

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann wrote two textbooks: “Cosmetic Dermatology: Principles and Practice” (New York: McGraw-Hill, 2002), and “Cosmeceuticals and Cosmetic Ingredients” (New York: McGraw-Hill, 2014), and a New York Times Best Sellers book for consumers, “The Skin Type Solution” (New York: Bantam Dell, 2006). Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems. Write to her at dermnews@mdedge.com

References

1. Zhang J et al. J Sep Sci. 2015 May;38(9):1552-60.

2. Yang HL et al. J Food Drug Anal. 2017 Oct;25(4):898-907.

3. Chiu HF et al. J Food Drug Anal. 2018 Jan;26(1):309-17.

4. Chen X et al. Drug Dev Ind Pharm. 2019 Jun;45(6):1009-16.

5. Nagai K. Zoolog Sci. 2013 Oct;30(10):783-93.

6. Latire T et al. Cytotechnology. 2017 Oct;69(5):815-29.

7. Li YC et al. Pharm Biol. 2013 Mar;51(3):289-97.

8. Jian-Ping D et al. Pharm Biol. 2010 Feb;48(2):122-7.

9. Lee K et al. Mol Biol Rep. 2012 Mar;39(3):3211-8.

10. Lopez E et al. Tissue Cell. 2000 Feb;32(1):95-101.

11. Rousseau M et al. Comp Biochem Physiol B Biochem Mol Biol. 2006 Sep;145(1):1-9.

12. Yang YL et al. Biomed Mater Eng. 2015;26 Suppl 1:S139-45.

13. Shao DZ et al. J Cosmet Sci. 2010 Mar-Apr;61(2):133-45.

14. Lee M et al. Skin Res Technol. 2020 Jan;26(1):76-80.

Publications
Topics
Sections

Pearl powder has been used for thousands of years in traditional Chinese medicine, as well as in cosmetics and as health food supplements in China and Taiwan.1,2 Because of its dense protein and mineral composition, it has been used to treat several skin and bone disorders, as well as palpitations, insomnia, and epilepsy.3,4 The pearl-farming industry itself was established in Japan and has existed for more than a century; today, pearls are cultured globally and continue to receive attention for conferring health benefits.5

White Mother Of Pearl Pinctada maxima oyster
Vladyslav Danilin/iStock/Getty Images

Calcium carbonate is the primary component of mollusk shells (roughly 95%), with the remainder an organic matrix including proteins, glycoproteins, and polysaccharides.6 Pearl powder is known to have exhibited antiaging, antioxidant, antiradiative, and tonic activities; in recent years, it has been incorporated into health foods for such properties and used in the clinical setting to treat ulcers (aphthous, gastric, and duodenal).4,7 Consisting of multiple active proteins, pearl powder is thought to be conducive to skin cell growth and effective for wound repair.4 This column focuses on recent research into the dermatologic potential of the powder derived from mother of pearl.
 

Wound healing

A decade ago, Jian-Ping et al. showed in mice that the water-soluble matrix of pearl powder (Hyriopsis cumingii) could significantly induce oral fibroblast proliferation and collagen accumulation, suppress matrix metalloproteinase-2 activity, and significantly foster TIMP-1 synthesis. The investigators concluded that the wound healing facilitated by pearl powder derives, in part, from its capacity to promote fibroblast mitosis, collagen deposition, and production of TIMP-1.8

Two years later, Lee et al. evaluated the effects of water-soluble nacre (mother of pearl) on second-degree burn wound healing in porcine skin as a proxy for human skin. They found that its application quickly led to burn-induced granulation areas filling with collagen, with normal skin appearance restored to wounded dermis and epidermis. Angiogenesis was also promoted by water-soluble nacre as was wound recovery in areas with apoptotic and necrotic cellular damage. Murine fibroblast NIH3T3 cells treated with water-soluble nacre also demonstrated augmented proliferation and collagen production. The researchers cited the restoration of angiogenesis and fibroblast activity as the primary benefits of water-soluble nacre, suggesting its potential as a wound therapy, preferable to powdered nacre due to better biocompatibility with less discomfort.9

The next year, Li et al. found that mother of pearl extract promoted cell migration of fibroblasts in cell culture, demonstrating its potential as a wound healing model.7In 2019, Chen et al. studied the effects of pearl powders of varying particle sizes to treat wounds in vitro and in vivo. They found that micro- and nanosized pearl powders augmented proliferation and migration of skin cells and shortened wound closure time. All powders also improved the biomechanical strength of healed skin, enhanced collagen formation and deposition, and expanded cutaneous angiogenesis, with nanoscale pearl powder displaying greatest efficiency.4

Skin tone and atopic dermatitis

In 2000, Lopez et al. implanted powdered nacre (mother of pearl derived from Pinctada maxima), which can promote and regulate bone-forming cells, into rat dermis to evaluate its effects on skin fibroblasts. They noted that the implant yielded well-vascularized tissue and improved extracellular matrix production, synthesis of substances involved in cellular adhesion and communication, and tissue regeneration (such as collagen types I and III). The investigators concluded that the powdered nacre contributed to the conditions necessary for improved skin tone and proper physiologic functioning of the skin.10

Rousseau et al. extracted lipids from the nacre of the oyster P. margaritifera to test on artificially dehydrated skin explants with the intention of developing new treatments for atopic dermatitis. The researchers determined that the lipids spurred a reconstitution of the intercellular material of the stratum corneum, concluding that new products to treat atopic dermatitis might be based on the signaling activity of nacre lipids.11

Antifibrotic and anti-inflammatory activity

A 2015 study by Yang et al. showed that a room-temperature superextraction system to yield the main active constituents of pearl was successful in enhancing their anti-inflammatory and antiapoptotic activity in human keratinocyte cells (HaCaT) exposed to low-dose UVB. The investigators combined pearl extract and poly (gamma-glutamic acid) hydrogels and observed reductions in inflammation and apoptosis of HaCaT cells. They concluded that a marketed pearl extract may be able to suppress radiation dermatitis present in keratinocytes.12

Dr. Leslie S. Baumann

Two years later, Latire et al. used human dermal fibroblasts in primary culture to assess the potential biological activities of the matrix macromolecular components extracted from the shells of two edible mollusks (the blue mussel Mytilus edulis and the Pacific oyster Crassostrea gigas). The investigators found that both extracts influenced metabolic functions of the cells and reduced type I collagen levels, with an associated rise in matrix metalloproteinase-1 activity. Given their findings implying the effectiveness of the extracts in facilitating the catabolic pathway of human dermal fibroblasts, the authors suggest that these shell matrices present the potential for use in treating fibrosis, especially for scleroderma.6

Antioxidant and antiaging activity

Shao et al. demonstrated 10 years ago that pearl powder provides a moisturizing effect on the skin, with ultramicro pearl powder delivering a more robust moisturizing result than water-soluble pearl powder. These two types of pearl powder, along with another one tested (ultranano pearl powder), also significantly diminished the activation of tyrosinase and free radicals. Water-soluble pearl powder did not perform as well as the other two formulations in free radical scavenging. The investigators suggested that their results support the use of pearl powder to combat aging and enhance beauty, and could be used in the clinical setting.13

In 2017, Yang et al. reported on the in vitro antihemolytic and antioxidant activity of pearl powder in shielding human erythrocytes against 2,2’-azobis(2-amidinopropane) dihydrochloride–induced oxidative damage to membrane proteins/lipids. The researchers contend that the strong antioxidant qualities of pearl powder could be applied to prevent or protect against various diseases resulting from free radical damage.2

Human trials: Antioxidant, antiaging, skin appearance

Chiu et al. studied the antioxidant activity of various pearl powder extracts in a randomized, placebo-controlled trial in 2018. They also investigated the life span–prolonging effects of the powders using wild-type Caenorhabditis elegans. Twenty healthy middle-aged subjects were separated into two groups (experimental and placebo), with 3 g of pearl powder administered in capsules to the former and 3 g of placebo to the latter over 8 weeks. Blood samples taken at the beginning and every 2 weeks during the trial and in the 10th week revealed maximum antioxidant activity of the pearl powder and prolongation of C. elegans lifespan by 18.87%. Subjects using pearl powder demonstrated significant increases in total antioxidant capacity, thiols, glutathione, and enzymic antioxidant activity, along with notably inhibited lipid peroxidation products. The investigators concluded that pearl powder extract acted as a potent antioxidant and its use may be warranted to treat degenerative conditions related to aging.3

A recent study of the perception of blue light on Korean women’s faces using blue pearl pigment revealed that the pigment does indeed elicit the perception of the blue-light effect, notably transparency and gloss, which is particularly valued in Korea.14

Conclusion

The use of mother of pearl and pearl powder in traditional Chinese medicine and as a cosmetic and food additive has a rich and lengthy history. Contemporary research clearly suggests interesting avenues for further investigation and some promising results. Much more research is necessary, though, to delineate the potential roles of pearl powder in the skin care arsenal.
 

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann wrote two textbooks: “Cosmetic Dermatology: Principles and Practice” (New York: McGraw-Hill, 2002), and “Cosmeceuticals and Cosmetic Ingredients” (New York: McGraw-Hill, 2014), and a New York Times Best Sellers book for consumers, “The Skin Type Solution” (New York: Bantam Dell, 2006). Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems. Write to her at dermnews@mdedge.com

References

1. Zhang J et al. J Sep Sci. 2015 May;38(9):1552-60.

2. Yang HL et al. J Food Drug Anal. 2017 Oct;25(4):898-907.

3. Chiu HF et al. J Food Drug Anal. 2018 Jan;26(1):309-17.

4. Chen X et al. Drug Dev Ind Pharm. 2019 Jun;45(6):1009-16.

5. Nagai K. Zoolog Sci. 2013 Oct;30(10):783-93.

6. Latire T et al. Cytotechnology. 2017 Oct;69(5):815-29.

7. Li YC et al. Pharm Biol. 2013 Mar;51(3):289-97.

8. Jian-Ping D et al. Pharm Biol. 2010 Feb;48(2):122-7.

9. Lee K et al. Mol Biol Rep. 2012 Mar;39(3):3211-8.

10. Lopez E et al. Tissue Cell. 2000 Feb;32(1):95-101.

11. Rousseau M et al. Comp Biochem Physiol B Biochem Mol Biol. 2006 Sep;145(1):1-9.

12. Yang YL et al. Biomed Mater Eng. 2015;26 Suppl 1:S139-45.

13. Shao DZ et al. J Cosmet Sci. 2010 Mar-Apr;61(2):133-45.

14. Lee M et al. Skin Res Technol. 2020 Jan;26(1):76-80.

Pearl powder has been used for thousands of years in traditional Chinese medicine, as well as in cosmetics and as health food supplements in China and Taiwan.1,2 Because of its dense protein and mineral composition, it has been used to treat several skin and bone disorders, as well as palpitations, insomnia, and epilepsy.3,4 The pearl-farming industry itself was established in Japan and has existed for more than a century; today, pearls are cultured globally and continue to receive attention for conferring health benefits.5

White Mother Of Pearl Pinctada maxima oyster
Vladyslav Danilin/iStock/Getty Images

Calcium carbonate is the primary component of mollusk shells (roughly 95%), with the remainder an organic matrix including proteins, glycoproteins, and polysaccharides.6 Pearl powder is known to have exhibited antiaging, antioxidant, antiradiative, and tonic activities; in recent years, it has been incorporated into health foods for such properties and used in the clinical setting to treat ulcers (aphthous, gastric, and duodenal).4,7 Consisting of multiple active proteins, pearl powder is thought to be conducive to skin cell growth and effective for wound repair.4 This column focuses on recent research into the dermatologic potential of the powder derived from mother of pearl.
 

Wound healing

A decade ago, Jian-Ping et al. showed in mice that the water-soluble matrix of pearl powder (Hyriopsis cumingii) could significantly induce oral fibroblast proliferation and collagen accumulation, suppress matrix metalloproteinase-2 activity, and significantly foster TIMP-1 synthesis. The investigators concluded that the wound healing facilitated by pearl powder derives, in part, from its capacity to promote fibroblast mitosis, collagen deposition, and production of TIMP-1.8

Two years later, Lee et al. evaluated the effects of water-soluble nacre (mother of pearl) on second-degree burn wound healing in porcine skin as a proxy for human skin. They found that its application quickly led to burn-induced granulation areas filling with collagen, with normal skin appearance restored to wounded dermis and epidermis. Angiogenesis was also promoted by water-soluble nacre as was wound recovery in areas with apoptotic and necrotic cellular damage. Murine fibroblast NIH3T3 cells treated with water-soluble nacre also demonstrated augmented proliferation and collagen production. The researchers cited the restoration of angiogenesis and fibroblast activity as the primary benefits of water-soluble nacre, suggesting its potential as a wound therapy, preferable to powdered nacre due to better biocompatibility with less discomfort.9

The next year, Li et al. found that mother of pearl extract promoted cell migration of fibroblasts in cell culture, demonstrating its potential as a wound healing model.7In 2019, Chen et al. studied the effects of pearl powders of varying particle sizes to treat wounds in vitro and in vivo. They found that micro- and nanosized pearl powders augmented proliferation and migration of skin cells and shortened wound closure time. All powders also improved the biomechanical strength of healed skin, enhanced collagen formation and deposition, and expanded cutaneous angiogenesis, with nanoscale pearl powder displaying greatest efficiency.4

Skin tone and atopic dermatitis

In 2000, Lopez et al. implanted powdered nacre (mother of pearl derived from Pinctada maxima), which can promote and regulate bone-forming cells, into rat dermis to evaluate its effects on skin fibroblasts. They noted that the implant yielded well-vascularized tissue and improved extracellular matrix production, synthesis of substances involved in cellular adhesion and communication, and tissue regeneration (such as collagen types I and III). The investigators concluded that the powdered nacre contributed to the conditions necessary for improved skin tone and proper physiologic functioning of the skin.10

Rousseau et al. extracted lipids from the nacre of the oyster P. margaritifera to test on artificially dehydrated skin explants with the intention of developing new treatments for atopic dermatitis. The researchers determined that the lipids spurred a reconstitution of the intercellular material of the stratum corneum, concluding that new products to treat atopic dermatitis might be based on the signaling activity of nacre lipids.11

Antifibrotic and anti-inflammatory activity

A 2015 study by Yang et al. showed that a room-temperature superextraction system to yield the main active constituents of pearl was successful in enhancing their anti-inflammatory and antiapoptotic activity in human keratinocyte cells (HaCaT) exposed to low-dose UVB. The investigators combined pearl extract and poly (gamma-glutamic acid) hydrogels and observed reductions in inflammation and apoptosis of HaCaT cells. They concluded that a marketed pearl extract may be able to suppress radiation dermatitis present in keratinocytes.12

Dr. Leslie S. Baumann

Two years later, Latire et al. used human dermal fibroblasts in primary culture to assess the potential biological activities of the matrix macromolecular components extracted from the shells of two edible mollusks (the blue mussel Mytilus edulis and the Pacific oyster Crassostrea gigas). The investigators found that both extracts influenced metabolic functions of the cells and reduced type I collagen levels, with an associated rise in matrix metalloproteinase-1 activity. Given their findings implying the effectiveness of the extracts in facilitating the catabolic pathway of human dermal fibroblasts, the authors suggest that these shell matrices present the potential for use in treating fibrosis, especially for scleroderma.6

Antioxidant and antiaging activity

Shao et al. demonstrated 10 years ago that pearl powder provides a moisturizing effect on the skin, with ultramicro pearl powder delivering a more robust moisturizing result than water-soluble pearl powder. These two types of pearl powder, along with another one tested (ultranano pearl powder), also significantly diminished the activation of tyrosinase and free radicals. Water-soluble pearl powder did not perform as well as the other two formulations in free radical scavenging. The investigators suggested that their results support the use of pearl powder to combat aging and enhance beauty, and could be used in the clinical setting.13

In 2017, Yang et al. reported on the in vitro antihemolytic and antioxidant activity of pearl powder in shielding human erythrocytes against 2,2’-azobis(2-amidinopropane) dihydrochloride–induced oxidative damage to membrane proteins/lipids. The researchers contend that the strong antioxidant qualities of pearl powder could be applied to prevent or protect against various diseases resulting from free radical damage.2

Human trials: Antioxidant, antiaging, skin appearance

Chiu et al. studied the antioxidant activity of various pearl powder extracts in a randomized, placebo-controlled trial in 2018. They also investigated the life span–prolonging effects of the powders using wild-type Caenorhabditis elegans. Twenty healthy middle-aged subjects were separated into two groups (experimental and placebo), with 3 g of pearl powder administered in capsules to the former and 3 g of placebo to the latter over 8 weeks. Blood samples taken at the beginning and every 2 weeks during the trial and in the 10th week revealed maximum antioxidant activity of the pearl powder and prolongation of C. elegans lifespan by 18.87%. Subjects using pearl powder demonstrated significant increases in total antioxidant capacity, thiols, glutathione, and enzymic antioxidant activity, along with notably inhibited lipid peroxidation products. The investigators concluded that pearl powder extract acted as a potent antioxidant and its use may be warranted to treat degenerative conditions related to aging.3

A recent study of the perception of blue light on Korean women’s faces using blue pearl pigment revealed that the pigment does indeed elicit the perception of the blue-light effect, notably transparency and gloss, which is particularly valued in Korea.14

Conclusion

The use of mother of pearl and pearl powder in traditional Chinese medicine and as a cosmetic and food additive has a rich and lengthy history. Contemporary research clearly suggests interesting avenues for further investigation and some promising results. Much more research is necessary, though, to delineate the potential roles of pearl powder in the skin care arsenal.
 

Dr. Baumann is a private practice dermatologist, researcher, author, and entrepreneur who practices in Miami. She founded the Cosmetic Dermatology Center at the University of Miami in 1997. Dr. Baumann wrote two textbooks: “Cosmetic Dermatology: Principles and Practice” (New York: McGraw-Hill, 2002), and “Cosmeceuticals and Cosmetic Ingredients” (New York: McGraw-Hill, 2014), and a New York Times Best Sellers book for consumers, “The Skin Type Solution” (New York: Bantam Dell, 2006). Dr. Baumann has received funding for advisory boards and/or clinical research trials from Allergan, Evolus, Galderma, and Revance. She is the founder and CEO of Skin Type Solutions Franchise Systems. Write to her at dermnews@mdedge.com

References

1. Zhang J et al. J Sep Sci. 2015 May;38(9):1552-60.

2. Yang HL et al. J Food Drug Anal. 2017 Oct;25(4):898-907.

3. Chiu HF et al. J Food Drug Anal. 2018 Jan;26(1):309-17.

4. Chen X et al. Drug Dev Ind Pharm. 2019 Jun;45(6):1009-16.

5. Nagai K. Zoolog Sci. 2013 Oct;30(10):783-93.

6. Latire T et al. Cytotechnology. 2017 Oct;69(5):815-29.

7. Li YC et al. Pharm Biol. 2013 Mar;51(3):289-97.

8. Jian-Ping D et al. Pharm Biol. 2010 Feb;48(2):122-7.

9. Lee K et al. Mol Biol Rep. 2012 Mar;39(3):3211-8.

10. Lopez E et al. Tissue Cell. 2000 Feb;32(1):95-101.

11. Rousseau M et al. Comp Biochem Physiol B Biochem Mol Biol. 2006 Sep;145(1):1-9.

12. Yang YL et al. Biomed Mater Eng. 2015;26 Suppl 1:S139-45.

13. Shao DZ et al. J Cosmet Sci. 2010 Mar-Apr;61(2):133-45.

14. Lee M et al. Skin Res Technol. 2020 Jan;26(1):76-80.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.