LayerRx Mapping ID
645
Slot System
Featured Buckets
Featured Buckets Admin
Reverse Chronological Sort
Allow Teaser Image
Medscape Lead Concept
39

Rosacea is in the eye of the beholder, expert says

Article Type
Changed
Sat, 02/26/2022 - 14:48

 

In the clinical experience of Emmy Graber, MD, MBA, rosacea is in the eye of the beholder.

Dr. Emmy Graber, a dermatologist in Boston
Dr. Emmy Graber

“It’s not really up to us as the providers as to what’s important to the patient or how bad their rosacea is,” she said during MedscapeLive’s annual Las Vegas Dermatology Seminar. “It really is up to the patient,” added Dr. Graber, president of The Dermatology Institute of Boston, who recommends asking patients about how severe they consider their rosacea to be, and what about rosacea bothers them most. Their responses may be surprising.



A study published in 2017 showed that complete resolution of even mild rosacea prolongs remission of rosacea, and most importantly, improves the quality of life for patients. “So, don’t discount what you consider to be mild rosacea in patients,” she said.

Skin care recommendations

“And don’t forget about basic skin care,” she advised. A recently published Chinese study of 999 rosacea patients and 1,010 controls with healthy skin found that a high frequency of cleansing and expansive use of cleansers were positively correlated with rosacea occurrence, suggesting that overcleansing can be a risk factor for rosacea. “Ask your patient, ‘how often are you cleaning your face?’ ” Dr. Graber suggested. “You might find that they’re overdoing it by washing three or four times a day. Several studies have shown that basic skin care alone improves rosacea.”

Skin care recommendations for patients with rosacea include avoiding chemical or physical exfoliants and alcohol-based topical products, and moisturizing and washing their faces with mild, synthetic detergent-based products rather than traditional soaps, which may further alkalinize and irritate the skin. “Patients should also be counseled to use physical-based sunscreens rather than chemical-based sunscreens,” she said.
 

Treating erythema

For treating erythema with topicals, a systematic review published in 2019 found the most evidence for brimonidine 0.33% gel, an alpha2-adrenergic agonist, and oxymetazoline 1% cream, an alpha1-adrenergic agonist. “Both of these products functionally constrict facial blood vessels,” and are Food and Drug Administration approved for treating persistent erythema, Dr. Graber said. “These products improve erythema within 3 hours of and up to 12 hours after application and overall, they are well tolerated.”

Rosacea.org
Subtype 1: Facial redness: Flushing and persistent redness. Visible blood vessels may also appear.

Based on clinical trial results, about 15% of patients on brimonidine report adverse reactions such as dermatitis, burning, pruritus, and erythema, compared with 8% of patients on oxymetazoline. At the same time, up to 20% of individuals on brimonidine report rebound erythema, compared with fewer than 1% of those using oxymetazoline. Laser and light therapies such as pulse-dye lasers, potassium-titanyl-phosphate lasers, and intense-pulse light devices are also effective in treating persistent erythema but are less effective for transient flushing.
 

 

 

Treatment of papules and pustules

For treating papules and pustules, the 2019 systemic review also found high-certainty evidence for using azelaic acid and topical ivermectin, and moderate-certainty evidence for using topical metronidazole and topical minocycline. “Topical ivermectin was demonstrated to be the most effective topical treatment for papulopustular rosacea and to provide the greatest psychological benefit to these patients,” Dr. Graber said.

In a double-blind, multicenter 15-week trial comparing azelaic acid 15% gel with metronidazole 0.75% gel in patients with papulopustular rosacea, both agents were found to be effective. But those treated with azelaic acid 15% gel had a greater reduction in lesion counts and erythema, and improvement in global assessments, compared with metronidazole 0.75% gel. However, the azelaic acid 15% gel was associated with more stinging compared with metronidazole 0.75% gel, although it was usually transient.

Another study, a double-blind, single-center, 15-week trial, compared the efficacy of azelaic acid 20% cream with metronidazole 0.75% cream. Both agents were found to be effective and had similar levels of reductions in papules and pustules. However, patients in the azelaic acid 20% cream arm had significantly higher physician ratings of global improvement, as well as overall higher patient satisfaction.

More recently, a phase 3 study of 962 patients found that ivermectin 1% cream once daily improved quality of life slightly more than metronidazole 0.75% cream twice daily. No difference in adverse events were noted between the two agents.

Other options for treating papules and pustules include topical minocycline 1.5% foam, which is FDA approved for rosacea, as well as second-line agents topical sodium sulfacetamide with sulfur cleanser (cream or lotion), and permethrin, Dr. Graber said.

As for treating papules and pustules with oral agents, the strongest evidence favors oral tetracyclines and isotretinoin, she noted.

Doxycycline, minocycline, tetracycline, and sarecycline can be used as monotherapy or coadministered with topical agents. “The addition of topical agents may also help to shorten the duration of antibiotic use, which is very important,” Dr. Graber said.

She noted that oral beta-blockers might be useful to treat persistent erythema and flushing because they antagonize the effects of sympathetic nerve stimulation and circulating catecholamines at b-adrenoceptors. Carvedilol and propranolol have been the most studied. The most common potential side effects are hypotension and bradycardia.

Dr. Graber disclosed that she is a consultant/adviser for Digital Diagnostics, Almirall, Hovione, Keratin Biosciences, La Roche Posay, Ortho Dermatologics, Sebacia, Sol-Gel, Verrica, and WebMD. She is also a research investigator for Hovione, Ortho Dermatologics, Sebacia, and she receives royalties from Wolters Kluwer Health.

MedscapeLive and this news organization are owned by the same parent company.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

In the clinical experience of Emmy Graber, MD, MBA, rosacea is in the eye of the beholder.

Dr. Emmy Graber, a dermatologist in Boston
Dr. Emmy Graber

“It’s not really up to us as the providers as to what’s important to the patient or how bad their rosacea is,” she said during MedscapeLive’s annual Las Vegas Dermatology Seminar. “It really is up to the patient,” added Dr. Graber, president of The Dermatology Institute of Boston, who recommends asking patients about how severe they consider their rosacea to be, and what about rosacea bothers them most. Their responses may be surprising.



A study published in 2017 showed that complete resolution of even mild rosacea prolongs remission of rosacea, and most importantly, improves the quality of life for patients. “So, don’t discount what you consider to be mild rosacea in patients,” she said.

Skin care recommendations

“And don’t forget about basic skin care,” she advised. A recently published Chinese study of 999 rosacea patients and 1,010 controls with healthy skin found that a high frequency of cleansing and expansive use of cleansers were positively correlated with rosacea occurrence, suggesting that overcleansing can be a risk factor for rosacea. “Ask your patient, ‘how often are you cleaning your face?’ ” Dr. Graber suggested. “You might find that they’re overdoing it by washing three or four times a day. Several studies have shown that basic skin care alone improves rosacea.”

Skin care recommendations for patients with rosacea include avoiding chemical or physical exfoliants and alcohol-based topical products, and moisturizing and washing their faces with mild, synthetic detergent-based products rather than traditional soaps, which may further alkalinize and irritate the skin. “Patients should also be counseled to use physical-based sunscreens rather than chemical-based sunscreens,” she said.
 

Treating erythema

For treating erythema with topicals, a systematic review published in 2019 found the most evidence for brimonidine 0.33% gel, an alpha2-adrenergic agonist, and oxymetazoline 1% cream, an alpha1-adrenergic agonist. “Both of these products functionally constrict facial blood vessels,” and are Food and Drug Administration approved for treating persistent erythema, Dr. Graber said. “These products improve erythema within 3 hours of and up to 12 hours after application and overall, they are well tolerated.”

Rosacea.org
Subtype 1: Facial redness: Flushing and persistent redness. Visible blood vessels may also appear.

Based on clinical trial results, about 15% of patients on brimonidine report adverse reactions such as dermatitis, burning, pruritus, and erythema, compared with 8% of patients on oxymetazoline. At the same time, up to 20% of individuals on brimonidine report rebound erythema, compared with fewer than 1% of those using oxymetazoline. Laser and light therapies such as pulse-dye lasers, potassium-titanyl-phosphate lasers, and intense-pulse light devices are also effective in treating persistent erythema but are less effective for transient flushing.
 

 

 

Treatment of papules and pustules

For treating papules and pustules, the 2019 systemic review also found high-certainty evidence for using azelaic acid and topical ivermectin, and moderate-certainty evidence for using topical metronidazole and topical minocycline. “Topical ivermectin was demonstrated to be the most effective topical treatment for papulopustular rosacea and to provide the greatest psychological benefit to these patients,” Dr. Graber said.

In a double-blind, multicenter 15-week trial comparing azelaic acid 15% gel with metronidazole 0.75% gel in patients with papulopustular rosacea, both agents were found to be effective. But those treated with azelaic acid 15% gel had a greater reduction in lesion counts and erythema, and improvement in global assessments, compared with metronidazole 0.75% gel. However, the azelaic acid 15% gel was associated with more stinging compared with metronidazole 0.75% gel, although it was usually transient.

Another study, a double-blind, single-center, 15-week trial, compared the efficacy of azelaic acid 20% cream with metronidazole 0.75% cream. Both agents were found to be effective and had similar levels of reductions in papules and pustules. However, patients in the azelaic acid 20% cream arm had significantly higher physician ratings of global improvement, as well as overall higher patient satisfaction.

More recently, a phase 3 study of 962 patients found that ivermectin 1% cream once daily improved quality of life slightly more than metronidazole 0.75% cream twice daily. No difference in adverse events were noted between the two agents.

Other options for treating papules and pustules include topical minocycline 1.5% foam, which is FDA approved for rosacea, as well as second-line agents topical sodium sulfacetamide with sulfur cleanser (cream or lotion), and permethrin, Dr. Graber said.

As for treating papules and pustules with oral agents, the strongest evidence favors oral tetracyclines and isotretinoin, she noted.

Doxycycline, minocycline, tetracycline, and sarecycline can be used as monotherapy or coadministered with topical agents. “The addition of topical agents may also help to shorten the duration of antibiotic use, which is very important,” Dr. Graber said.

She noted that oral beta-blockers might be useful to treat persistent erythema and flushing because they antagonize the effects of sympathetic nerve stimulation and circulating catecholamines at b-adrenoceptors. Carvedilol and propranolol have been the most studied. The most common potential side effects are hypotension and bradycardia.

Dr. Graber disclosed that she is a consultant/adviser for Digital Diagnostics, Almirall, Hovione, Keratin Biosciences, La Roche Posay, Ortho Dermatologics, Sebacia, Sol-Gel, Verrica, and WebMD. She is also a research investigator for Hovione, Ortho Dermatologics, Sebacia, and she receives royalties from Wolters Kluwer Health.

MedscapeLive and this news organization are owned by the same parent company.

 

In the clinical experience of Emmy Graber, MD, MBA, rosacea is in the eye of the beholder.

Dr. Emmy Graber, a dermatologist in Boston
Dr. Emmy Graber

“It’s not really up to us as the providers as to what’s important to the patient or how bad their rosacea is,” she said during MedscapeLive’s annual Las Vegas Dermatology Seminar. “It really is up to the patient,” added Dr. Graber, president of The Dermatology Institute of Boston, who recommends asking patients about how severe they consider their rosacea to be, and what about rosacea bothers them most. Their responses may be surprising.



A study published in 2017 showed that complete resolution of even mild rosacea prolongs remission of rosacea, and most importantly, improves the quality of life for patients. “So, don’t discount what you consider to be mild rosacea in patients,” she said.

Skin care recommendations

“And don’t forget about basic skin care,” she advised. A recently published Chinese study of 999 rosacea patients and 1,010 controls with healthy skin found that a high frequency of cleansing and expansive use of cleansers were positively correlated with rosacea occurrence, suggesting that overcleansing can be a risk factor for rosacea. “Ask your patient, ‘how often are you cleaning your face?’ ” Dr. Graber suggested. “You might find that they’re overdoing it by washing three or four times a day. Several studies have shown that basic skin care alone improves rosacea.”

Skin care recommendations for patients with rosacea include avoiding chemical or physical exfoliants and alcohol-based topical products, and moisturizing and washing their faces with mild, synthetic detergent-based products rather than traditional soaps, which may further alkalinize and irritate the skin. “Patients should also be counseled to use physical-based sunscreens rather than chemical-based sunscreens,” she said.
 

Treating erythema

For treating erythema with topicals, a systematic review published in 2019 found the most evidence for brimonidine 0.33% gel, an alpha2-adrenergic agonist, and oxymetazoline 1% cream, an alpha1-adrenergic agonist. “Both of these products functionally constrict facial blood vessels,” and are Food and Drug Administration approved for treating persistent erythema, Dr. Graber said. “These products improve erythema within 3 hours of and up to 12 hours after application and overall, they are well tolerated.”

Rosacea.org
Subtype 1: Facial redness: Flushing and persistent redness. Visible blood vessels may also appear.

Based on clinical trial results, about 15% of patients on brimonidine report adverse reactions such as dermatitis, burning, pruritus, and erythema, compared with 8% of patients on oxymetazoline. At the same time, up to 20% of individuals on brimonidine report rebound erythema, compared with fewer than 1% of those using oxymetazoline. Laser and light therapies such as pulse-dye lasers, potassium-titanyl-phosphate lasers, and intense-pulse light devices are also effective in treating persistent erythema but are less effective for transient flushing.
 

 

 

Treatment of papules and pustules

For treating papules and pustules, the 2019 systemic review also found high-certainty evidence for using azelaic acid and topical ivermectin, and moderate-certainty evidence for using topical metronidazole and topical minocycline. “Topical ivermectin was demonstrated to be the most effective topical treatment for papulopustular rosacea and to provide the greatest psychological benefit to these patients,” Dr. Graber said.

In a double-blind, multicenter 15-week trial comparing azelaic acid 15% gel with metronidazole 0.75% gel in patients with papulopustular rosacea, both agents were found to be effective. But those treated with azelaic acid 15% gel had a greater reduction in lesion counts and erythema, and improvement in global assessments, compared with metronidazole 0.75% gel. However, the azelaic acid 15% gel was associated with more stinging compared with metronidazole 0.75% gel, although it was usually transient.

Another study, a double-blind, single-center, 15-week trial, compared the efficacy of azelaic acid 20% cream with metronidazole 0.75% cream. Both agents were found to be effective and had similar levels of reductions in papules and pustules. However, patients in the azelaic acid 20% cream arm had significantly higher physician ratings of global improvement, as well as overall higher patient satisfaction.

More recently, a phase 3 study of 962 patients found that ivermectin 1% cream once daily improved quality of life slightly more than metronidazole 0.75% cream twice daily. No difference in adverse events were noted between the two agents.

Other options for treating papules and pustules include topical minocycline 1.5% foam, which is FDA approved for rosacea, as well as second-line agents topical sodium sulfacetamide with sulfur cleanser (cream or lotion), and permethrin, Dr. Graber said.

As for treating papules and pustules with oral agents, the strongest evidence favors oral tetracyclines and isotretinoin, she noted.

Doxycycline, minocycline, tetracycline, and sarecycline can be used as monotherapy or coadministered with topical agents. “The addition of topical agents may also help to shorten the duration of antibiotic use, which is very important,” Dr. Graber said.

She noted that oral beta-blockers might be useful to treat persistent erythema and flushing because they antagonize the effects of sympathetic nerve stimulation and circulating catecholamines at b-adrenoceptors. Carvedilol and propranolol have been the most studied. The most common potential side effects are hypotension and bradycardia.

Dr. Graber disclosed that she is a consultant/adviser for Digital Diagnostics, Almirall, Hovione, Keratin Biosciences, La Roche Posay, Ortho Dermatologics, Sebacia, Sol-Gel, Verrica, and WebMD. She is also a research investigator for Hovione, Ortho Dermatologics, Sebacia, and she receives royalties from Wolters Kluwer Health.

MedscapeLive and this news organization are owned by the same parent company.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM MEDSCAPELIVE LAS VEGAS DERMATOLOGY SEMINAR

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

Microbiome studies among those awarded National Rosacea Society grants

Article Type
Changed
Tue, 11/23/2021 - 09:53

 

A study on the role of the ocular surface microbiome in rosacea pathogenesis and an investigation of elevated intracellular signals in rosacea lesions earned new research funding from the National Rosacea Society (NRS) this year, as part of the organization’s research grants program.

Rosacea, subtype 4: Eye irritation (ocular rosacea). Watery or bloodshot appearance, irritation, burning, or stinging.
National Rosacea Society

The NRS research grants program was created to increase knowledge and understanding of not only the potential causes of rosacea, but other aspects of the disease that may inform prevention, treatment, or a potential cure, according to the press release announcing the recipients.



New research grant recipient Sezen Karakus, MD, of the Johns Hopkins Wilmer Eye Institute, Baltimore, received $15,000 for a study on the contribution of the ocular surface microbiome to the development of rosacea. Ocular rosacea can result in corneal complications severe enough to affect vision, and identifying the microorganisms on the ocular surface may lead to new treatment strategies, Dr. Karakus said in the release. He will collaborate on this research with dermatologist Noori Kim, MD, of Johns Hopkins University, Baltimore.

A second new research grant went to Emmanuel Contassot, MD, project leader in the dermatology department at of the University Hospital of Basel, Switzerland, who received $5,000 to investigate whether certain elevated intracellular signals in rosacea lesions may promote the skin inflammation that may be a root cause of the condition.

The NRS also renewed its support of a pair of ongoing studies. Michelle Trautwein, MD, of the Institute for Biodiversity Science and Sustainability at the California Academy of Sciences, continues her work on the first study to sequence the genome of Demodex mites; the study also identifies associated bacteria that may play a role in rosacea.

Demodex mite
National Rosacea Society
Demodex mite


A second ongoing study by Tissa Hata, MD, of the University of California, San Diego, focuses on the normalization of the microbiome in people with rosacea. Dr. Hata’s work identifies types of bacteria associated with rosacea, as well as bacteria that may be associated with healthy skin after successful treatment of rosacea, including Cutibacterium acnes and Staphylococcus epidermidis.

The deadline to submit research proposals for next year’s grants is June 17, 2022. Researchers can find forms and instructions at the research grants section of the NRS website or by contacting the National Rosacea Society at 111 Lions Dr., Suite 216, Barrington, Ill., 60010, by telephone at 1-888-662-5874, or by email at info@rosacea.org.

Publications
Topics
Sections

 

A study on the role of the ocular surface microbiome in rosacea pathogenesis and an investigation of elevated intracellular signals in rosacea lesions earned new research funding from the National Rosacea Society (NRS) this year, as part of the organization’s research grants program.

Rosacea, subtype 4: Eye irritation (ocular rosacea). Watery or bloodshot appearance, irritation, burning, or stinging.
National Rosacea Society

The NRS research grants program was created to increase knowledge and understanding of not only the potential causes of rosacea, but other aspects of the disease that may inform prevention, treatment, or a potential cure, according to the press release announcing the recipients.



New research grant recipient Sezen Karakus, MD, of the Johns Hopkins Wilmer Eye Institute, Baltimore, received $15,000 for a study on the contribution of the ocular surface microbiome to the development of rosacea. Ocular rosacea can result in corneal complications severe enough to affect vision, and identifying the microorganisms on the ocular surface may lead to new treatment strategies, Dr. Karakus said in the release. He will collaborate on this research with dermatologist Noori Kim, MD, of Johns Hopkins University, Baltimore.

A second new research grant went to Emmanuel Contassot, MD, project leader in the dermatology department at of the University Hospital of Basel, Switzerland, who received $5,000 to investigate whether certain elevated intracellular signals in rosacea lesions may promote the skin inflammation that may be a root cause of the condition.

The NRS also renewed its support of a pair of ongoing studies. Michelle Trautwein, MD, of the Institute for Biodiversity Science and Sustainability at the California Academy of Sciences, continues her work on the first study to sequence the genome of Demodex mites; the study also identifies associated bacteria that may play a role in rosacea.

Demodex mite
National Rosacea Society
Demodex mite


A second ongoing study by Tissa Hata, MD, of the University of California, San Diego, focuses on the normalization of the microbiome in people with rosacea. Dr. Hata’s work identifies types of bacteria associated with rosacea, as well as bacteria that may be associated with healthy skin after successful treatment of rosacea, including Cutibacterium acnes and Staphylococcus epidermidis.

The deadline to submit research proposals for next year’s grants is June 17, 2022. Researchers can find forms and instructions at the research grants section of the NRS website or by contacting the National Rosacea Society at 111 Lions Dr., Suite 216, Barrington, Ill., 60010, by telephone at 1-888-662-5874, or by email at info@rosacea.org.

 

A study on the role of the ocular surface microbiome in rosacea pathogenesis and an investigation of elevated intracellular signals in rosacea lesions earned new research funding from the National Rosacea Society (NRS) this year, as part of the organization’s research grants program.

Rosacea, subtype 4: Eye irritation (ocular rosacea). Watery or bloodshot appearance, irritation, burning, or stinging.
National Rosacea Society

The NRS research grants program was created to increase knowledge and understanding of not only the potential causes of rosacea, but other aspects of the disease that may inform prevention, treatment, or a potential cure, according to the press release announcing the recipients.



New research grant recipient Sezen Karakus, MD, of the Johns Hopkins Wilmer Eye Institute, Baltimore, received $15,000 for a study on the contribution of the ocular surface microbiome to the development of rosacea. Ocular rosacea can result in corneal complications severe enough to affect vision, and identifying the microorganisms on the ocular surface may lead to new treatment strategies, Dr. Karakus said in the release. He will collaborate on this research with dermatologist Noori Kim, MD, of Johns Hopkins University, Baltimore.

A second new research grant went to Emmanuel Contassot, MD, project leader in the dermatology department at of the University Hospital of Basel, Switzerland, who received $5,000 to investigate whether certain elevated intracellular signals in rosacea lesions may promote the skin inflammation that may be a root cause of the condition.

The NRS also renewed its support of a pair of ongoing studies. Michelle Trautwein, MD, of the Institute for Biodiversity Science and Sustainability at the California Academy of Sciences, continues her work on the first study to sequence the genome of Demodex mites; the study also identifies associated bacteria that may play a role in rosacea.

Demodex mite
National Rosacea Society
Demodex mite


A second ongoing study by Tissa Hata, MD, of the University of California, San Diego, focuses on the normalization of the microbiome in people with rosacea. Dr. Hata’s work identifies types of bacteria associated with rosacea, as well as bacteria that may be associated with healthy skin after successful treatment of rosacea, including Cutibacterium acnes and Staphylococcus epidermidis.

The deadline to submit research proposals for next year’s grants is June 17, 2022. Researchers can find forms and instructions at the research grants section of the NRS website or by contacting the National Rosacea Society at 111 Lions Dr., Suite 216, Barrington, Ill., 60010, by telephone at 1-888-662-5874, or by email at info@rosacea.org.

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

Contact Allergy to Topical Medicaments, Part 1: A Double-edged Sword

Article Type
Changed
Wed, 11/10/2021 - 13:13
Display Headline
Contact Allergy to Topical Medicaments, Part 1: A Double-edged Sword

Topical medications frequently are prescribed in dermatology and provide the advantages of direct skin penetration and targeted application while typically sparing patients from systemic effects. Adverse cutaneous effects include allergic contact dermatitis (ACD), irritant contact dermatitis (ICD), photosensitivity, urticaria, hyperpigmentation or hypopigmentation, atrophy, periorificial dermatitis, and acneform eruptions. Allergic contact dermatitis can develop from the active drug or vehicle components.

Patients with medicament ACD often present with symptoms of pruritus and dermatitis at the site of topical application. They may express concern that the medication is no longer working or seems to be making things worse. Certain sites are more prone to developing medicament dermatitis, including the face, groin, and lower legs. Older adults may be more at risk. Other risk factors include pre-existing skin diseases such as stasis dermatitis, acne, psoriasis, atopic dermatitis, and genital dermatoses.1 A review of 14,911 patch-tested patients from a single referral clinic revealed that 17.4% had iatrogenic contact dermatitis, with the most common culprits being topical antibiotics, antiseptics, and steroids.2

In this 2-part series, we will focus on the active drug as a source of ACD. Part 1 explores ACD associated with acne and rosacea medications, antimicrobials, antihistamines, and topical pain preparations.

 

Acne and Rosacea Medications

Retinoids—Topical retinoids are first-line acne treatments that help normalize skin keratinization. Irritant contact dermatitis from retinoids is a well-known and common side effect. Although far less common than ICD, ACD from topical retinoid use has been reported.3,4 Reactions to tretinoin are most frequently reported in the literature compared to adapalene gel5 and tazarotene foam, which have lower potential for sensitization.6 Allergic contact dermatitis also has been reported from retinyl palmitate7,8 in cosmetic creams and from occupational exposure in settings of industrial vitamin A production.9 Both ICD and ACD from topical retinoids can present with pruritus, erythema, and scaling. Given this clinical overlap between ACD and ICD, patch testing is crucial in differentiating the underlying etiology of the dermatitis.

Benzoyl Peroxide—Benzoyl peroxide (BP) is another popular topical acne treatment that targets Cutibacterium acnes, a bacterium often implicated in the pathogenesis of acne vulgaris. Similar to retinoids, ICD is more common than ACD. Several cases of ACD to BP have been reported.10-14 Occasionally, honey-colored crusting associated with ACD to BP can mimic impetigo.10 Aside from use of BP as an acne treatment, other potential exposures to BP include bleached flour13 and orthopedic bone cement. Occupations at risk for potential BP exposure include dental technicians15 and those working in plastic manufacturing.

Brimonidine—Brimonidine tartrate is a selective α2-adrenergic agonist initially used to treat open-angle glaucoma and also is used as a topical treatment for rosacea. Allergic reactions to brimonidine eye drops may present with periorbital hyperpigmentation and pruritic bullous lesions.16 Case reports of topical brimonidine ACD have demonstrated mixed patch test results, with positive patch tests to Mirvaso (Galderma) as is but negative patch tests to pure brimonidine tartrate 0.33%.17,18 Ringuet and Houle19 reported the first known positive patch test reaction to pure topical brimonidine, testing with brimonidine tartrate 1% in petrolatum.20,21 Clinicians should be attuned to ACD to topical brimonidine in patients previously treated for glaucoma, as prior use of ophthalmic preparations may result in sensitization.18,20

Antimicrobials

Clindamycin—Clindamycin targets bacterial protein synthesis and is an effective adjunct in the treatment of acne. Despite its widespread and often long-term use, topical clindamycin is a weak sensitizer.22 To date, limited case reports on ACD to topical clindamycin exist.23-28 Rare clinical patterns of ACD to clindamycin include mimickers of irritant retinoid dermatitis, erythema multiforme, or pustular rosacea.25,26,29

 

 

Metronidazole—Metronidazole is a bactericidal agent that disrupts nucleic acid synthesis with additional anti-inflammatory properties used in the treatment of rosacea. Allergic contact dermatitis to topical metronidazole has been reported.30-34 In 2006, Beutner at al35 patch tested 215 patients using metronidazole gel 1%, which revealed no positive reactions to indicate contact sensitization. Similarly, Jappe et al36 found no positive reactions to metronidazole 2% in petrolatum in their prospective analysis of 78 rosacea patients, further highlighting the exceptionally low incidence of ACD. Cross-reaction with isothiazolinone, which shares structurally similar properties to metronidazole, has been speculated.31,34 One patient developed an acute reaction to metronidazole gel 0.75% within 24 hours of application, suggesting that isothiazolinone may act as a sensitizer, though this relationship has not been proven.31

Neomycin—Neomycin blocks bacterial protein synthesis and is available in both prescription and over-the-counter (OTC) formulations. It commonly is used to treat and prevent superficial wound infections as an OTC antibiotic and also has otic, ophthalmologic, gastroenterologic, urologic, and peritoneal formulations. It also can be used in the dental and veterinary fields and is present in some animal feeds and in trace amounts in some vaccines for humans. Neomycin is a common antibiotic contact allergen, and the most recently reported 2017-2018 North American Contact Dermatitis Group data cycle placed it at number 12 with 5.4% positivity.37 Co-reactions with bacitracin can occur, substantially limiting OTC topical antibiotic options for allergic patients. A safe alternative for patients with neomycin (and bacitracin and polymyxin) contact allergy is prescription mupirocin.

Bacitracin—Bacitracin interferes with peptidoglycan and cell-wall synthesis to treat superficial cutaneous infections. Similar to neomycin, it also can be found in OTC antibiotic ointments as well as in antibacterial bandages. There are several case reports of patients with both type IV delayed hypersensitivity (contact dermatitis) and type I anaphylactic reactions to bacitracin38-40; patch testers should be aware of this rare association. Bacitracin was positive in 5.5% of patch tested patients in the 2017-2018 North American Contact Dermatitis Group data cycle,37 and as with neomycin, bacitracin also is commonly patch tested in most screening patch test series.

Polymyxin—Polymyxin is a polypeptide topical antibiotic that is used to treat superficial wound infections and can be used in combination with neomycin and/or bacitracin. Historically, it is a less common antibiotic allergen; however, it is now frequently included in comprehensive patch test series, as the frequency of positive reactions seems to be increasing, probably due to polysensitization with neomycin and bacitracin.

Nystatin—Nystatin is an antifungal that binds to ergosterol and disrupts the cell wall. Cases exist of ACD to topical nystatin as well as systemic ACD from oral exposure, though both are quite rare. Authors have surmised that the overall low rates of ACD may be due to poor skin absorption of nystatin, which also can confound patch testing.41,42 For patients with suspected ACD to nystatin, repeat open application testing also can be performed to confirm allergy.

 

 

Imidazole Antifungals—Similar to nystatins, imidazole antifungals also work by disrupting the fungal cell wall. Imidazole antifungal preparations that have been reported to cause ACD include clotrimazole, miconazole, econazole, and isoconazole, and although cross-reactivity patterns have been described, they are not always reproducible with patch testing.43 In one reported case, tioconazole found in an antifungal nail lacquer triggered ACD involving not only the fingers and toes but also the trunk.44 Erythema multiforme–like reactions also have been described from topical use.45 Commercial patch test preparations of the most common imidazole allergens do exist. Nonimidazole antifungals remain a safe option for allergic patients.

Antihistamines

Antihistamines, or H1-receptor antagonists, are marketed to be applied topically for relief of pruritus associated with allergic cutaneous reactions. Ironically, they are known to be potent sensitizers themselves. There are 6 main chemical classes of antihistamines: phenothiazines, ethylenediamines, ethanolamines, alkylamines, piperazines, and piperidines. Goossens and Linsen46 patch tested 12,460 patients from 1978 to 1997 and found the most positive reactions to promethazine (phenothiazine)(n=12), followed by diphenhydramine (ethanolamine)(n=8) and clemizole (benzimidazole)(n=6). The authors also noted cross-reactions between diphenhydramine derivatives and between promethazine and chlorpromazine.46

Doxepin is a tricyclic antidepressant with antihistamine activity and is a well-documented sensitizer.47-52 Taylor et al47 evaluated 97 patients with chronic dermatoses, and patch testing revealed 17 (17.5%) positive reactions to doxepin cream, 13 (76.5%) of which were positive reactions to both the commercial cream and the active ingredient. Patch testing using doxepin dilution as low as 0.5% in petrolatum is sufficient to provoke a strong (++) allergic reaction.50,51 Early-onset ACD following the use of doxepin cream suggests the possibility of prior sensitization, perhaps with a structurally similar phenothiazine drug.51 A keen suspicion for ACD in patients using doxepin cream for longer than the recommended duration can help make the diagnosis.49,52

 

Topical Analgesics

Nonsteroidal Anti-inflammatory Drugs—Ketoprofen is one of the most frequent culprits of photoallergic contact dermatitis. Pruritic, papulovesicular, and bullous lesions typically develop acutely weeks after exposure. Prolonged photosensitivity is common and can last years after discontinuation of the nonsteroidal anti-inflammatory drug.53 Cases of cross-reactions and co-sensitization to structurally similar substances have been reported, including to benzophenone-related chemicals in sunscreen and aldehyde groups in fragrance mix.53,54

Diclofenac gel generally is well tolerated in the topical treatment of joint pain and inflammation. In the setting of ACD, patients typically present with dermatitis localized to the area of application.55 Immediate cessation and avoidance of topical diclofenac are crucial components of management. Although systemic contact dermatitis has been reported with oral diclofenac use,56 a recent report suggested that oral diclofenac may be well tolerated for some patients with topical ACD.57

 

 

Publications on bufexamac-induced ACD mainly consist of international reports, as this medication has been discontinued in the United States. Bufexamac is a highly sensitizing agent that can lead to severe polymorphic eruptions requiring treatment with prednisolone and even hospitalization.58 In one Australian case report, a mother developed an edematous, erythematous, papulovesicular eruption on the breast while breastfeeding her baby, who was being treated with bufexamac cream 5% for infantile eczema.59 Carprofen-induced photoallergic contact dermatitis is associated with occupational exposure in pharmaceutical workers.60,61 A few case reports on other nonsteroidal anti-inflammatory drugs, including etofenamate and aceclofenac, have been published.62,63

Compounded Medications—Compounded topical analgesics, which help to control pain via multiple combined effects, have gained increasing popularity in the management of chronic neuropathic pain disorders. Only a few recent retrospective studies assessing the efficacy and safety of these medications have mentioned suspected allergic cutaneous reactions.62,63 In 2015, Turrentine et al64 reported a case of ACD to cyclobenzaprine in a compound containing ketamine 10%, diclofenac 5%, baclofen 2%, bupivacaine 1%, cyclobenzaprine 2%, gabapentin 6%, ibuprofen 3%, and pentoxifylline 3% in a proprietary cream base. When patients present with suspected ACD to a compounded pain medication, obtaining individual components for patch testing is key to determining the allergic ingredient(s). We suspect that we will see a rise in reports of ACD as these topical compounds become readily adopted in clinical practices.

Patch Testing for Diagnosis

When patients present with symptoms concerning for ACD to medicaments, the astute clinician should promptly stop the suspected topical medication and consider patch testing. For common allergens such as neomycin, bacitracin, or ethylenediamine, commercial patch test preparations exist and should be used; however, for drugs that do not have a commercial patch test preparation, the patient’s product can be applied as is, keeping in mind that certain preparations (such as retinoids) can cause irritant patch test reactions, which may confound the reading. Alternatively, individual ingredients in the medication’s formulation can be requested from the manufacturer or a compounding pharmacy for targeted testing. Suggested concentrations for patch testing based on the literature and expert reference are listed in the Table. The authors (M.R., A.R.A.) frequently rely on an expert reference66 to determine ideal concentrations for patch testing. Referral to a specialized patch test clinic may be appropriate.

 

Final Interpretation

Although their intent is to heal, topical medicaments also can be a source of ACD. The astute clinician should consider ACD when topicals either no longer seem to help the patient or trigger new-onset dermatitis. Patch testing directly with the culprit medicament, or individual medication ingredients when needed, can lead to the diagnosis, though caution is advised. Stay tuned for part 2 of this series in which we will discuss ACD to topical steroids, immunomodulators, and anesthetic medications.

References
  1. Davis MD. Unusual patterns in contact dermatitis: medicaments. Dermatol Clin. 2009;27:289-297, vi. doi:10.1016/j.det.2009.05.003
  2. Gilissen L, Goossens A. Frequency and trends of contact allergy to and iatrogenic contact dermatitis caused by topical drugs over a 25-year period. Contact Dermatitis. 2016;75:290-302. doi:10.1111/cod.12621
  3. Balato N, Patruno C, Lembo G, et al. Allergic contact dermatitis from retinoic acid. Contact Dermatitis. 1995;32:51. doi:10.1111/j.1600-0536.1995.tb00846.x
  4. Berg JE, Bowman JP, Saenz AB. Cumulative irritation potential and contact sensitization potential of tazarotene foam 0.1% in 2 phase 1 patch studies. Cutis. 2012;90:206-211.
  5. Numata T, Jo R, Kobayashi Y, et al. Allergic contact dermatitis caused by adapalene. Contact Dermatitis. 2015;73:187-188. doi:10.1111/cod.12410
  6. Anderson A, Gebauer K. Periorbital allergic contact dermatitis resulting from topical retinoic acid use. Australas J Dermatol. 2014;55:152-153. doi:10.1111/ajd.12041
  7. Blondeel A. Contact allergy to vitamin A. Contact Dermatitis. 1984;11:191-192. doi:10.1111/j.1600-0536.1984.tb00976.x
  8. Manzano D, Aguirre A, Gardeazabal J, et al. Allergic contact dermatitis from tocopheryl acetate (vitamin E) and retinol palmitate (vitamin A) in a moisturizing cream. Contact Dermatitis. 1994;31:324. doi:10.1111/j.1600-0536.1994.tb02030.x
  9. Heidenheim M, Jemec GB. Occupational allergic contact dermatitis from vitamin A acetate. Contact Dermatitis. 1995;33:439. doi:10.1111/j.1600-0536.1995.tb02091.x
  10. Kim C, Craiglow BG, Watsky KL, et al. Allergic contact dermatitis to benzoyl peroxide resembling impetigo. Pediatr Dermatol. 2015;32:E161-E162. doi:10.1111/pde.12585
  11. Sandre M, Skotnicki-Grant S. A case of a paediatric patient with allergic contact dermatitis to benzoyl peroxide. J Cutan Med Surg. 2018;22:226-228. doi:10.1177/1203475417733462
  12. Corazza M, Amendolagine G, Musmeci D, et al. Sometimes even Dr Google is wrong: an unusual contact dermatitis caused by benzoyl peroxide. Contact Dermatitis. 2018;79:380-381. doi:10.1111/cod.13086
  13. Adelman M, Mohammad T, Kerr H. Allergic contact dermatitis due to benzoyl peroxide from an unlikely source. Dermatitis. 2019;30:230-231. doi:10.1097/DER.0000000000000470
  14. Gatica-Ortega ME, Pastor-Nieto MA. Allergic contact dermatitis to Glycyrrhiza inflata root extract in an anti-acne cosmetic product [published online April 28, 2021]. Contact Dermatitis. doi:10.1111/cod.13872
  15. Ockenfels HM, Uter W, Lessmann H, et al. Patch testing with benzoyl peroxide: reaction profile and interpretation of positive patch test reactions. Contact Dermatitis. 2009;61:209-216. doi:10.1111/j.1600-0536.2009.01603.x
  16. Sodhi PK, Verma L, Ratan J. Dermatological side effects of brimonidine: a report of three cases. J Dermatol. 2003;30:697-700. doi:10.1111/j.1346-8138.2003.tb00461.x
  17. Swanson LA, Warshaw EM. Allergic contact dermatitis to topical brimonidine tartrate gel 0.33% for treatment of rosacea. J Am Acad Dermatol. 2014;71:832-833. doi:10.1016/j.jaad.2014.05.073
  18. Bangsgaard N, Fischer LA, Zachariae C. Sensitization to and allergic contact dermatitis caused by Mirvaso(®)(brimonidine tartrate) for treatment of rosacea—2 cases. Contact Dermatitis. 2016;74:378-379. doi:10.1111/cod.12547
  19. Ringuet J, Houle MC. Case report: allergic contact dermatitis to topical brimonidine demonstrated with patch testing: insights on evaluation of brimonidine sensitization. J Cutan Med Surg. 2018;22:636-638. doi:10.1177/1203475418789020
  20. Cookson H, McFadden J, White J, et al. Allergic contact dermatitis caused by Mirvaso®, brimonidine tartrate gel 0.33%, a new topical treatment for rosaceal erythema. Contact Dermatitis. 2015;73:366-367. doi:10.1111/cod.12476
  21. Rajagopalan A, Rajagopalan B. Allergic contact dermatitis to topical brimonidine. Australas J Dermatol. 2015;56:235. doi:10.1111/ajd.12299
  22. Veraldi S, Brena M, Barbareschi M. Allergic contact dermatitis caused by topical antiacne drugs. Expert Rev Clin Pharmacol. 2015;8:377-381. doi:10.1586/17512433.2015.1046839
  23. Vejlstrup E, Menné T. Contact dermatitis from clindamycin. Contact Dermatitis. 1995;32:110. doi:10.1111/j.1600-0536.1995.tb00759.x
  24. García R, Galindo PA, Feo F, et al. Delayed allergic reactions to amoxycillin and clindamycin. Contact Dermatitis. 1996;35:116-117. doi:10.1111/j.1600-0536.1996.tb02312.x
  25. Muñoz D, Del Pozo MD, Audicana M, et al. Erythema-multiforme-like eruption from antibiotics of 3 different groups. Contact Dermatitis. 1996;34:227-228. doi:10.1111/j.1600-0536.1996.tb02187.x
  26. Romita P, Ettorre G, Corazza M, et al. Allergic contact dermatitis caused by clindamycin mimicking ‘retinoid flare.’ Contact Dermatitis. 2017;77:181-182. doi:10.1111/cod.12784
  27. Veraldi S, Guanziroli E, Ferrucci S, et al. Allergic contact dermatitis caused by clindamycin. Contact Dermatitis. 2019;80:68-69. doi:10.1111/cod.13133
  28. Voller LM, Kullberg SA, Warshaw EM. Axillary allergic contact dermatitis to topical clindamycin. Contact Dermatitis. 2020;82:313-314. doi:10.1111/cod.13465
  29. de Kort WJ, de Groot AC. Clindamycin allergy presenting as rosacea. Contact Dermatitis. 1989;20:72-73. doi:10.1111/j.1600-0536.1989.tb03108.x
  30. Vincenzi C, Lucente P, Ricci C, et al. Facial contact dermatitis due to metronidazole. Contact Dermatitis. 1997;36:116-117. doi:10.1111/j.1600-0536.1997.tb00434.x
  31. Wolf R, Orion E, Matz H. Co-existing sensitivity to metronidazole and isothiazolinone. Clin Exp Dermatol. 2003;28:506-507. doi:10.1046/j.1365-2230.2003.01364.x
  32. Madsen JT, Thormann J, Kerre S, et al. Allergic contact dermatitis to topical metronidazole—3 cases. Contact Dermatitis. 2007;56:364-366. doi:10.1111/j.1600-0536.2006.01064.x
  33. Fernández-Jorge B, Goday Buján J, Fernández-Torres R, et al. Concomitant allergic contact dermatitis from diphenhydramine and metronidazole. Contact Dermatitis. 2008;59:115-116. doi:10.1111/j.1600-0536.2008.01332.x
  34. Madsen JT, Lorentzen HF, Paulsen E. Contact sensitization to metronidazole from possible occupational exposure. Contact Dermatitis. 2009;60:117-118. doi:10.1111/j.1600-0536.2008.01490.x
  35. Beutner KR, Lemke S, Calvarese B. A look at the safety of metronidazole 1% gel: cumulative irritation, contact sensitization, phototoxicity, and photoallergy potential. Cutis. 2006;77(4 suppl):12-17.
  36. Jappe U, Schäfer T, Schnuch A, et al. Contact allergy in patients with rosacea: a clinic-based, prospective epidemiological study. J Eur Acad Dermatol Venereol. 2008;22:1208-1214. doi:10.1111/j.1468-3083.2008.02778.x
  37. DeKoven JG, Silverberg JI, Warshaw EM, et al. North American Contact Dermatitis Group Patch Test Results: 2017-2018. Dermatitis. 2021;32:111-123. doi:10.1097/DER.0000000000000729
  38. Comaish JS, Cunliffe WJ. Absorption of drugs from varicose ulcers: a cause of anaphylaxis. Br J Clin Pract. 1967;21:97-98.
  39. Roupe G, Strannegård O. Anaphylactic shock elicited by topical administration of bacitracin. Arch Dermatol. 1969;100:450-452.
  40. Farley M, Pak H, Carregal V, et al. Anaphylaxis to topically applied bacitracin. Am J Contact Dermat. 1995;6:28-31.
  41. Barranco R, Tornero P, de Barrio M, et al. Type IV hypersensitivity to oral nystatin. Contact Dermatitis. 2001;45:60. doi:10.1034/j.1600-0536.2001.045001060.x
  42. Cooper SM, Shaw S. Contact allergy to nystatin: an unusual allergen. Contact Dermatitis. 1999;41:120. doi:10.1111/j.1600-0536.1999.tb06254.x
  43. Dooms-Goossens A, Matura M, Drieghe J, et al. Contact allergy to imidazoles used as antimycotic agents. Contact Dermatitis. 1995;33:73-77. doi:10.1111/j.1600-0536.1995.tb00504.x
  44. Pérez-Mesonero R, Schneller-Pavelescu L, Ochando-Ibernón G, et al. Is tioconazole contact dermatitis still a concern? bringing allergic contact dermatitis caused by topical tioconazole back into the spotlight. Contact Dermatitis. 2019;80:168-169.
  45. Tang MM, Corti MA, Stirnimann R, et al. Severe cutaneous allergic reactions following topical antifungal therapy. Contact Dermatitis. 2013;68:56-57.
  46. Goossens A, Linsen G. Contact allergy to antihistamines is not common. Contact Dermatitis. 1998;39:38. doi:10.1111/j.1600-0536.1998.tb05817.x
  47. Taylor JS, Praditsuwan P, Handel D, et al. Allergic contact dermatitis from doxepin cream. one-year patch test clinic experience. Arch Dermatol. 1996;132:515-518.
  48. Bilbao I, Aguirre A, Vicente JM, et al. Allergic contact dermatitis due to 5% doxepin cream. Contact Dermatitis. 1996;35:254-255. doi:10.1111/j.1600-0536.1996.tb02374.x
  49. Shelley WB, Shelley ED, Talanin NY. Self-potentiating allergic contact dermatitis caused by doxepin hydrochloride cream. J Am Acad Dermatol. 1996;34:143-144. doi:10.1016/s0190-9622(96)90864-6
  50. Wakelin SH, Rycroft RJ. Allergic contact dermatitis from doxepin. Contact Dermatitis. 1999;40:214. doi:10.1111/j.1600-0536.1999.tb06037.x
  51. Horn HM, Tidman MJ, Aldridge RD. Allergic contact dermatitis due to doxepin cream in a patient with dystrophic epidermolysis bullosa. Contact Dermatitis. 2001;45:115. doi:10.1034/j.1600-0536.2001.045002115.x
  52. Bonnel RA, La Grenade L, Karwoski CB, et al. Allergic contact dermatitis from topical doxepin: Food and Drug Administration’s postmarketing surveillance experience. J Am Acad Dermatol. 2003;48:294-296. doi:10.1067/mjd.2003.46
  53. Devleeschouwer V, Roelandts R, Garmyn M, et al. Allergic and photoallergic contact dermatitis from ketoprofen: results of (photo) patch testing and follow-up of 42 patients. Contact Dermatitis. 2008;58:159-166. doi:10.1111/j.1600-0536.2007.01296.x
  54. Foti C, Bonamonte D, Conserva A, et al. Allergic and photoallergic contact dermatitis from ketoprofen: evaluation of cross-reactivities by a combination of photopatch testing and computerized conformational analysis. Curr Pharm Des. 2008;14:2833-2839. doi:10.2174/138161208786369696
  55. Gulin SJ, Chiriac A. Diclofenac-induced allergic contact dermatitis: a series of four patients. Drug Saf Case Rep. 2016;3:15. doi:10.1007/s40800-016-0039-3
  56. Lakshmi C, Srinivas CR. Systemic (allergic) contact dermatitis to diclofenac. Indian J Dermatol Venereol Leprol. 2011;77:536. doi:10.4103/0378-6323.82424
  57. Beutner C, Forkel S, Kreipe K, et al. Contact allergy to topical diclofenac with systemic tolerance [published online August 22, 2021]. Contact Dermatitis. doi:10.1111/cod.13961
  58. Pan Y, Nixon R. Allergic contact dermatitis to topical preparations of bufexamac. Australas J Dermatol. 2012;53:207-210. doi:10.1111/j.1440-0960.2012.00876.x
  59. Nakada T, Matsuzawa Y. Allergic contact dermatitis syndrome from bufexamac for nursing infant. Dermatitis. 2012;23:185-186. doi:10.1097/DER.0b013e318260d774
  60. Kerr AC, Muller F, Ferguson J, et al. Occupational carprofen photoallergic contact dermatitis. Br J Dermatol. 2008;159:1303-1308. doi:10.1111/j.1365-2133.2008.08847.x
  61. Kiely C, Murphy G. Photoallergic contact dermatitis caused by occupational exposure to the canine non-steroidal anti-inflammatory drug carprofen. Contact Dermatitis. 2010;63:364-365. doi:10.1111/j.1600-0536.2010.01820.x
  62. Somberg J, Molnar J. Retrospective evaluation on the analgesic activities of 2 compounded topical creams and voltaren gel in chronic noncancer pain. Am J Ther. 2015;22:342-349. doi:10.1097/MJT.0000000000000275
  63. Lee HG, Grossman SK, Valdes-Rodriguez R, et al. Topical ketamine-amitriptyline-lidocaine for chronic pruritus: a retrospective study assessing efficacy and tolerability. J Am Acad Dermatol. 2017;76:760-761. doi:10.1016/j.jaad.2016.10.030
  64. Turrentine JE, Marrazzo G, Cruz PD Jr. Novel use of patch testing in the first report of allergic contact dermatitis to cyclobenzaprine. Dermatitis. 2015;26:60-61. doi:10.1097/DER.0000000000000099
  65. de Groot A. Patch Testing. 3rd ed. acdegroot publishing; 2008.
  66. de Groot A. Patch Testing. 4th ed. acdegroot publishing; 2018.
Article PDF
Author and Disclosure Information

Ms. Ng and Dr. Reeder are from the Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison. Dr. Atwater is from the Department of Dermatology, Duke University School of Medicine, Durham, North Carolina, and Eli Lilly and Company, Indianapolis, Indiana.

Ms. Ng and Dr. Reeder report no conflict of interest. Dr. Atwater is Immediate Past President of the American Contact Dermatitis Society (ACDS) and is an employee of Eli Lilly and Company.

This article is the first of a 2-part series. Part 2 will appear in January 2022.

Correspondence: Margo Reeder, MD, 1 S Park St, 7th Fl, Madison, WI 53715 (mreeder@dermatology.wisc.edu).

Issue
Cutis - 108(5)
Publications
Topics
Page Number
271-275
Sections
Author and Disclosure Information

Ms. Ng and Dr. Reeder are from the Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison. Dr. Atwater is from the Department of Dermatology, Duke University School of Medicine, Durham, North Carolina, and Eli Lilly and Company, Indianapolis, Indiana.

Ms. Ng and Dr. Reeder report no conflict of interest. Dr. Atwater is Immediate Past President of the American Contact Dermatitis Society (ACDS) and is an employee of Eli Lilly and Company.

This article is the first of a 2-part series. Part 2 will appear in January 2022.

Correspondence: Margo Reeder, MD, 1 S Park St, 7th Fl, Madison, WI 53715 (mreeder@dermatology.wisc.edu).

Author and Disclosure Information

Ms. Ng and Dr. Reeder are from the Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison. Dr. Atwater is from the Department of Dermatology, Duke University School of Medicine, Durham, North Carolina, and Eli Lilly and Company, Indianapolis, Indiana.

Ms. Ng and Dr. Reeder report no conflict of interest. Dr. Atwater is Immediate Past President of the American Contact Dermatitis Society (ACDS) and is an employee of Eli Lilly and Company.

This article is the first of a 2-part series. Part 2 will appear in January 2022.

Correspondence: Margo Reeder, MD, 1 S Park St, 7th Fl, Madison, WI 53715 (mreeder@dermatology.wisc.edu).

Article PDF
Article PDF

Topical medications frequently are prescribed in dermatology and provide the advantages of direct skin penetration and targeted application while typically sparing patients from systemic effects. Adverse cutaneous effects include allergic contact dermatitis (ACD), irritant contact dermatitis (ICD), photosensitivity, urticaria, hyperpigmentation or hypopigmentation, atrophy, periorificial dermatitis, and acneform eruptions. Allergic contact dermatitis can develop from the active drug or vehicle components.

Patients with medicament ACD often present with symptoms of pruritus and dermatitis at the site of topical application. They may express concern that the medication is no longer working or seems to be making things worse. Certain sites are more prone to developing medicament dermatitis, including the face, groin, and lower legs. Older adults may be more at risk. Other risk factors include pre-existing skin diseases such as stasis dermatitis, acne, psoriasis, atopic dermatitis, and genital dermatoses.1 A review of 14,911 patch-tested patients from a single referral clinic revealed that 17.4% had iatrogenic contact dermatitis, with the most common culprits being topical antibiotics, antiseptics, and steroids.2

In this 2-part series, we will focus on the active drug as a source of ACD. Part 1 explores ACD associated with acne and rosacea medications, antimicrobials, antihistamines, and topical pain preparations.

 

Acne and Rosacea Medications

Retinoids—Topical retinoids are first-line acne treatments that help normalize skin keratinization. Irritant contact dermatitis from retinoids is a well-known and common side effect. Although far less common than ICD, ACD from topical retinoid use has been reported.3,4 Reactions to tretinoin are most frequently reported in the literature compared to adapalene gel5 and tazarotene foam, which have lower potential for sensitization.6 Allergic contact dermatitis also has been reported from retinyl palmitate7,8 in cosmetic creams and from occupational exposure in settings of industrial vitamin A production.9 Both ICD and ACD from topical retinoids can present with pruritus, erythema, and scaling. Given this clinical overlap between ACD and ICD, patch testing is crucial in differentiating the underlying etiology of the dermatitis.

Benzoyl Peroxide—Benzoyl peroxide (BP) is another popular topical acne treatment that targets Cutibacterium acnes, a bacterium often implicated in the pathogenesis of acne vulgaris. Similar to retinoids, ICD is more common than ACD. Several cases of ACD to BP have been reported.10-14 Occasionally, honey-colored crusting associated with ACD to BP can mimic impetigo.10 Aside from use of BP as an acne treatment, other potential exposures to BP include bleached flour13 and orthopedic bone cement. Occupations at risk for potential BP exposure include dental technicians15 and those working in plastic manufacturing.

Brimonidine—Brimonidine tartrate is a selective α2-adrenergic agonist initially used to treat open-angle glaucoma and also is used as a topical treatment for rosacea. Allergic reactions to brimonidine eye drops may present with periorbital hyperpigmentation and pruritic bullous lesions.16 Case reports of topical brimonidine ACD have demonstrated mixed patch test results, with positive patch tests to Mirvaso (Galderma) as is but negative patch tests to pure brimonidine tartrate 0.33%.17,18 Ringuet and Houle19 reported the first known positive patch test reaction to pure topical brimonidine, testing with brimonidine tartrate 1% in petrolatum.20,21 Clinicians should be attuned to ACD to topical brimonidine in patients previously treated for glaucoma, as prior use of ophthalmic preparations may result in sensitization.18,20

Antimicrobials

Clindamycin—Clindamycin targets bacterial protein synthesis and is an effective adjunct in the treatment of acne. Despite its widespread and often long-term use, topical clindamycin is a weak sensitizer.22 To date, limited case reports on ACD to topical clindamycin exist.23-28 Rare clinical patterns of ACD to clindamycin include mimickers of irritant retinoid dermatitis, erythema multiforme, or pustular rosacea.25,26,29

 

 

Metronidazole—Metronidazole is a bactericidal agent that disrupts nucleic acid synthesis with additional anti-inflammatory properties used in the treatment of rosacea. Allergic contact dermatitis to topical metronidazole has been reported.30-34 In 2006, Beutner at al35 patch tested 215 patients using metronidazole gel 1%, which revealed no positive reactions to indicate contact sensitization. Similarly, Jappe et al36 found no positive reactions to metronidazole 2% in petrolatum in their prospective analysis of 78 rosacea patients, further highlighting the exceptionally low incidence of ACD. Cross-reaction with isothiazolinone, which shares structurally similar properties to metronidazole, has been speculated.31,34 One patient developed an acute reaction to metronidazole gel 0.75% within 24 hours of application, suggesting that isothiazolinone may act as a sensitizer, though this relationship has not been proven.31

Neomycin—Neomycin blocks bacterial protein synthesis and is available in both prescription and over-the-counter (OTC) formulations. It commonly is used to treat and prevent superficial wound infections as an OTC antibiotic and also has otic, ophthalmologic, gastroenterologic, urologic, and peritoneal formulations. It also can be used in the dental and veterinary fields and is present in some animal feeds and in trace amounts in some vaccines for humans. Neomycin is a common antibiotic contact allergen, and the most recently reported 2017-2018 North American Contact Dermatitis Group data cycle placed it at number 12 with 5.4% positivity.37 Co-reactions with bacitracin can occur, substantially limiting OTC topical antibiotic options for allergic patients. A safe alternative for patients with neomycin (and bacitracin and polymyxin) contact allergy is prescription mupirocin.

Bacitracin—Bacitracin interferes with peptidoglycan and cell-wall synthesis to treat superficial cutaneous infections. Similar to neomycin, it also can be found in OTC antibiotic ointments as well as in antibacterial bandages. There are several case reports of patients with both type IV delayed hypersensitivity (contact dermatitis) and type I anaphylactic reactions to bacitracin38-40; patch testers should be aware of this rare association. Bacitracin was positive in 5.5% of patch tested patients in the 2017-2018 North American Contact Dermatitis Group data cycle,37 and as with neomycin, bacitracin also is commonly patch tested in most screening patch test series.

Polymyxin—Polymyxin is a polypeptide topical antibiotic that is used to treat superficial wound infections and can be used in combination with neomycin and/or bacitracin. Historically, it is a less common antibiotic allergen; however, it is now frequently included in comprehensive patch test series, as the frequency of positive reactions seems to be increasing, probably due to polysensitization with neomycin and bacitracin.

Nystatin—Nystatin is an antifungal that binds to ergosterol and disrupts the cell wall. Cases exist of ACD to topical nystatin as well as systemic ACD from oral exposure, though both are quite rare. Authors have surmised that the overall low rates of ACD may be due to poor skin absorption of nystatin, which also can confound patch testing.41,42 For patients with suspected ACD to nystatin, repeat open application testing also can be performed to confirm allergy.

 

 

Imidazole Antifungals—Similar to nystatins, imidazole antifungals also work by disrupting the fungal cell wall. Imidazole antifungal preparations that have been reported to cause ACD include clotrimazole, miconazole, econazole, and isoconazole, and although cross-reactivity patterns have been described, they are not always reproducible with patch testing.43 In one reported case, tioconazole found in an antifungal nail lacquer triggered ACD involving not only the fingers and toes but also the trunk.44 Erythema multiforme–like reactions also have been described from topical use.45 Commercial patch test preparations of the most common imidazole allergens do exist. Nonimidazole antifungals remain a safe option for allergic patients.

Antihistamines

Antihistamines, or H1-receptor antagonists, are marketed to be applied topically for relief of pruritus associated with allergic cutaneous reactions. Ironically, they are known to be potent sensitizers themselves. There are 6 main chemical classes of antihistamines: phenothiazines, ethylenediamines, ethanolamines, alkylamines, piperazines, and piperidines. Goossens and Linsen46 patch tested 12,460 patients from 1978 to 1997 and found the most positive reactions to promethazine (phenothiazine)(n=12), followed by diphenhydramine (ethanolamine)(n=8) and clemizole (benzimidazole)(n=6). The authors also noted cross-reactions between diphenhydramine derivatives and between promethazine and chlorpromazine.46

Doxepin is a tricyclic antidepressant with antihistamine activity and is a well-documented sensitizer.47-52 Taylor et al47 evaluated 97 patients with chronic dermatoses, and patch testing revealed 17 (17.5%) positive reactions to doxepin cream, 13 (76.5%) of which were positive reactions to both the commercial cream and the active ingredient. Patch testing using doxepin dilution as low as 0.5% in petrolatum is sufficient to provoke a strong (++) allergic reaction.50,51 Early-onset ACD following the use of doxepin cream suggests the possibility of prior sensitization, perhaps with a structurally similar phenothiazine drug.51 A keen suspicion for ACD in patients using doxepin cream for longer than the recommended duration can help make the diagnosis.49,52

 

Topical Analgesics

Nonsteroidal Anti-inflammatory Drugs—Ketoprofen is one of the most frequent culprits of photoallergic contact dermatitis. Pruritic, papulovesicular, and bullous lesions typically develop acutely weeks after exposure. Prolonged photosensitivity is common and can last years after discontinuation of the nonsteroidal anti-inflammatory drug.53 Cases of cross-reactions and co-sensitization to structurally similar substances have been reported, including to benzophenone-related chemicals in sunscreen and aldehyde groups in fragrance mix.53,54

Diclofenac gel generally is well tolerated in the topical treatment of joint pain and inflammation. In the setting of ACD, patients typically present with dermatitis localized to the area of application.55 Immediate cessation and avoidance of topical diclofenac are crucial components of management. Although systemic contact dermatitis has been reported with oral diclofenac use,56 a recent report suggested that oral diclofenac may be well tolerated for some patients with topical ACD.57

 

 

Publications on bufexamac-induced ACD mainly consist of international reports, as this medication has been discontinued in the United States. Bufexamac is a highly sensitizing agent that can lead to severe polymorphic eruptions requiring treatment with prednisolone and even hospitalization.58 In one Australian case report, a mother developed an edematous, erythematous, papulovesicular eruption on the breast while breastfeeding her baby, who was being treated with bufexamac cream 5% for infantile eczema.59 Carprofen-induced photoallergic contact dermatitis is associated with occupational exposure in pharmaceutical workers.60,61 A few case reports on other nonsteroidal anti-inflammatory drugs, including etofenamate and aceclofenac, have been published.62,63

Compounded Medications—Compounded topical analgesics, which help to control pain via multiple combined effects, have gained increasing popularity in the management of chronic neuropathic pain disorders. Only a few recent retrospective studies assessing the efficacy and safety of these medications have mentioned suspected allergic cutaneous reactions.62,63 In 2015, Turrentine et al64 reported a case of ACD to cyclobenzaprine in a compound containing ketamine 10%, diclofenac 5%, baclofen 2%, bupivacaine 1%, cyclobenzaprine 2%, gabapentin 6%, ibuprofen 3%, and pentoxifylline 3% in a proprietary cream base. When patients present with suspected ACD to a compounded pain medication, obtaining individual components for patch testing is key to determining the allergic ingredient(s). We suspect that we will see a rise in reports of ACD as these topical compounds become readily adopted in clinical practices.

Patch Testing for Diagnosis

When patients present with symptoms concerning for ACD to medicaments, the astute clinician should promptly stop the suspected topical medication and consider patch testing. For common allergens such as neomycin, bacitracin, or ethylenediamine, commercial patch test preparations exist and should be used; however, for drugs that do not have a commercial patch test preparation, the patient’s product can be applied as is, keeping in mind that certain preparations (such as retinoids) can cause irritant patch test reactions, which may confound the reading. Alternatively, individual ingredients in the medication’s formulation can be requested from the manufacturer or a compounding pharmacy for targeted testing. Suggested concentrations for patch testing based on the literature and expert reference are listed in the Table. The authors (M.R., A.R.A.) frequently rely on an expert reference66 to determine ideal concentrations for patch testing. Referral to a specialized patch test clinic may be appropriate.

 

Final Interpretation

Although their intent is to heal, topical medicaments also can be a source of ACD. The astute clinician should consider ACD when topicals either no longer seem to help the patient or trigger new-onset dermatitis. Patch testing directly with the culprit medicament, or individual medication ingredients when needed, can lead to the diagnosis, though caution is advised. Stay tuned for part 2 of this series in which we will discuss ACD to topical steroids, immunomodulators, and anesthetic medications.

Topical medications frequently are prescribed in dermatology and provide the advantages of direct skin penetration and targeted application while typically sparing patients from systemic effects. Adverse cutaneous effects include allergic contact dermatitis (ACD), irritant contact dermatitis (ICD), photosensitivity, urticaria, hyperpigmentation or hypopigmentation, atrophy, periorificial dermatitis, and acneform eruptions. Allergic contact dermatitis can develop from the active drug or vehicle components.

Patients with medicament ACD often present with symptoms of pruritus and dermatitis at the site of topical application. They may express concern that the medication is no longer working or seems to be making things worse. Certain sites are more prone to developing medicament dermatitis, including the face, groin, and lower legs. Older adults may be more at risk. Other risk factors include pre-existing skin diseases such as stasis dermatitis, acne, psoriasis, atopic dermatitis, and genital dermatoses.1 A review of 14,911 patch-tested patients from a single referral clinic revealed that 17.4% had iatrogenic contact dermatitis, with the most common culprits being topical antibiotics, antiseptics, and steroids.2

In this 2-part series, we will focus on the active drug as a source of ACD. Part 1 explores ACD associated with acne and rosacea medications, antimicrobials, antihistamines, and topical pain preparations.

 

Acne and Rosacea Medications

Retinoids—Topical retinoids are first-line acne treatments that help normalize skin keratinization. Irritant contact dermatitis from retinoids is a well-known and common side effect. Although far less common than ICD, ACD from topical retinoid use has been reported.3,4 Reactions to tretinoin are most frequently reported in the literature compared to adapalene gel5 and tazarotene foam, which have lower potential for sensitization.6 Allergic contact dermatitis also has been reported from retinyl palmitate7,8 in cosmetic creams and from occupational exposure in settings of industrial vitamin A production.9 Both ICD and ACD from topical retinoids can present with pruritus, erythema, and scaling. Given this clinical overlap between ACD and ICD, patch testing is crucial in differentiating the underlying etiology of the dermatitis.

Benzoyl Peroxide—Benzoyl peroxide (BP) is another popular topical acne treatment that targets Cutibacterium acnes, a bacterium often implicated in the pathogenesis of acne vulgaris. Similar to retinoids, ICD is more common than ACD. Several cases of ACD to BP have been reported.10-14 Occasionally, honey-colored crusting associated with ACD to BP can mimic impetigo.10 Aside from use of BP as an acne treatment, other potential exposures to BP include bleached flour13 and orthopedic bone cement. Occupations at risk for potential BP exposure include dental technicians15 and those working in plastic manufacturing.

Brimonidine—Brimonidine tartrate is a selective α2-adrenergic agonist initially used to treat open-angle glaucoma and also is used as a topical treatment for rosacea. Allergic reactions to brimonidine eye drops may present with periorbital hyperpigmentation and pruritic bullous lesions.16 Case reports of topical brimonidine ACD have demonstrated mixed patch test results, with positive patch tests to Mirvaso (Galderma) as is but negative patch tests to pure brimonidine tartrate 0.33%.17,18 Ringuet and Houle19 reported the first known positive patch test reaction to pure topical brimonidine, testing with brimonidine tartrate 1% in petrolatum.20,21 Clinicians should be attuned to ACD to topical brimonidine in patients previously treated for glaucoma, as prior use of ophthalmic preparations may result in sensitization.18,20

Antimicrobials

Clindamycin—Clindamycin targets bacterial protein synthesis and is an effective adjunct in the treatment of acne. Despite its widespread and often long-term use, topical clindamycin is a weak sensitizer.22 To date, limited case reports on ACD to topical clindamycin exist.23-28 Rare clinical patterns of ACD to clindamycin include mimickers of irritant retinoid dermatitis, erythema multiforme, or pustular rosacea.25,26,29

 

 

Metronidazole—Metronidazole is a bactericidal agent that disrupts nucleic acid synthesis with additional anti-inflammatory properties used in the treatment of rosacea. Allergic contact dermatitis to topical metronidazole has been reported.30-34 In 2006, Beutner at al35 patch tested 215 patients using metronidazole gel 1%, which revealed no positive reactions to indicate contact sensitization. Similarly, Jappe et al36 found no positive reactions to metronidazole 2% in petrolatum in their prospective analysis of 78 rosacea patients, further highlighting the exceptionally low incidence of ACD. Cross-reaction with isothiazolinone, which shares structurally similar properties to metronidazole, has been speculated.31,34 One patient developed an acute reaction to metronidazole gel 0.75% within 24 hours of application, suggesting that isothiazolinone may act as a sensitizer, though this relationship has not been proven.31

Neomycin—Neomycin blocks bacterial protein synthesis and is available in both prescription and over-the-counter (OTC) formulations. It commonly is used to treat and prevent superficial wound infections as an OTC antibiotic and also has otic, ophthalmologic, gastroenterologic, urologic, and peritoneal formulations. It also can be used in the dental and veterinary fields and is present in some animal feeds and in trace amounts in some vaccines for humans. Neomycin is a common antibiotic contact allergen, and the most recently reported 2017-2018 North American Contact Dermatitis Group data cycle placed it at number 12 with 5.4% positivity.37 Co-reactions with bacitracin can occur, substantially limiting OTC topical antibiotic options for allergic patients. A safe alternative for patients with neomycin (and bacitracin and polymyxin) contact allergy is prescription mupirocin.

Bacitracin—Bacitracin interferes with peptidoglycan and cell-wall synthesis to treat superficial cutaneous infections. Similar to neomycin, it also can be found in OTC antibiotic ointments as well as in antibacterial bandages. There are several case reports of patients with both type IV delayed hypersensitivity (contact dermatitis) and type I anaphylactic reactions to bacitracin38-40; patch testers should be aware of this rare association. Bacitracin was positive in 5.5% of patch tested patients in the 2017-2018 North American Contact Dermatitis Group data cycle,37 and as with neomycin, bacitracin also is commonly patch tested in most screening patch test series.

Polymyxin—Polymyxin is a polypeptide topical antibiotic that is used to treat superficial wound infections and can be used in combination with neomycin and/or bacitracin. Historically, it is a less common antibiotic allergen; however, it is now frequently included in comprehensive patch test series, as the frequency of positive reactions seems to be increasing, probably due to polysensitization with neomycin and bacitracin.

Nystatin—Nystatin is an antifungal that binds to ergosterol and disrupts the cell wall. Cases exist of ACD to topical nystatin as well as systemic ACD from oral exposure, though both are quite rare. Authors have surmised that the overall low rates of ACD may be due to poor skin absorption of nystatin, which also can confound patch testing.41,42 For patients with suspected ACD to nystatin, repeat open application testing also can be performed to confirm allergy.

 

 

Imidazole Antifungals—Similar to nystatins, imidazole antifungals also work by disrupting the fungal cell wall. Imidazole antifungal preparations that have been reported to cause ACD include clotrimazole, miconazole, econazole, and isoconazole, and although cross-reactivity patterns have been described, they are not always reproducible with patch testing.43 In one reported case, tioconazole found in an antifungal nail lacquer triggered ACD involving not only the fingers and toes but also the trunk.44 Erythema multiforme–like reactions also have been described from topical use.45 Commercial patch test preparations of the most common imidazole allergens do exist. Nonimidazole antifungals remain a safe option for allergic patients.

Antihistamines

Antihistamines, or H1-receptor antagonists, are marketed to be applied topically for relief of pruritus associated with allergic cutaneous reactions. Ironically, they are known to be potent sensitizers themselves. There are 6 main chemical classes of antihistamines: phenothiazines, ethylenediamines, ethanolamines, alkylamines, piperazines, and piperidines. Goossens and Linsen46 patch tested 12,460 patients from 1978 to 1997 and found the most positive reactions to promethazine (phenothiazine)(n=12), followed by diphenhydramine (ethanolamine)(n=8) and clemizole (benzimidazole)(n=6). The authors also noted cross-reactions between diphenhydramine derivatives and between promethazine and chlorpromazine.46

Doxepin is a tricyclic antidepressant with antihistamine activity and is a well-documented sensitizer.47-52 Taylor et al47 evaluated 97 patients with chronic dermatoses, and patch testing revealed 17 (17.5%) positive reactions to doxepin cream, 13 (76.5%) of which were positive reactions to both the commercial cream and the active ingredient. Patch testing using doxepin dilution as low as 0.5% in petrolatum is sufficient to provoke a strong (++) allergic reaction.50,51 Early-onset ACD following the use of doxepin cream suggests the possibility of prior sensitization, perhaps with a structurally similar phenothiazine drug.51 A keen suspicion for ACD in patients using doxepin cream for longer than the recommended duration can help make the diagnosis.49,52

 

Topical Analgesics

Nonsteroidal Anti-inflammatory Drugs—Ketoprofen is one of the most frequent culprits of photoallergic contact dermatitis. Pruritic, papulovesicular, and bullous lesions typically develop acutely weeks after exposure. Prolonged photosensitivity is common and can last years after discontinuation of the nonsteroidal anti-inflammatory drug.53 Cases of cross-reactions and co-sensitization to structurally similar substances have been reported, including to benzophenone-related chemicals in sunscreen and aldehyde groups in fragrance mix.53,54

Diclofenac gel generally is well tolerated in the topical treatment of joint pain and inflammation. In the setting of ACD, patients typically present with dermatitis localized to the area of application.55 Immediate cessation and avoidance of topical diclofenac are crucial components of management. Although systemic contact dermatitis has been reported with oral diclofenac use,56 a recent report suggested that oral diclofenac may be well tolerated for some patients with topical ACD.57

 

 

Publications on bufexamac-induced ACD mainly consist of international reports, as this medication has been discontinued in the United States. Bufexamac is a highly sensitizing agent that can lead to severe polymorphic eruptions requiring treatment with prednisolone and even hospitalization.58 In one Australian case report, a mother developed an edematous, erythematous, papulovesicular eruption on the breast while breastfeeding her baby, who was being treated with bufexamac cream 5% for infantile eczema.59 Carprofen-induced photoallergic contact dermatitis is associated with occupational exposure in pharmaceutical workers.60,61 A few case reports on other nonsteroidal anti-inflammatory drugs, including etofenamate and aceclofenac, have been published.62,63

Compounded Medications—Compounded topical analgesics, which help to control pain via multiple combined effects, have gained increasing popularity in the management of chronic neuropathic pain disorders. Only a few recent retrospective studies assessing the efficacy and safety of these medications have mentioned suspected allergic cutaneous reactions.62,63 In 2015, Turrentine et al64 reported a case of ACD to cyclobenzaprine in a compound containing ketamine 10%, diclofenac 5%, baclofen 2%, bupivacaine 1%, cyclobenzaprine 2%, gabapentin 6%, ibuprofen 3%, and pentoxifylline 3% in a proprietary cream base. When patients present with suspected ACD to a compounded pain medication, obtaining individual components for patch testing is key to determining the allergic ingredient(s). We suspect that we will see a rise in reports of ACD as these topical compounds become readily adopted in clinical practices.

Patch Testing for Diagnosis

When patients present with symptoms concerning for ACD to medicaments, the astute clinician should promptly stop the suspected topical medication and consider patch testing. For common allergens such as neomycin, bacitracin, or ethylenediamine, commercial patch test preparations exist and should be used; however, for drugs that do not have a commercial patch test preparation, the patient’s product can be applied as is, keeping in mind that certain preparations (such as retinoids) can cause irritant patch test reactions, which may confound the reading. Alternatively, individual ingredients in the medication’s formulation can be requested from the manufacturer or a compounding pharmacy for targeted testing. Suggested concentrations for patch testing based on the literature and expert reference are listed in the Table. The authors (M.R., A.R.A.) frequently rely on an expert reference66 to determine ideal concentrations for patch testing. Referral to a specialized patch test clinic may be appropriate.

 

Final Interpretation

Although their intent is to heal, topical medicaments also can be a source of ACD. The astute clinician should consider ACD when topicals either no longer seem to help the patient or trigger new-onset dermatitis. Patch testing directly with the culprit medicament, or individual medication ingredients when needed, can lead to the diagnosis, though caution is advised. Stay tuned for part 2 of this series in which we will discuss ACD to topical steroids, immunomodulators, and anesthetic medications.

References
  1. Davis MD. Unusual patterns in contact dermatitis: medicaments. Dermatol Clin. 2009;27:289-297, vi. doi:10.1016/j.det.2009.05.003
  2. Gilissen L, Goossens A. Frequency and trends of contact allergy to and iatrogenic contact dermatitis caused by topical drugs over a 25-year period. Contact Dermatitis. 2016;75:290-302. doi:10.1111/cod.12621
  3. Balato N, Patruno C, Lembo G, et al. Allergic contact dermatitis from retinoic acid. Contact Dermatitis. 1995;32:51. doi:10.1111/j.1600-0536.1995.tb00846.x
  4. Berg JE, Bowman JP, Saenz AB. Cumulative irritation potential and contact sensitization potential of tazarotene foam 0.1% in 2 phase 1 patch studies. Cutis. 2012;90:206-211.
  5. Numata T, Jo R, Kobayashi Y, et al. Allergic contact dermatitis caused by adapalene. Contact Dermatitis. 2015;73:187-188. doi:10.1111/cod.12410
  6. Anderson A, Gebauer K. Periorbital allergic contact dermatitis resulting from topical retinoic acid use. Australas J Dermatol. 2014;55:152-153. doi:10.1111/ajd.12041
  7. Blondeel A. Contact allergy to vitamin A. Contact Dermatitis. 1984;11:191-192. doi:10.1111/j.1600-0536.1984.tb00976.x
  8. Manzano D, Aguirre A, Gardeazabal J, et al. Allergic contact dermatitis from tocopheryl acetate (vitamin E) and retinol palmitate (vitamin A) in a moisturizing cream. Contact Dermatitis. 1994;31:324. doi:10.1111/j.1600-0536.1994.tb02030.x
  9. Heidenheim M, Jemec GB. Occupational allergic contact dermatitis from vitamin A acetate. Contact Dermatitis. 1995;33:439. doi:10.1111/j.1600-0536.1995.tb02091.x
  10. Kim C, Craiglow BG, Watsky KL, et al. Allergic contact dermatitis to benzoyl peroxide resembling impetigo. Pediatr Dermatol. 2015;32:E161-E162. doi:10.1111/pde.12585
  11. Sandre M, Skotnicki-Grant S. A case of a paediatric patient with allergic contact dermatitis to benzoyl peroxide. J Cutan Med Surg. 2018;22:226-228. doi:10.1177/1203475417733462
  12. Corazza M, Amendolagine G, Musmeci D, et al. Sometimes even Dr Google is wrong: an unusual contact dermatitis caused by benzoyl peroxide. Contact Dermatitis. 2018;79:380-381. doi:10.1111/cod.13086
  13. Adelman M, Mohammad T, Kerr H. Allergic contact dermatitis due to benzoyl peroxide from an unlikely source. Dermatitis. 2019;30:230-231. doi:10.1097/DER.0000000000000470
  14. Gatica-Ortega ME, Pastor-Nieto MA. Allergic contact dermatitis to Glycyrrhiza inflata root extract in an anti-acne cosmetic product [published online April 28, 2021]. Contact Dermatitis. doi:10.1111/cod.13872
  15. Ockenfels HM, Uter W, Lessmann H, et al. Patch testing with benzoyl peroxide: reaction profile and interpretation of positive patch test reactions. Contact Dermatitis. 2009;61:209-216. doi:10.1111/j.1600-0536.2009.01603.x
  16. Sodhi PK, Verma L, Ratan J. Dermatological side effects of brimonidine: a report of three cases. J Dermatol. 2003;30:697-700. doi:10.1111/j.1346-8138.2003.tb00461.x
  17. Swanson LA, Warshaw EM. Allergic contact dermatitis to topical brimonidine tartrate gel 0.33% for treatment of rosacea. J Am Acad Dermatol. 2014;71:832-833. doi:10.1016/j.jaad.2014.05.073
  18. Bangsgaard N, Fischer LA, Zachariae C. Sensitization to and allergic contact dermatitis caused by Mirvaso(®)(brimonidine tartrate) for treatment of rosacea—2 cases. Contact Dermatitis. 2016;74:378-379. doi:10.1111/cod.12547
  19. Ringuet J, Houle MC. Case report: allergic contact dermatitis to topical brimonidine demonstrated with patch testing: insights on evaluation of brimonidine sensitization. J Cutan Med Surg. 2018;22:636-638. doi:10.1177/1203475418789020
  20. Cookson H, McFadden J, White J, et al. Allergic contact dermatitis caused by Mirvaso®, brimonidine tartrate gel 0.33%, a new topical treatment for rosaceal erythema. Contact Dermatitis. 2015;73:366-367. doi:10.1111/cod.12476
  21. Rajagopalan A, Rajagopalan B. Allergic contact dermatitis to topical brimonidine. Australas J Dermatol. 2015;56:235. doi:10.1111/ajd.12299
  22. Veraldi S, Brena M, Barbareschi M. Allergic contact dermatitis caused by topical antiacne drugs. Expert Rev Clin Pharmacol. 2015;8:377-381. doi:10.1586/17512433.2015.1046839
  23. Vejlstrup E, Menné T. Contact dermatitis from clindamycin. Contact Dermatitis. 1995;32:110. doi:10.1111/j.1600-0536.1995.tb00759.x
  24. García R, Galindo PA, Feo F, et al. Delayed allergic reactions to amoxycillin and clindamycin. Contact Dermatitis. 1996;35:116-117. doi:10.1111/j.1600-0536.1996.tb02312.x
  25. Muñoz D, Del Pozo MD, Audicana M, et al. Erythema-multiforme-like eruption from antibiotics of 3 different groups. Contact Dermatitis. 1996;34:227-228. doi:10.1111/j.1600-0536.1996.tb02187.x
  26. Romita P, Ettorre G, Corazza M, et al. Allergic contact dermatitis caused by clindamycin mimicking ‘retinoid flare.’ Contact Dermatitis. 2017;77:181-182. doi:10.1111/cod.12784
  27. Veraldi S, Guanziroli E, Ferrucci S, et al. Allergic contact dermatitis caused by clindamycin. Contact Dermatitis. 2019;80:68-69. doi:10.1111/cod.13133
  28. Voller LM, Kullberg SA, Warshaw EM. Axillary allergic contact dermatitis to topical clindamycin. Contact Dermatitis. 2020;82:313-314. doi:10.1111/cod.13465
  29. de Kort WJ, de Groot AC. Clindamycin allergy presenting as rosacea. Contact Dermatitis. 1989;20:72-73. doi:10.1111/j.1600-0536.1989.tb03108.x
  30. Vincenzi C, Lucente P, Ricci C, et al. Facial contact dermatitis due to metronidazole. Contact Dermatitis. 1997;36:116-117. doi:10.1111/j.1600-0536.1997.tb00434.x
  31. Wolf R, Orion E, Matz H. Co-existing sensitivity to metronidazole and isothiazolinone. Clin Exp Dermatol. 2003;28:506-507. doi:10.1046/j.1365-2230.2003.01364.x
  32. Madsen JT, Thormann J, Kerre S, et al. Allergic contact dermatitis to topical metronidazole—3 cases. Contact Dermatitis. 2007;56:364-366. doi:10.1111/j.1600-0536.2006.01064.x
  33. Fernández-Jorge B, Goday Buján J, Fernández-Torres R, et al. Concomitant allergic contact dermatitis from diphenhydramine and metronidazole. Contact Dermatitis. 2008;59:115-116. doi:10.1111/j.1600-0536.2008.01332.x
  34. Madsen JT, Lorentzen HF, Paulsen E. Contact sensitization to metronidazole from possible occupational exposure. Contact Dermatitis. 2009;60:117-118. doi:10.1111/j.1600-0536.2008.01490.x
  35. Beutner KR, Lemke S, Calvarese B. A look at the safety of metronidazole 1% gel: cumulative irritation, contact sensitization, phototoxicity, and photoallergy potential. Cutis. 2006;77(4 suppl):12-17.
  36. Jappe U, Schäfer T, Schnuch A, et al. Contact allergy in patients with rosacea: a clinic-based, prospective epidemiological study. J Eur Acad Dermatol Venereol. 2008;22:1208-1214. doi:10.1111/j.1468-3083.2008.02778.x
  37. DeKoven JG, Silverberg JI, Warshaw EM, et al. North American Contact Dermatitis Group Patch Test Results: 2017-2018. Dermatitis. 2021;32:111-123. doi:10.1097/DER.0000000000000729
  38. Comaish JS, Cunliffe WJ. Absorption of drugs from varicose ulcers: a cause of anaphylaxis. Br J Clin Pract. 1967;21:97-98.
  39. Roupe G, Strannegård O. Anaphylactic shock elicited by topical administration of bacitracin. Arch Dermatol. 1969;100:450-452.
  40. Farley M, Pak H, Carregal V, et al. Anaphylaxis to topically applied bacitracin. Am J Contact Dermat. 1995;6:28-31.
  41. Barranco R, Tornero P, de Barrio M, et al. Type IV hypersensitivity to oral nystatin. Contact Dermatitis. 2001;45:60. doi:10.1034/j.1600-0536.2001.045001060.x
  42. Cooper SM, Shaw S. Contact allergy to nystatin: an unusual allergen. Contact Dermatitis. 1999;41:120. doi:10.1111/j.1600-0536.1999.tb06254.x
  43. Dooms-Goossens A, Matura M, Drieghe J, et al. Contact allergy to imidazoles used as antimycotic agents. Contact Dermatitis. 1995;33:73-77. doi:10.1111/j.1600-0536.1995.tb00504.x
  44. Pérez-Mesonero R, Schneller-Pavelescu L, Ochando-Ibernón G, et al. Is tioconazole contact dermatitis still a concern? bringing allergic contact dermatitis caused by topical tioconazole back into the spotlight. Contact Dermatitis. 2019;80:168-169.
  45. Tang MM, Corti MA, Stirnimann R, et al. Severe cutaneous allergic reactions following topical antifungal therapy. Contact Dermatitis. 2013;68:56-57.
  46. Goossens A, Linsen G. Contact allergy to antihistamines is not common. Contact Dermatitis. 1998;39:38. doi:10.1111/j.1600-0536.1998.tb05817.x
  47. Taylor JS, Praditsuwan P, Handel D, et al. Allergic contact dermatitis from doxepin cream. one-year patch test clinic experience. Arch Dermatol. 1996;132:515-518.
  48. Bilbao I, Aguirre A, Vicente JM, et al. Allergic contact dermatitis due to 5% doxepin cream. Contact Dermatitis. 1996;35:254-255. doi:10.1111/j.1600-0536.1996.tb02374.x
  49. Shelley WB, Shelley ED, Talanin NY. Self-potentiating allergic contact dermatitis caused by doxepin hydrochloride cream. J Am Acad Dermatol. 1996;34:143-144. doi:10.1016/s0190-9622(96)90864-6
  50. Wakelin SH, Rycroft RJ. Allergic contact dermatitis from doxepin. Contact Dermatitis. 1999;40:214. doi:10.1111/j.1600-0536.1999.tb06037.x
  51. Horn HM, Tidman MJ, Aldridge RD. Allergic contact dermatitis due to doxepin cream in a patient with dystrophic epidermolysis bullosa. Contact Dermatitis. 2001;45:115. doi:10.1034/j.1600-0536.2001.045002115.x
  52. Bonnel RA, La Grenade L, Karwoski CB, et al. Allergic contact dermatitis from topical doxepin: Food and Drug Administration’s postmarketing surveillance experience. J Am Acad Dermatol. 2003;48:294-296. doi:10.1067/mjd.2003.46
  53. Devleeschouwer V, Roelandts R, Garmyn M, et al. Allergic and photoallergic contact dermatitis from ketoprofen: results of (photo) patch testing and follow-up of 42 patients. Contact Dermatitis. 2008;58:159-166. doi:10.1111/j.1600-0536.2007.01296.x
  54. Foti C, Bonamonte D, Conserva A, et al. Allergic and photoallergic contact dermatitis from ketoprofen: evaluation of cross-reactivities by a combination of photopatch testing and computerized conformational analysis. Curr Pharm Des. 2008;14:2833-2839. doi:10.2174/138161208786369696
  55. Gulin SJ, Chiriac A. Diclofenac-induced allergic contact dermatitis: a series of four patients. Drug Saf Case Rep. 2016;3:15. doi:10.1007/s40800-016-0039-3
  56. Lakshmi C, Srinivas CR. Systemic (allergic) contact dermatitis to diclofenac. Indian J Dermatol Venereol Leprol. 2011;77:536. doi:10.4103/0378-6323.82424
  57. Beutner C, Forkel S, Kreipe K, et al. Contact allergy to topical diclofenac with systemic tolerance [published online August 22, 2021]. Contact Dermatitis. doi:10.1111/cod.13961
  58. Pan Y, Nixon R. Allergic contact dermatitis to topical preparations of bufexamac. Australas J Dermatol. 2012;53:207-210. doi:10.1111/j.1440-0960.2012.00876.x
  59. Nakada T, Matsuzawa Y. Allergic contact dermatitis syndrome from bufexamac for nursing infant. Dermatitis. 2012;23:185-186. doi:10.1097/DER.0b013e318260d774
  60. Kerr AC, Muller F, Ferguson J, et al. Occupational carprofen photoallergic contact dermatitis. Br J Dermatol. 2008;159:1303-1308. doi:10.1111/j.1365-2133.2008.08847.x
  61. Kiely C, Murphy G. Photoallergic contact dermatitis caused by occupational exposure to the canine non-steroidal anti-inflammatory drug carprofen. Contact Dermatitis. 2010;63:364-365. doi:10.1111/j.1600-0536.2010.01820.x
  62. Somberg J, Molnar J. Retrospective evaluation on the analgesic activities of 2 compounded topical creams and voltaren gel in chronic noncancer pain. Am J Ther. 2015;22:342-349. doi:10.1097/MJT.0000000000000275
  63. Lee HG, Grossman SK, Valdes-Rodriguez R, et al. Topical ketamine-amitriptyline-lidocaine for chronic pruritus: a retrospective study assessing efficacy and tolerability. J Am Acad Dermatol. 2017;76:760-761. doi:10.1016/j.jaad.2016.10.030
  64. Turrentine JE, Marrazzo G, Cruz PD Jr. Novel use of patch testing in the first report of allergic contact dermatitis to cyclobenzaprine. Dermatitis. 2015;26:60-61. doi:10.1097/DER.0000000000000099
  65. de Groot A. Patch Testing. 3rd ed. acdegroot publishing; 2008.
  66. de Groot A. Patch Testing. 4th ed. acdegroot publishing; 2018.
References
  1. Davis MD. Unusual patterns in contact dermatitis: medicaments. Dermatol Clin. 2009;27:289-297, vi. doi:10.1016/j.det.2009.05.003
  2. Gilissen L, Goossens A. Frequency and trends of contact allergy to and iatrogenic contact dermatitis caused by topical drugs over a 25-year period. Contact Dermatitis. 2016;75:290-302. doi:10.1111/cod.12621
  3. Balato N, Patruno C, Lembo G, et al. Allergic contact dermatitis from retinoic acid. Contact Dermatitis. 1995;32:51. doi:10.1111/j.1600-0536.1995.tb00846.x
  4. Berg JE, Bowman JP, Saenz AB. Cumulative irritation potential and contact sensitization potential of tazarotene foam 0.1% in 2 phase 1 patch studies. Cutis. 2012;90:206-211.
  5. Numata T, Jo R, Kobayashi Y, et al. Allergic contact dermatitis caused by adapalene. Contact Dermatitis. 2015;73:187-188. doi:10.1111/cod.12410
  6. Anderson A, Gebauer K. Periorbital allergic contact dermatitis resulting from topical retinoic acid use. Australas J Dermatol. 2014;55:152-153. doi:10.1111/ajd.12041
  7. Blondeel A. Contact allergy to vitamin A. Contact Dermatitis. 1984;11:191-192. doi:10.1111/j.1600-0536.1984.tb00976.x
  8. Manzano D, Aguirre A, Gardeazabal J, et al. Allergic contact dermatitis from tocopheryl acetate (vitamin E) and retinol palmitate (vitamin A) in a moisturizing cream. Contact Dermatitis. 1994;31:324. doi:10.1111/j.1600-0536.1994.tb02030.x
  9. Heidenheim M, Jemec GB. Occupational allergic contact dermatitis from vitamin A acetate. Contact Dermatitis. 1995;33:439. doi:10.1111/j.1600-0536.1995.tb02091.x
  10. Kim C, Craiglow BG, Watsky KL, et al. Allergic contact dermatitis to benzoyl peroxide resembling impetigo. Pediatr Dermatol. 2015;32:E161-E162. doi:10.1111/pde.12585
  11. Sandre M, Skotnicki-Grant S. A case of a paediatric patient with allergic contact dermatitis to benzoyl peroxide. J Cutan Med Surg. 2018;22:226-228. doi:10.1177/1203475417733462
  12. Corazza M, Amendolagine G, Musmeci D, et al. Sometimes even Dr Google is wrong: an unusual contact dermatitis caused by benzoyl peroxide. Contact Dermatitis. 2018;79:380-381. doi:10.1111/cod.13086
  13. Adelman M, Mohammad T, Kerr H. Allergic contact dermatitis due to benzoyl peroxide from an unlikely source. Dermatitis. 2019;30:230-231. doi:10.1097/DER.0000000000000470
  14. Gatica-Ortega ME, Pastor-Nieto MA. Allergic contact dermatitis to Glycyrrhiza inflata root extract in an anti-acne cosmetic product [published online April 28, 2021]. Contact Dermatitis. doi:10.1111/cod.13872
  15. Ockenfels HM, Uter W, Lessmann H, et al. Patch testing with benzoyl peroxide: reaction profile and interpretation of positive patch test reactions. Contact Dermatitis. 2009;61:209-216. doi:10.1111/j.1600-0536.2009.01603.x
  16. Sodhi PK, Verma L, Ratan J. Dermatological side effects of brimonidine: a report of three cases. J Dermatol. 2003;30:697-700. doi:10.1111/j.1346-8138.2003.tb00461.x
  17. Swanson LA, Warshaw EM. Allergic contact dermatitis to topical brimonidine tartrate gel 0.33% for treatment of rosacea. J Am Acad Dermatol. 2014;71:832-833. doi:10.1016/j.jaad.2014.05.073
  18. Bangsgaard N, Fischer LA, Zachariae C. Sensitization to and allergic contact dermatitis caused by Mirvaso(®)(brimonidine tartrate) for treatment of rosacea—2 cases. Contact Dermatitis. 2016;74:378-379. doi:10.1111/cod.12547
  19. Ringuet J, Houle MC. Case report: allergic contact dermatitis to topical brimonidine demonstrated with patch testing: insights on evaluation of brimonidine sensitization. J Cutan Med Surg. 2018;22:636-638. doi:10.1177/1203475418789020
  20. Cookson H, McFadden J, White J, et al. Allergic contact dermatitis caused by Mirvaso®, brimonidine tartrate gel 0.33%, a new topical treatment for rosaceal erythema. Contact Dermatitis. 2015;73:366-367. doi:10.1111/cod.12476
  21. Rajagopalan A, Rajagopalan B. Allergic contact dermatitis to topical brimonidine. Australas J Dermatol. 2015;56:235. doi:10.1111/ajd.12299
  22. Veraldi S, Brena M, Barbareschi M. Allergic contact dermatitis caused by topical antiacne drugs. Expert Rev Clin Pharmacol. 2015;8:377-381. doi:10.1586/17512433.2015.1046839
  23. Vejlstrup E, Menné T. Contact dermatitis from clindamycin. Contact Dermatitis. 1995;32:110. doi:10.1111/j.1600-0536.1995.tb00759.x
  24. García R, Galindo PA, Feo F, et al. Delayed allergic reactions to amoxycillin and clindamycin. Contact Dermatitis. 1996;35:116-117. doi:10.1111/j.1600-0536.1996.tb02312.x
  25. Muñoz D, Del Pozo MD, Audicana M, et al. Erythema-multiforme-like eruption from antibiotics of 3 different groups. Contact Dermatitis. 1996;34:227-228. doi:10.1111/j.1600-0536.1996.tb02187.x
  26. Romita P, Ettorre G, Corazza M, et al. Allergic contact dermatitis caused by clindamycin mimicking ‘retinoid flare.’ Contact Dermatitis. 2017;77:181-182. doi:10.1111/cod.12784
  27. Veraldi S, Guanziroli E, Ferrucci S, et al. Allergic contact dermatitis caused by clindamycin. Contact Dermatitis. 2019;80:68-69. doi:10.1111/cod.13133
  28. Voller LM, Kullberg SA, Warshaw EM. Axillary allergic contact dermatitis to topical clindamycin. Contact Dermatitis. 2020;82:313-314. doi:10.1111/cod.13465
  29. de Kort WJ, de Groot AC. Clindamycin allergy presenting as rosacea. Contact Dermatitis. 1989;20:72-73. doi:10.1111/j.1600-0536.1989.tb03108.x
  30. Vincenzi C, Lucente P, Ricci C, et al. Facial contact dermatitis due to metronidazole. Contact Dermatitis. 1997;36:116-117. doi:10.1111/j.1600-0536.1997.tb00434.x
  31. Wolf R, Orion E, Matz H. Co-existing sensitivity to metronidazole and isothiazolinone. Clin Exp Dermatol. 2003;28:506-507. doi:10.1046/j.1365-2230.2003.01364.x
  32. Madsen JT, Thormann J, Kerre S, et al. Allergic contact dermatitis to topical metronidazole—3 cases. Contact Dermatitis. 2007;56:364-366. doi:10.1111/j.1600-0536.2006.01064.x
  33. Fernández-Jorge B, Goday Buján J, Fernández-Torres R, et al. Concomitant allergic contact dermatitis from diphenhydramine and metronidazole. Contact Dermatitis. 2008;59:115-116. doi:10.1111/j.1600-0536.2008.01332.x
  34. Madsen JT, Lorentzen HF, Paulsen E. Contact sensitization to metronidazole from possible occupational exposure. Contact Dermatitis. 2009;60:117-118. doi:10.1111/j.1600-0536.2008.01490.x
  35. Beutner KR, Lemke S, Calvarese B. A look at the safety of metronidazole 1% gel: cumulative irritation, contact sensitization, phototoxicity, and photoallergy potential. Cutis. 2006;77(4 suppl):12-17.
  36. Jappe U, Schäfer T, Schnuch A, et al. Contact allergy in patients with rosacea: a clinic-based, prospective epidemiological study. J Eur Acad Dermatol Venereol. 2008;22:1208-1214. doi:10.1111/j.1468-3083.2008.02778.x
  37. DeKoven JG, Silverberg JI, Warshaw EM, et al. North American Contact Dermatitis Group Patch Test Results: 2017-2018. Dermatitis. 2021;32:111-123. doi:10.1097/DER.0000000000000729
  38. Comaish JS, Cunliffe WJ. Absorption of drugs from varicose ulcers: a cause of anaphylaxis. Br J Clin Pract. 1967;21:97-98.
  39. Roupe G, Strannegård O. Anaphylactic shock elicited by topical administration of bacitracin. Arch Dermatol. 1969;100:450-452.
  40. Farley M, Pak H, Carregal V, et al. Anaphylaxis to topically applied bacitracin. Am J Contact Dermat. 1995;6:28-31.
  41. Barranco R, Tornero P, de Barrio M, et al. Type IV hypersensitivity to oral nystatin. Contact Dermatitis. 2001;45:60. doi:10.1034/j.1600-0536.2001.045001060.x
  42. Cooper SM, Shaw S. Contact allergy to nystatin: an unusual allergen. Contact Dermatitis. 1999;41:120. doi:10.1111/j.1600-0536.1999.tb06254.x
  43. Dooms-Goossens A, Matura M, Drieghe J, et al. Contact allergy to imidazoles used as antimycotic agents. Contact Dermatitis. 1995;33:73-77. doi:10.1111/j.1600-0536.1995.tb00504.x
  44. Pérez-Mesonero R, Schneller-Pavelescu L, Ochando-Ibernón G, et al. Is tioconazole contact dermatitis still a concern? bringing allergic contact dermatitis caused by topical tioconazole back into the spotlight. Contact Dermatitis. 2019;80:168-169.
  45. Tang MM, Corti MA, Stirnimann R, et al. Severe cutaneous allergic reactions following topical antifungal therapy. Contact Dermatitis. 2013;68:56-57.
  46. Goossens A, Linsen G. Contact allergy to antihistamines is not common. Contact Dermatitis. 1998;39:38. doi:10.1111/j.1600-0536.1998.tb05817.x
  47. Taylor JS, Praditsuwan P, Handel D, et al. Allergic contact dermatitis from doxepin cream. one-year patch test clinic experience. Arch Dermatol. 1996;132:515-518.
  48. Bilbao I, Aguirre A, Vicente JM, et al. Allergic contact dermatitis due to 5% doxepin cream. Contact Dermatitis. 1996;35:254-255. doi:10.1111/j.1600-0536.1996.tb02374.x
  49. Shelley WB, Shelley ED, Talanin NY. Self-potentiating allergic contact dermatitis caused by doxepin hydrochloride cream. J Am Acad Dermatol. 1996;34:143-144. doi:10.1016/s0190-9622(96)90864-6
  50. Wakelin SH, Rycroft RJ. Allergic contact dermatitis from doxepin. Contact Dermatitis. 1999;40:214. doi:10.1111/j.1600-0536.1999.tb06037.x
  51. Horn HM, Tidman MJ, Aldridge RD. Allergic contact dermatitis due to doxepin cream in a patient with dystrophic epidermolysis bullosa. Contact Dermatitis. 2001;45:115. doi:10.1034/j.1600-0536.2001.045002115.x
  52. Bonnel RA, La Grenade L, Karwoski CB, et al. Allergic contact dermatitis from topical doxepin: Food and Drug Administration’s postmarketing surveillance experience. J Am Acad Dermatol. 2003;48:294-296. doi:10.1067/mjd.2003.46
  53. Devleeschouwer V, Roelandts R, Garmyn M, et al. Allergic and photoallergic contact dermatitis from ketoprofen: results of (photo) patch testing and follow-up of 42 patients. Contact Dermatitis. 2008;58:159-166. doi:10.1111/j.1600-0536.2007.01296.x
  54. Foti C, Bonamonte D, Conserva A, et al. Allergic and photoallergic contact dermatitis from ketoprofen: evaluation of cross-reactivities by a combination of photopatch testing and computerized conformational analysis. Curr Pharm Des. 2008;14:2833-2839. doi:10.2174/138161208786369696
  55. Gulin SJ, Chiriac A. Diclofenac-induced allergic contact dermatitis: a series of four patients. Drug Saf Case Rep. 2016;3:15. doi:10.1007/s40800-016-0039-3
  56. Lakshmi C, Srinivas CR. Systemic (allergic) contact dermatitis to diclofenac. Indian J Dermatol Venereol Leprol. 2011;77:536. doi:10.4103/0378-6323.82424
  57. Beutner C, Forkel S, Kreipe K, et al. Contact allergy to topical diclofenac with systemic tolerance [published online August 22, 2021]. Contact Dermatitis. doi:10.1111/cod.13961
  58. Pan Y, Nixon R. Allergic contact dermatitis to topical preparations of bufexamac. Australas J Dermatol. 2012;53:207-210. doi:10.1111/j.1440-0960.2012.00876.x
  59. Nakada T, Matsuzawa Y. Allergic contact dermatitis syndrome from bufexamac for nursing infant. Dermatitis. 2012;23:185-186. doi:10.1097/DER.0b013e318260d774
  60. Kerr AC, Muller F, Ferguson J, et al. Occupational carprofen photoallergic contact dermatitis. Br J Dermatol. 2008;159:1303-1308. doi:10.1111/j.1365-2133.2008.08847.x
  61. Kiely C, Murphy G. Photoallergic contact dermatitis caused by occupational exposure to the canine non-steroidal anti-inflammatory drug carprofen. Contact Dermatitis. 2010;63:364-365. doi:10.1111/j.1600-0536.2010.01820.x
  62. Somberg J, Molnar J. Retrospective evaluation on the analgesic activities of 2 compounded topical creams and voltaren gel in chronic noncancer pain. Am J Ther. 2015;22:342-349. doi:10.1097/MJT.0000000000000275
  63. Lee HG, Grossman SK, Valdes-Rodriguez R, et al. Topical ketamine-amitriptyline-lidocaine for chronic pruritus: a retrospective study assessing efficacy and tolerability. J Am Acad Dermatol. 2017;76:760-761. doi:10.1016/j.jaad.2016.10.030
  64. Turrentine JE, Marrazzo G, Cruz PD Jr. Novel use of patch testing in the first report of allergic contact dermatitis to cyclobenzaprine. Dermatitis. 2015;26:60-61. doi:10.1097/DER.0000000000000099
  65. de Groot A. Patch Testing. 3rd ed. acdegroot publishing; 2008.
  66. de Groot A. Patch Testing. 4th ed. acdegroot publishing; 2018.
Issue
Cutis - 108(5)
Issue
Cutis - 108(5)
Page Number
271-275
Page Number
271-275
Publications
Publications
Topics
Article Type
Display Headline
Contact Allergy to Topical Medicaments, Part 1: A Double-edged Sword
Display Headline
Contact Allergy to Topical Medicaments, Part 1: A Double-edged Sword
Sections
Inside the Article

Practice Points

  • Allergic contact dermatitis should be suspected in patients with persistent or worsening dermatitis after use of topical medications.
  • Prior sensitization is not always apparent, and cross-reactions may occur between structurally similar compounds.
  • Although most medicaments can be patch tested as is, patch testing to the individual components may be necessary to identify the causative allergen.
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
Article PDF Media

Granulomatous Facial Dermatoses

Article Type
Changed
Fri, 02/11/2022 - 13:46
Display Headline
Granulomatous Facial Dermatoses

Cutaneous granulomatous diseases encompass many entities that are skin-limited or systemic. The prototypical cutaneous granuloma is a painless, rounded, well-defined, red-pink or flesh-colored papule1 and is smooth, owing to minimal epidermal involvement. Examples of conditions that present with such lesions include granulomatous periorificial dermatitis (GPD), granulomatous rosacea (GR), lupus miliaris disseminatus faciei (LMDF), and papular sarcoidosis. These entities commonly are seen on the face and can be a source of distress to patients when they are extensive. Several reports have raised the possibility that these conditions lie on a spectrum.2-4 We present 2 cases of patients with facial papular granulomas, discuss potential causes of the lesions, review historical aspects from the literature, and highlight the challenges that these lesions can pose to the clinician.

Case Reports

Patient 1—A 10-year-old Ethiopian girl with a history of atopic dermatitis presented with a facial rash of 4 months’ duration. Her pediatrician initially treated the rash as pityriasis alba and prescribed hydrocortisone cream. Two months into treatment, the patient developed an otherwise asymptomatic, unilateral, papular dermatosis on the right cheek. She subsequently was switched to treatment with benzoyl peroxide and topical clindamycin, which she had been using for 2 months with no improvement at the time of the current presentation. The lesions then spread bilaterally and periorally.

At the current presentation, physical examination demonstrated fine, diffuse, follicular-based, flesh-colored papules over both cheeks, the right side of the nose, and the perioral region (Figure 1). A biopsy of a papular lesion from the right cheek revealed well-formed, noncaseating granulomas in the superficial and mid dermis with an associated lymphocytic infiltrate (Figure 2). No organisms were identified on acid-fast, Fite, or periodic acid–Schiff staining. A tuberculin skin test was negative. A chest radiograph showed small calcified hilar lymph nodes bilaterally. Pulmonary function tests were unremarkable. Calcium and angiotensin-converting enzyme levels were normal.

FIGURE 1. Multiple pink-yellow, smooth, dome-shaped papules on the bilateral cheeks, chin, and nose in patient 1.

The patient denied any fever, chills, hemoptysis, cough, dyspnea, lymphadenopathy, scleral or conjunctival pain or erythema, visual disturbances, or arthralgias. Hydroxychloroquine 200 mg twice daily was started with minimal improvement after 5 months. Methotrexate 20 mg once weekly was then added. Topical fluocinonide 0.05% also was started at this time, as the patient had required several prednisone tapers over the past 3 months for symptomatic relief. The lesions improved minimally after 5 more months of treatment, at which time she had developed inflammatory papules, pustules, and open comedones in the same areas as well as the glabella.

FIGURE 2. Papular lesion in patient 1 prior to treatment. Magnified view of noncaseating granuloma with lymphocytic infiltrate in the superficial dermis (H&E, original magnification ×10).

Repeat biopsy of a papular lesion demonstrated noncaseating granulomas and an associated chronic lymphocytic infiltrate in a follicular and perifollicular distribution (Figure 3). Biopsy of a pustule demonstrated acute Demodex folliculitis. Fluocinonide was stopped, and anti-mite therapy with ivermectin, permethrin cream 5%, and selenium sulfide lotion 2.5% was started, with good response from the pustular lesions.

FIGURE 3. Histologic view of papular lesion in patient 1 after treatment with hydroxychloroquine, methotrexate, and topical fluocinonide. Magnified view of poorly defined granulomas with lymphocytic infiltrates in the mid and superficial dermis (H&E, original magnification ×10).

The patient continued taking methotrexate 20 mg once weekly during this time, with improvement in the papular lesions. She discontinued methotrexate after 12 months with complete resolution. At follow-up 12 months after stopping the methotrexate (roughly 2 years after initial presentation), she showed sustained resolution, with small pitted scars on both cheeks and the nasal tip.

Patient 2—A 33-year-old Ethiopian woman presented with a facial rash of 15 years’ duration. The lesions had been accumulating slowly and were asymptomatic. Physical examination revealed multiple follicular-based, flesh-colored, and erythematous papules on the cheeks, chin, perioral area, and forehead (Figure 4). There were no pustules or telangiectasias. Treatment with tretinoin cream 0.05% for 6 months offered minimal relief.

FIGURE 4. Numerous flesh-colored, dome-shaped papules are seen over parts of the right face in patient 2, including the inferolateral forehead, temple, and cheek, but not the upper eyelid.

 

 

Biopsy of a papule from the left mandible showed superficial vascular telangiectasias, noncaseating granulomas comprising epithelioid histiocytes and lymphocytes in the superficial dermis, and a perifollicular lymphocytic infiltrate (Figure 5). No organisms were identified on Fite or Gomori methenamine silver staining.

FIGURE 5. Histologic view of a papular lesion in patient 2. Magnified view of the superficial dermis demonstrated epithelioid and lymphocytic infiltrates, some of which were trying to form granulomas. Superficial dermal telangiectasias also were present (H&E, original magnification ×10).

Comment

The first step in differentiating cutaneous granulomatous lesions should be to distinguish infectious from noninfectious causes.1 Noninfectious cutaneous granulomas can appear nearly anywhere; however, certain processes have a predilection for the face, including GPD, GR, LMDF, and papular sarcoidosis.5-7 These conditions generally present with papular granulomas with features as described above.

Granulomatous Periorificial Dermatitis—In 1970, Gianotti and colleagues8 briefly described the first possible cases of GPD in 5 children. The eruption comprised numerous yellow, dome-shaped papules in a mostly perioral distribution. Tuberculin and the Kveim tests were nonreactive; histopathology was described as sarcoid-type and not necessarily follicular or perifollicular.8 In 1974, Marten et al9 described 22 Afro-Caribbean children with flesh-colored, papular eruptions on the face that did not show histologic granulomatous changes but were morphologically similar to the reports by Gianotti et al.8 By 1989, Frieden and colleagues10 described this facial eruption as “granulomatous perioral dermatitis in children”. Additionally, the investigators observed granulomatous infiltrates in a perifollicular distribution and suggested follicular disruption as a possible cause. It was clear from the case discussions that these eruptions were not uncommonly diagnosed as papular sarcoidosis.10 The following year, Williams et al11 reported 5 cases of similar papular eruptions in 5 Afro-Caribbean children, coining the term facial Afro-Caribbean eruption.11 Knautz and Lesher12 referred to this entity as “childhood GPD” in 1996 to avoid limiting the diagnosis to Afro-Caribbean patients and to a perioral distribution; this is the most popular current terminology.12 Since then, reports of extrafacial involvement and disease in adults have been published.13,14

Granulomatous periorificial dermatitis often is seen in the perinasal, periocular, and perioral regions of the face.2 It is associated with topical steroid exposure.5 Histologically, noncaseating granulomas around the upper half of undisrupted hair follicles with a lymphocytic infiltrate are typical.13 Treatment should begin with cessation of any topical steroids; first-line agents are oral tetracycline or macrolide antibiotics.5 These agents can be used alone or in combination with topical erythromycin, metronidazole, or sulfur-based lotions.13 Rarely, GPD presents extrafacially.13 Even so, it usually resolves within 2 weeks to 6 months, especially with therapy; scarring is unusual.5,13,15

Granulomatous Rosacea—A report in the early 20th century described patients with tuberculoid granulomas resembling papular rosacea; the initial belief was that this finding represented a rosacealike tuberculid eruption.5 However, this belief was questioned by Snapp,16 among others, who demonstrated near universal lack of reactivity to tuberculin among 20 of these patients in 1949; more recent evidence has substantiated these findings.17 Still, Snapp16 postulated that these rosacealike granulomatous lesions were distinct from classic rosacea because they lacked vascular symptoms and pustules and were recalcitrant to rosacea treatment modalities.

 

 

In 1970, Mullanax and colleagues18 introduced the term granulomatous rosacea, reiterating that this entity was not tuberculous. They documented papulopustular lesions as well as telangiectasias, raising the possibility that GR does overlap with acne rosacea. More recent studies have established the current theory that GR is a histologic variant of acne rosacea because, in addition to typical granulomatous papules, its microscopic features can be seen across subtypes of acne rosacea.19,20

Various causes have been proposed for GR. Demodex mites have been reported in association with GR for nearly 30 years.19,20 In the past 10 years, molecular studies have started to define the role of metalloproteinases, UV radiation, and cutaneous peptides in the pathogenesis of acne rosacea and GR.21,22

Granulomatous rosacea typically is seen in middle-aged women.20,23 Hallmarks of rosacea, such as facial erythema, flushing, telangiectasias, pustules, and rhinophyma, are not always present in GR.5,20,23 Lesions usually are distributed around the central face, although extension to the cheeks, total facial involvement, and extrafacial lesions are possible.5,20 Histologically, perifollicular and follicular-based noncaseating granulomas with dilatation of the dermal papillary vasculature are seen.17,23 As a whole, rosacea is comparatively uncommon in dark-skinned patients; when it does occur, GR is a frequent presentation.24

First-line treatment for GR is tetracycline antibiotics.5 Unresponsive cases have been treated—largely anecdotally—with topical modalities (eg, metronidazole, steroids, immunomodulators), systemic agents (eg, dapsone, erythromycin, isotretinoin), and other therapies.5 Granulomatous rosacea tends to have a chronic course.5,23

Lupus Miliaris Disseminatus Faciei—Classic LMDF demonstrates caseating perifollicular granulomas histologically.6,17,25 Lesions tend to appear on the central face, particularly the eyelids, and can be seen extrafacially.3,6,25,26 Although LMDF originally was categorized as a tuberculid eruption, this no longer is thought to be the case.27 It is now regarded by some as a variant of GR25; however, LMDF responds poorly to tetracyclines, is more common in males, and lacks rosacealike vascular abnormalities, leading some to question this association.3,6,17 In the past 20 years, some have proposed renaming LMDF to better reflect its clinical course and to consider it independent of tuberculosis and GR.28 It usually resolves spontaneously after 1 to 3 years, leaving pitted scars.3,6

 

 

Papular Sarcoidosis—The first potential documented case of sarcoidosis was by Hutchinson29 in 1869 in a patient seen in London. The author labeled purple plaques on the index patient’s legs and hands as “livid papillary psoriasis.” In 1889, Besnier30 described a patient with violaceous swellings on the nose, ears, and fingers, which he called “lupus pernio”; his contemporary, Tenneson,31 published a case of lupus pernio and described its histologic profile as comprising epithelioid cells and giant cells. It was not until 1899 that the term sarkoid was used to describe these cutaneous lesions by Boeck,32 who thought they were reminiscent of sarcoma. In 1915, Kuznitsky and Bittorf33 described a patient with cutaneous lesions histologically consistent with Boeck’s sarkoid but additionally with hilar lymphadenopathy and pulmonary infiltrates. Around 1916 or 1917, Schaumann34 described patients with cutaneous lesions and additionally with involvement of pulmonary, osseous, hepatosplenic, and tonsillar tissue. These reports are among the first to recognize the multisystemic nature of sarcoidosis. The first possible case of childhood sarcoidosis might have been reported by Osler35 in the United States in 1898.

In the past century or so, an ongoing effort by researchers has focused on identifying etiologic triggers for sarcoidosis. Microbial agents have been considered in this role, with Mycobacterium and Propionibacterium organisms the most intensively studied; the possibility that foreign material contributes to the formation of granulomas also has been raised.36 Current models of the pathogenesis of sarcoidosis involve an interplay between the immune system in genetically predisposed patients and an infection that leads to a hyperimmune type 1 T–helper cell response that clears the infection but not antigens generated by the microbes and the acute host response, including proteins such as serum amyloid A and vimentin.36,37 These antigens aggregate and serve as a nidus for granuloma formation and maintenance long after infection has resolved.

Cutaneous lesions of sarcoidosis include macules, papules, plaques, and lupus pernio, as well as lesions arising within scars or tattoos, with many less common presentations.7,38 Papular sarcoidosis is common on the face but also can involve the extremities.4,7 Strictly, at least 2 organ systems must be involved to diagnose sarcoidosis, but this is debatable.4,7 Among 41 patients with cutaneous sarcoidosis, 24 (58.5%) had systemic disease; cutaneous lesions were the presenting sign in 87.5% (21/24) of patients.38 Histologic analysis, regardless of the lesion, usually shows noncaseating so-called “naked” granulomas, which have minimal lymphocytic infiltrate associated with the epithelioid histiocytes.38,39 Perifollicular granulomas are possible but unusual.40

Treatment depends on the extent of cutaneous and systemic involvement. Pharmacotherapeutic modalities include topical steroids, immunomodulators, and retinoids; systemic immunomodulators and immunosuppressants; and biologic agents.7 Isolated cutaneous sarcoidosis, particularly the papular variant, usually is associated with acute disease lasting less than 2 years, with resolution of skin lesions.7,38 That said, a recent report suggested that cutaneous sarcoidosis can progress to multisystemic disease as long as 7 years after the initial diagnosis.41

Clinical and Histologic Overlap—Despite this categorization of noninfectious facial granulomatous conditions, each has some clinical and histologic overlap with the others, which must be considered when encountering a granulomatous facial dermatosis. Both GPD and GR tend to present with lesions near the eyes, mouth, and nose, although GR can extend to lateral aspects of the face, below the mandible, and the forehead and has different demographic features.15,20,23 Granulomas in both GPD and GR generally are noncaseating and form in a follicular or perifollicular distribution within the dermis.2,15,23 Lupus miliaris disseminatus faciei and GR share a similar facial distribution in some cases.17,20 Even papular cutaneous sarcoidosis has masqueraded as GR clinically and histologically.4

 

 

Diagnostic and Treatment Difficulty—Our cases illustrate the range of difficulty in evaluating and managing patients with facial papular granulomas. On one hand, our adult patient’s clinical and histologic findings were highly consistent with GR; on the other hand, our younger patient had clinicopathologic features of both sarcoidosis and GPD at varying times. Both conditions are more common in dark-skinned patients.11,42

Juvenile sarcoidosis is comparatively rare, with a reported annual incidence of 0.22 to 0.27 for every 100,000 children younger than 15 years; however, juvenile sarcoidosis commonly presents around 8 to 15 years of age.43

It is unusual for sarcoid granulomas to be isolated to the skin, much less to the face.4,7,43,44 Patient 1 initially presented in this manner and lacked convincing laboratory or radiographic evidence of systemic sarcoidosis. Bilateral hilar calcifications in sarcoidosis are more typical among adults after 5 to 20 years; there were no signs or symptoms of active infection that could account for the pulmonary and cutaneous lesions.45

The presence of perifollicular granulomas with associated lymphocytic infiltrates on repeat biopsy, coupled with the use of topical steroids, made it difficult to rule out a contribution by GPD to her clinical course. That her lesions resolved with pitted scarring while she was taking methotrexate and after topical steroids had been stopped could be the result of successful management or spontaneous resolution of her dermatosis; both papular sarcoidosis and GPD tend to have a self-limited course.7,13

Conclusion

We present 2 cases of papular facial granulomas in patients with similar skin types who had different clinical courses. Evaluation of such lesions remains challenging given the similarity between specific entities that present in this manner. Certainly, it is reasonable to consider a spectrum upon which all of these conditions fall, in light of the findings of these cases and those reported previously.

References
  1. Beretta-Piccoli BT, Mainetti C, Peeters M-A, et al. Cutaneous granulomatosis: a comprehensive review. Clin Rev Allergy Immunol. 2018;54:131-146. doi:10.1007/s12016-017-8666-8
  2. Lucas CR, Korman NJ, Gilliam AC. Granulomatous periorificial dermatitis: a variant of granulomatous rosacea in children? J Cutan Med Surg. 2009;13:115-118. doi:10.2310/7750.2008.07088
  3. van de Scheur MR, van der Waal RIF, Starink TM. Lupus miliaris disseminatus faciei: a distinctive rosacea-like syndrome and not a granulomatous form of rosacea. Dermatology. 2003;206:120-123. doi:10.1159/000068457
  4. Simonart T, Lowy M, Rasquin F, et al. Overlap of sarcoidosis and rosacea. Dermatology. 1997;194:416-418. doi:10.1159/000246165
  5. Lee GL, Zirwas MJ. Granulomatous rosacea and periorificial dermatitis: controversies and review of management. Dermatol Clin. 2015;33:447-455. doi:10.1016/j.det.2015.03.009
  6. Michaels JD, Cook-Norris RH, Lehman JS, et al. Adult with papular eruption of the central aspect of the face. J Am Acad Dermatol. 2014;71:410-412. doi:10.1016/j.jaad.2012.06.039
  7. Wanat KA, Rosenbach M. Cutaneous sarcoidosis. Clin Chest Med. 2015;38:685-702. doi:10.1016/j.ccm.2015.08.010
  8. Gianotti F, Ermacora E, Benelli MG, et al. Particulière dermatite peri-orale infantile. observations sur 5 cas. Bull Soc Fr Dermatol Syphiligr. 1970;77:341.
  9. Marten RH, Presbury DG, Adamson JE, et al. An unusual papular and acneiform facial eruption in the negro child. Br J Dermatol. 1974;91:435-438. doi:10.1111/j.1365-2133.1974.tb13083.x
  10. Frieden IJ, Prose NS, Fletcher V, et al. Granulomatous perioral dermatitis in children. Arch Dermatol. 1989;125:369-373.
  11. Williams HC, Ashworth J, Pembroke AC, et al. FACE—facial Afro-Caribbean childhood eruption. Clin Exp Dermatol. 1990;15:163-166. doi:10.1111/j.1365-2230.1990.tb02063.x
  12. Knautz MA, Lesher JL Jr. Childhood granulomatous periorificial dermatitis. Pediatr Dermatol. 1996;13:131-134. doi:10.1111/j.1525-1470.1996.tb01419.x
  13. Urbatsch AJ, Frieden I, Williams ML, et al. Extrafacial and generalized granulomatous periorificial dermatitis. Arch Dermatol. 2002;138:1354-1358. doi:10.1001/archderm.138.10.1354
  14. Vincenzi C, Parente G, Tosti A. Perioral granulomatous dermatitis: two cases treated with clarithromycin. J Dermatol Treat. 2000;11:57-61.
  15. Kim YJ, Shin JW, Lee JS, et al. Childhood granulomatous periorificial dermatitis. Ann Dermatol. 2011;23:386-388. doi:10.5021/ad.2011.23.3.386
  16. Snapp RH. Lewandowsky’s rosacea-like eruption; a clinical study. J Invest Dermatol. 1949;13:175-190. doi:10.1038/jid.1949.86
  17. Chougule A, Chatterjee D, Sethi S, et al. Granulomatous rosacea versus lupus miliaris disseminatus faciei—2 faces of facial granulomatous disorder: a clinicohistological and molecular study. Am J Dermatopathol. 2018;40:819-823. doi:10.1097/DAD.0000000000001243
  18. Mullanax MG, Kierland RR. Granulomatous rosacea. Arch Dermatol. 1970;101:206-211.
  19. Sánchez JL, Berlingeri-Ramos AC, Dueño DV. Granulomatous rosacea. Am J Dermatopathol. 2008;30:6-9. doi:10.1097/DAD.0b013e31815bc191
  20. Helm KF, Menz J, Gibson LE, et al. A clinical and histopathologic study of granulomatous rosacea. J Am Acad Dermatol. 1991;25:1038-1043. doi:10.1016/0190-9622(91)70304-k
  21. Kanada KN, Nakatsuji T, Gallo RL. Doxycycline indirectly inhibits proteolytic activation of tryptic kallikrein-related peptidases and activation of cathelicidin. J Invest Dermatol. 2012;132:1435-1442. doi:10.1038/jid.2012.14
  22. Jang YH, Sim JH, Kang HY, et al. Immunohistochemical expression of matrix metalloproteinases in the granulomatous rosacea compared with the non-granulomatous rosacea. J Eur Acad Dermatol Venereol. 2011;25:544-548. doi:10.1111/j.1468-3083.2010.03825.x
  23. Khokhar O, Khachemoune A. A case of granulomatous rosacea: sorting granulomatous rosacea from other granulomatous diseases that affect the face. Dermatol Online J. 2004;10:6.
  24. Rosen T, Stone MS. Acne rosacea in blacks. J Am Acad Dermatol. 1987;17:70-73. doi:10.1016/s0190-9622(87)70173-x
  25. Adams AK, Davis JL, Davis MDP, et al. What is your diagnosis? granulomatous rosacea (lupus miliaris disseminatus faciei, acne agminata). Cutis. 2008;82:103-112.
  26. Shitara A. Lupus miliaris disseminatus faciei. Int J Dermatol. 1984;23:542-544. doi:10.1111/j.1365-4362.1984.tb04206.x
  27. Hodak E, Trattner A, Feuerman H, et al. Lupus miliaris disseminatus faciei—the DNA of Mycobacterium tuberculosis is not detectable in active lesions by polymerase chain reaction. Br J Dermatol. 1997;137:614-619. doi: 10.1111/j.1365-2133.1997.tb03797.x
  28. Skowron F, Causeret AS, Pabion C, et al. F.I.GU.R.E.: facial idiopathic granulomas with regressive evolution. Dermatology. 2000;201:287-289. doi:10.1159/000051539
  29. Hutchinson J. Case of livid papillary psoriasis. In: London J, Churchill A, eds. Illustrations of Clinical Surgery. J&A Churchill; 1877:42-43.
  30. Besnier E. Lupus pernio of the face [in French]. Ann Dermatol Syphiligr (Paris). 1889;10:33-36.
  31. Tenneson H. Lupus pernio. Ann Dermatol Syphiligr (Paris). 1889;10:333-336.
  32. Boeck C. Multiple benign sarkoid of the skin [in Norwegian]. Norsk Mag Laegevidensk. 1899;14:1321-1334.
  33. Kuznitsky E, Bittorf A. Sarkoid mit beteiligung innerer organe. Münch Med Wochenschr. 1915;62:1349-1353.
  34. Schaumann J. Etude sur le lupus pernio et ses rapports avec les sarcoides et la tuberculose. Ann Dermatol Syphiligr. 1916-1917;6:357-373.
  35. Osler W. On chronic symmetrical enlargement of the salivary and lacrimal glands. Am J Med Sci. 1898;115:27-30.
  36. Chen ES, Moller DR. Etiologies of sarcoidosis. Clin Rev Allergy Immunol. 2015;49:6-18. doi:10.1007/s12016-015-8481-z
  37. Eberhardt C, Thillai M, Parker R, et al. Proteomic analysis of Kveim reagent identifies targets of cellular immunity in sarcoidosis. PLoS One. 2017;12:e0170285. doi:10.1371/journal.pone.0170285
  38. Esteves TC, Aparicio G, Ferrer B, et al. Prognostic value of skin lesions in sarcoidosis: clinical and histopathological clues. Eur J Dermatol. 2015;25:556-562. doi:10.1684/ejd.2015.2666
  39. Cardoso JC, Cravo M, Reis JP, et al. Cutaneous sarcoidosis: a histopathological study. J Eur Acad Dermatol Venereol. 2009;23:678-682. doi:10.1111/j.1468-3083.2009.03153.x
  40. Mangas C, Fernández-Figueras M-T, Fité E, et al. Clinical spectrum and histological analysis of 32 cases of specific cutaneous sarcoidosis. J Cutan Pathol. 2006;33:772-777. doi:10.1111/j.1600-0560.2006.00563.x
  41. García-Colmenero L, Sánchez-Schmidt JM, Barranco C, et al. The natural history of cutaneous sarcoidosis. clinical spectrum and histological analysis of 40 cases. Int J Dermatol. 2019;58:178-184. doi: 10.1111/ijd.14218
  42. Shetty AK, Gedalia A. Childhood sarcoidosis: a rare but fascinating disorder. Pediatr Rheumatol Online J. 2008;6:16. doi:10.1186/1546-0096-6-16
  43. Milman N, Hoffmann AL, Byg KE. Sarcoidosis in children. epidemiology in Danes, clinical features, diagnosis, treatment and prognosis. Acta Paediatr. 1998;87:871-878. doi:10.1080/08035259875001366244. S¸ims¸ek A, Çelikten H, Yapıcı I. Isolated cutaneous sarcoidosis. Arch Bronconeumol. 2016;52:220.
  44. Scadding JG. The late stages of pulmonary sarcoidosis. Postgrad Med J. 1970;46:530-536. doi:10.1136/pgmj.46.538.530
Article PDF
Author and Disclosure Information

Dr. Teran is from the Department of Dermatology, The University of Texas Medical Branch, Galveston. Drs. Belote, Cropley, Gru, and Zlotoff are from the Department of Dermatology, University of Virginia Health System, Charlottesville. Dr. Gru also is from the Department of Pathology.

The authors report no conflict of interest.

Correspondence: Victor A. Teran, MD (vateran@utmb.edu).

Issue
Cutis - 108(4)
Publications
Topics
Page Number
E5-E10
Sections
Author and Disclosure Information

Dr. Teran is from the Department of Dermatology, The University of Texas Medical Branch, Galveston. Drs. Belote, Cropley, Gru, and Zlotoff are from the Department of Dermatology, University of Virginia Health System, Charlottesville. Dr. Gru also is from the Department of Pathology.

The authors report no conflict of interest.

Correspondence: Victor A. Teran, MD (vateran@utmb.edu).

Author and Disclosure Information

Dr. Teran is from the Department of Dermatology, The University of Texas Medical Branch, Galveston. Drs. Belote, Cropley, Gru, and Zlotoff are from the Department of Dermatology, University of Virginia Health System, Charlottesville. Dr. Gru also is from the Department of Pathology.

The authors report no conflict of interest.

Correspondence: Victor A. Teran, MD (vateran@utmb.edu).

Article PDF
Article PDF

Cutaneous granulomatous diseases encompass many entities that are skin-limited or systemic. The prototypical cutaneous granuloma is a painless, rounded, well-defined, red-pink or flesh-colored papule1 and is smooth, owing to minimal epidermal involvement. Examples of conditions that present with such lesions include granulomatous periorificial dermatitis (GPD), granulomatous rosacea (GR), lupus miliaris disseminatus faciei (LMDF), and papular sarcoidosis. These entities commonly are seen on the face and can be a source of distress to patients when they are extensive. Several reports have raised the possibility that these conditions lie on a spectrum.2-4 We present 2 cases of patients with facial papular granulomas, discuss potential causes of the lesions, review historical aspects from the literature, and highlight the challenges that these lesions can pose to the clinician.

Case Reports

Patient 1—A 10-year-old Ethiopian girl with a history of atopic dermatitis presented with a facial rash of 4 months’ duration. Her pediatrician initially treated the rash as pityriasis alba and prescribed hydrocortisone cream. Two months into treatment, the patient developed an otherwise asymptomatic, unilateral, papular dermatosis on the right cheek. She subsequently was switched to treatment with benzoyl peroxide and topical clindamycin, which she had been using for 2 months with no improvement at the time of the current presentation. The lesions then spread bilaterally and periorally.

At the current presentation, physical examination demonstrated fine, diffuse, follicular-based, flesh-colored papules over both cheeks, the right side of the nose, and the perioral region (Figure 1). A biopsy of a papular lesion from the right cheek revealed well-formed, noncaseating granulomas in the superficial and mid dermis with an associated lymphocytic infiltrate (Figure 2). No organisms were identified on acid-fast, Fite, or periodic acid–Schiff staining. A tuberculin skin test was negative. A chest radiograph showed small calcified hilar lymph nodes bilaterally. Pulmonary function tests were unremarkable. Calcium and angiotensin-converting enzyme levels were normal.

FIGURE 1. Multiple pink-yellow, smooth, dome-shaped papules on the bilateral cheeks, chin, and nose in patient 1.

The patient denied any fever, chills, hemoptysis, cough, dyspnea, lymphadenopathy, scleral or conjunctival pain or erythema, visual disturbances, or arthralgias. Hydroxychloroquine 200 mg twice daily was started with minimal improvement after 5 months. Methotrexate 20 mg once weekly was then added. Topical fluocinonide 0.05% also was started at this time, as the patient had required several prednisone tapers over the past 3 months for symptomatic relief. The lesions improved minimally after 5 more months of treatment, at which time she had developed inflammatory papules, pustules, and open comedones in the same areas as well as the glabella.

FIGURE 2. Papular lesion in patient 1 prior to treatment. Magnified view of noncaseating granuloma with lymphocytic infiltrate in the superficial dermis (H&E, original magnification ×10).

Repeat biopsy of a papular lesion demonstrated noncaseating granulomas and an associated chronic lymphocytic infiltrate in a follicular and perifollicular distribution (Figure 3). Biopsy of a pustule demonstrated acute Demodex folliculitis. Fluocinonide was stopped, and anti-mite therapy with ivermectin, permethrin cream 5%, and selenium sulfide lotion 2.5% was started, with good response from the pustular lesions.

FIGURE 3. Histologic view of papular lesion in patient 1 after treatment with hydroxychloroquine, methotrexate, and topical fluocinonide. Magnified view of poorly defined granulomas with lymphocytic infiltrates in the mid and superficial dermis (H&E, original magnification ×10).

The patient continued taking methotrexate 20 mg once weekly during this time, with improvement in the papular lesions. She discontinued methotrexate after 12 months with complete resolution. At follow-up 12 months after stopping the methotrexate (roughly 2 years after initial presentation), she showed sustained resolution, with small pitted scars on both cheeks and the nasal tip.

Patient 2—A 33-year-old Ethiopian woman presented with a facial rash of 15 years’ duration. The lesions had been accumulating slowly and were asymptomatic. Physical examination revealed multiple follicular-based, flesh-colored, and erythematous papules on the cheeks, chin, perioral area, and forehead (Figure 4). There were no pustules or telangiectasias. Treatment with tretinoin cream 0.05% for 6 months offered minimal relief.

FIGURE 4. Numerous flesh-colored, dome-shaped papules are seen over parts of the right face in patient 2, including the inferolateral forehead, temple, and cheek, but not the upper eyelid.

 

 

Biopsy of a papule from the left mandible showed superficial vascular telangiectasias, noncaseating granulomas comprising epithelioid histiocytes and lymphocytes in the superficial dermis, and a perifollicular lymphocytic infiltrate (Figure 5). No organisms were identified on Fite or Gomori methenamine silver staining.

FIGURE 5. Histologic view of a papular lesion in patient 2. Magnified view of the superficial dermis demonstrated epithelioid and lymphocytic infiltrates, some of which were trying to form granulomas. Superficial dermal telangiectasias also were present (H&E, original magnification ×10).

Comment

The first step in differentiating cutaneous granulomatous lesions should be to distinguish infectious from noninfectious causes.1 Noninfectious cutaneous granulomas can appear nearly anywhere; however, certain processes have a predilection for the face, including GPD, GR, LMDF, and papular sarcoidosis.5-7 These conditions generally present with papular granulomas with features as described above.

Granulomatous Periorificial Dermatitis—In 1970, Gianotti and colleagues8 briefly described the first possible cases of GPD in 5 children. The eruption comprised numerous yellow, dome-shaped papules in a mostly perioral distribution. Tuberculin and the Kveim tests were nonreactive; histopathology was described as sarcoid-type and not necessarily follicular or perifollicular.8 In 1974, Marten et al9 described 22 Afro-Caribbean children with flesh-colored, papular eruptions on the face that did not show histologic granulomatous changes but were morphologically similar to the reports by Gianotti et al.8 By 1989, Frieden and colleagues10 described this facial eruption as “granulomatous perioral dermatitis in children”. Additionally, the investigators observed granulomatous infiltrates in a perifollicular distribution and suggested follicular disruption as a possible cause. It was clear from the case discussions that these eruptions were not uncommonly diagnosed as papular sarcoidosis.10 The following year, Williams et al11 reported 5 cases of similar papular eruptions in 5 Afro-Caribbean children, coining the term facial Afro-Caribbean eruption.11 Knautz and Lesher12 referred to this entity as “childhood GPD” in 1996 to avoid limiting the diagnosis to Afro-Caribbean patients and to a perioral distribution; this is the most popular current terminology.12 Since then, reports of extrafacial involvement and disease in adults have been published.13,14

Granulomatous periorificial dermatitis often is seen in the perinasal, periocular, and perioral regions of the face.2 It is associated with topical steroid exposure.5 Histologically, noncaseating granulomas around the upper half of undisrupted hair follicles with a lymphocytic infiltrate are typical.13 Treatment should begin with cessation of any topical steroids; first-line agents are oral tetracycline or macrolide antibiotics.5 These agents can be used alone or in combination with topical erythromycin, metronidazole, or sulfur-based lotions.13 Rarely, GPD presents extrafacially.13 Even so, it usually resolves within 2 weeks to 6 months, especially with therapy; scarring is unusual.5,13,15

Granulomatous Rosacea—A report in the early 20th century described patients with tuberculoid granulomas resembling papular rosacea; the initial belief was that this finding represented a rosacealike tuberculid eruption.5 However, this belief was questioned by Snapp,16 among others, who demonstrated near universal lack of reactivity to tuberculin among 20 of these patients in 1949; more recent evidence has substantiated these findings.17 Still, Snapp16 postulated that these rosacealike granulomatous lesions were distinct from classic rosacea because they lacked vascular symptoms and pustules and were recalcitrant to rosacea treatment modalities.

 

 

In 1970, Mullanax and colleagues18 introduced the term granulomatous rosacea, reiterating that this entity was not tuberculous. They documented papulopustular lesions as well as telangiectasias, raising the possibility that GR does overlap with acne rosacea. More recent studies have established the current theory that GR is a histologic variant of acne rosacea because, in addition to typical granulomatous papules, its microscopic features can be seen across subtypes of acne rosacea.19,20

Various causes have been proposed for GR. Demodex mites have been reported in association with GR for nearly 30 years.19,20 In the past 10 years, molecular studies have started to define the role of metalloproteinases, UV radiation, and cutaneous peptides in the pathogenesis of acne rosacea and GR.21,22

Granulomatous rosacea typically is seen in middle-aged women.20,23 Hallmarks of rosacea, such as facial erythema, flushing, telangiectasias, pustules, and rhinophyma, are not always present in GR.5,20,23 Lesions usually are distributed around the central face, although extension to the cheeks, total facial involvement, and extrafacial lesions are possible.5,20 Histologically, perifollicular and follicular-based noncaseating granulomas with dilatation of the dermal papillary vasculature are seen.17,23 As a whole, rosacea is comparatively uncommon in dark-skinned patients; when it does occur, GR is a frequent presentation.24

First-line treatment for GR is tetracycline antibiotics.5 Unresponsive cases have been treated—largely anecdotally—with topical modalities (eg, metronidazole, steroids, immunomodulators), systemic agents (eg, dapsone, erythromycin, isotretinoin), and other therapies.5 Granulomatous rosacea tends to have a chronic course.5,23

Lupus Miliaris Disseminatus Faciei—Classic LMDF demonstrates caseating perifollicular granulomas histologically.6,17,25 Lesions tend to appear on the central face, particularly the eyelids, and can be seen extrafacially.3,6,25,26 Although LMDF originally was categorized as a tuberculid eruption, this no longer is thought to be the case.27 It is now regarded by some as a variant of GR25; however, LMDF responds poorly to tetracyclines, is more common in males, and lacks rosacealike vascular abnormalities, leading some to question this association.3,6,17 In the past 20 years, some have proposed renaming LMDF to better reflect its clinical course and to consider it independent of tuberculosis and GR.28 It usually resolves spontaneously after 1 to 3 years, leaving pitted scars.3,6

 

 

Papular Sarcoidosis—The first potential documented case of sarcoidosis was by Hutchinson29 in 1869 in a patient seen in London. The author labeled purple plaques on the index patient’s legs and hands as “livid papillary psoriasis.” In 1889, Besnier30 described a patient with violaceous swellings on the nose, ears, and fingers, which he called “lupus pernio”; his contemporary, Tenneson,31 published a case of lupus pernio and described its histologic profile as comprising epithelioid cells and giant cells. It was not until 1899 that the term sarkoid was used to describe these cutaneous lesions by Boeck,32 who thought they were reminiscent of sarcoma. In 1915, Kuznitsky and Bittorf33 described a patient with cutaneous lesions histologically consistent with Boeck’s sarkoid but additionally with hilar lymphadenopathy and pulmonary infiltrates. Around 1916 or 1917, Schaumann34 described patients with cutaneous lesions and additionally with involvement of pulmonary, osseous, hepatosplenic, and tonsillar tissue. These reports are among the first to recognize the multisystemic nature of sarcoidosis. The first possible case of childhood sarcoidosis might have been reported by Osler35 in the United States in 1898.

In the past century or so, an ongoing effort by researchers has focused on identifying etiologic triggers for sarcoidosis. Microbial agents have been considered in this role, with Mycobacterium and Propionibacterium organisms the most intensively studied; the possibility that foreign material contributes to the formation of granulomas also has been raised.36 Current models of the pathogenesis of sarcoidosis involve an interplay between the immune system in genetically predisposed patients and an infection that leads to a hyperimmune type 1 T–helper cell response that clears the infection but not antigens generated by the microbes and the acute host response, including proteins such as serum amyloid A and vimentin.36,37 These antigens aggregate and serve as a nidus for granuloma formation and maintenance long after infection has resolved.

Cutaneous lesions of sarcoidosis include macules, papules, plaques, and lupus pernio, as well as lesions arising within scars or tattoos, with many less common presentations.7,38 Papular sarcoidosis is common on the face but also can involve the extremities.4,7 Strictly, at least 2 organ systems must be involved to diagnose sarcoidosis, but this is debatable.4,7 Among 41 patients with cutaneous sarcoidosis, 24 (58.5%) had systemic disease; cutaneous lesions were the presenting sign in 87.5% (21/24) of patients.38 Histologic analysis, regardless of the lesion, usually shows noncaseating so-called “naked” granulomas, which have minimal lymphocytic infiltrate associated with the epithelioid histiocytes.38,39 Perifollicular granulomas are possible but unusual.40

Treatment depends on the extent of cutaneous and systemic involvement. Pharmacotherapeutic modalities include topical steroids, immunomodulators, and retinoids; systemic immunomodulators and immunosuppressants; and biologic agents.7 Isolated cutaneous sarcoidosis, particularly the papular variant, usually is associated with acute disease lasting less than 2 years, with resolution of skin lesions.7,38 That said, a recent report suggested that cutaneous sarcoidosis can progress to multisystemic disease as long as 7 years after the initial diagnosis.41

Clinical and Histologic Overlap—Despite this categorization of noninfectious facial granulomatous conditions, each has some clinical and histologic overlap with the others, which must be considered when encountering a granulomatous facial dermatosis. Both GPD and GR tend to present with lesions near the eyes, mouth, and nose, although GR can extend to lateral aspects of the face, below the mandible, and the forehead and has different demographic features.15,20,23 Granulomas in both GPD and GR generally are noncaseating and form in a follicular or perifollicular distribution within the dermis.2,15,23 Lupus miliaris disseminatus faciei and GR share a similar facial distribution in some cases.17,20 Even papular cutaneous sarcoidosis has masqueraded as GR clinically and histologically.4

 

 

Diagnostic and Treatment Difficulty—Our cases illustrate the range of difficulty in evaluating and managing patients with facial papular granulomas. On one hand, our adult patient’s clinical and histologic findings were highly consistent with GR; on the other hand, our younger patient had clinicopathologic features of both sarcoidosis and GPD at varying times. Both conditions are more common in dark-skinned patients.11,42

Juvenile sarcoidosis is comparatively rare, with a reported annual incidence of 0.22 to 0.27 for every 100,000 children younger than 15 years; however, juvenile sarcoidosis commonly presents around 8 to 15 years of age.43

It is unusual for sarcoid granulomas to be isolated to the skin, much less to the face.4,7,43,44 Patient 1 initially presented in this manner and lacked convincing laboratory or radiographic evidence of systemic sarcoidosis. Bilateral hilar calcifications in sarcoidosis are more typical among adults after 5 to 20 years; there were no signs or symptoms of active infection that could account for the pulmonary and cutaneous lesions.45

The presence of perifollicular granulomas with associated lymphocytic infiltrates on repeat biopsy, coupled with the use of topical steroids, made it difficult to rule out a contribution by GPD to her clinical course. That her lesions resolved with pitted scarring while she was taking methotrexate and after topical steroids had been stopped could be the result of successful management or spontaneous resolution of her dermatosis; both papular sarcoidosis and GPD tend to have a self-limited course.7,13

Conclusion

We present 2 cases of papular facial granulomas in patients with similar skin types who had different clinical courses. Evaluation of such lesions remains challenging given the similarity between specific entities that present in this manner. Certainly, it is reasonable to consider a spectrum upon which all of these conditions fall, in light of the findings of these cases and those reported previously.

Cutaneous granulomatous diseases encompass many entities that are skin-limited or systemic. The prototypical cutaneous granuloma is a painless, rounded, well-defined, red-pink or flesh-colored papule1 and is smooth, owing to minimal epidermal involvement. Examples of conditions that present with such lesions include granulomatous periorificial dermatitis (GPD), granulomatous rosacea (GR), lupus miliaris disseminatus faciei (LMDF), and papular sarcoidosis. These entities commonly are seen on the face and can be a source of distress to patients when they are extensive. Several reports have raised the possibility that these conditions lie on a spectrum.2-4 We present 2 cases of patients with facial papular granulomas, discuss potential causes of the lesions, review historical aspects from the literature, and highlight the challenges that these lesions can pose to the clinician.

Case Reports

Patient 1—A 10-year-old Ethiopian girl with a history of atopic dermatitis presented with a facial rash of 4 months’ duration. Her pediatrician initially treated the rash as pityriasis alba and prescribed hydrocortisone cream. Two months into treatment, the patient developed an otherwise asymptomatic, unilateral, papular dermatosis on the right cheek. She subsequently was switched to treatment with benzoyl peroxide and topical clindamycin, which she had been using for 2 months with no improvement at the time of the current presentation. The lesions then spread bilaterally and periorally.

At the current presentation, physical examination demonstrated fine, diffuse, follicular-based, flesh-colored papules over both cheeks, the right side of the nose, and the perioral region (Figure 1). A biopsy of a papular lesion from the right cheek revealed well-formed, noncaseating granulomas in the superficial and mid dermis with an associated lymphocytic infiltrate (Figure 2). No organisms were identified on acid-fast, Fite, or periodic acid–Schiff staining. A tuberculin skin test was negative. A chest radiograph showed small calcified hilar lymph nodes bilaterally. Pulmonary function tests were unremarkable. Calcium and angiotensin-converting enzyme levels were normal.

FIGURE 1. Multiple pink-yellow, smooth, dome-shaped papules on the bilateral cheeks, chin, and nose in patient 1.

The patient denied any fever, chills, hemoptysis, cough, dyspnea, lymphadenopathy, scleral or conjunctival pain or erythema, visual disturbances, or arthralgias. Hydroxychloroquine 200 mg twice daily was started with minimal improvement after 5 months. Methotrexate 20 mg once weekly was then added. Topical fluocinonide 0.05% also was started at this time, as the patient had required several prednisone tapers over the past 3 months for symptomatic relief. The lesions improved minimally after 5 more months of treatment, at which time she had developed inflammatory papules, pustules, and open comedones in the same areas as well as the glabella.

FIGURE 2. Papular lesion in patient 1 prior to treatment. Magnified view of noncaseating granuloma with lymphocytic infiltrate in the superficial dermis (H&E, original magnification ×10).

Repeat biopsy of a papular lesion demonstrated noncaseating granulomas and an associated chronic lymphocytic infiltrate in a follicular and perifollicular distribution (Figure 3). Biopsy of a pustule demonstrated acute Demodex folliculitis. Fluocinonide was stopped, and anti-mite therapy with ivermectin, permethrin cream 5%, and selenium sulfide lotion 2.5% was started, with good response from the pustular lesions.

FIGURE 3. Histologic view of papular lesion in patient 1 after treatment with hydroxychloroquine, methotrexate, and topical fluocinonide. Magnified view of poorly defined granulomas with lymphocytic infiltrates in the mid and superficial dermis (H&E, original magnification ×10).

The patient continued taking methotrexate 20 mg once weekly during this time, with improvement in the papular lesions. She discontinued methotrexate after 12 months with complete resolution. At follow-up 12 months after stopping the methotrexate (roughly 2 years after initial presentation), she showed sustained resolution, with small pitted scars on both cheeks and the nasal tip.

Patient 2—A 33-year-old Ethiopian woman presented with a facial rash of 15 years’ duration. The lesions had been accumulating slowly and were asymptomatic. Physical examination revealed multiple follicular-based, flesh-colored, and erythematous papules on the cheeks, chin, perioral area, and forehead (Figure 4). There were no pustules or telangiectasias. Treatment with tretinoin cream 0.05% for 6 months offered minimal relief.

FIGURE 4. Numerous flesh-colored, dome-shaped papules are seen over parts of the right face in patient 2, including the inferolateral forehead, temple, and cheek, but not the upper eyelid.

 

 

Biopsy of a papule from the left mandible showed superficial vascular telangiectasias, noncaseating granulomas comprising epithelioid histiocytes and lymphocytes in the superficial dermis, and a perifollicular lymphocytic infiltrate (Figure 5). No organisms were identified on Fite or Gomori methenamine silver staining.

FIGURE 5. Histologic view of a papular lesion in patient 2. Magnified view of the superficial dermis demonstrated epithelioid and lymphocytic infiltrates, some of which were trying to form granulomas. Superficial dermal telangiectasias also were present (H&E, original magnification ×10).

Comment

The first step in differentiating cutaneous granulomatous lesions should be to distinguish infectious from noninfectious causes.1 Noninfectious cutaneous granulomas can appear nearly anywhere; however, certain processes have a predilection for the face, including GPD, GR, LMDF, and papular sarcoidosis.5-7 These conditions generally present with papular granulomas with features as described above.

Granulomatous Periorificial Dermatitis—In 1970, Gianotti and colleagues8 briefly described the first possible cases of GPD in 5 children. The eruption comprised numerous yellow, dome-shaped papules in a mostly perioral distribution. Tuberculin and the Kveim tests were nonreactive; histopathology was described as sarcoid-type and not necessarily follicular or perifollicular.8 In 1974, Marten et al9 described 22 Afro-Caribbean children with flesh-colored, papular eruptions on the face that did not show histologic granulomatous changes but were morphologically similar to the reports by Gianotti et al.8 By 1989, Frieden and colleagues10 described this facial eruption as “granulomatous perioral dermatitis in children”. Additionally, the investigators observed granulomatous infiltrates in a perifollicular distribution and suggested follicular disruption as a possible cause. It was clear from the case discussions that these eruptions were not uncommonly diagnosed as papular sarcoidosis.10 The following year, Williams et al11 reported 5 cases of similar papular eruptions in 5 Afro-Caribbean children, coining the term facial Afro-Caribbean eruption.11 Knautz and Lesher12 referred to this entity as “childhood GPD” in 1996 to avoid limiting the diagnosis to Afro-Caribbean patients and to a perioral distribution; this is the most popular current terminology.12 Since then, reports of extrafacial involvement and disease in adults have been published.13,14

Granulomatous periorificial dermatitis often is seen in the perinasal, periocular, and perioral regions of the face.2 It is associated with topical steroid exposure.5 Histologically, noncaseating granulomas around the upper half of undisrupted hair follicles with a lymphocytic infiltrate are typical.13 Treatment should begin with cessation of any topical steroids; first-line agents are oral tetracycline or macrolide antibiotics.5 These agents can be used alone or in combination with topical erythromycin, metronidazole, or sulfur-based lotions.13 Rarely, GPD presents extrafacially.13 Even so, it usually resolves within 2 weeks to 6 months, especially with therapy; scarring is unusual.5,13,15

Granulomatous Rosacea—A report in the early 20th century described patients with tuberculoid granulomas resembling papular rosacea; the initial belief was that this finding represented a rosacealike tuberculid eruption.5 However, this belief was questioned by Snapp,16 among others, who demonstrated near universal lack of reactivity to tuberculin among 20 of these patients in 1949; more recent evidence has substantiated these findings.17 Still, Snapp16 postulated that these rosacealike granulomatous lesions were distinct from classic rosacea because they lacked vascular symptoms and pustules and were recalcitrant to rosacea treatment modalities.

 

 

In 1970, Mullanax and colleagues18 introduced the term granulomatous rosacea, reiterating that this entity was not tuberculous. They documented papulopustular lesions as well as telangiectasias, raising the possibility that GR does overlap with acne rosacea. More recent studies have established the current theory that GR is a histologic variant of acne rosacea because, in addition to typical granulomatous papules, its microscopic features can be seen across subtypes of acne rosacea.19,20

Various causes have been proposed for GR. Demodex mites have been reported in association with GR for nearly 30 years.19,20 In the past 10 years, molecular studies have started to define the role of metalloproteinases, UV radiation, and cutaneous peptides in the pathogenesis of acne rosacea and GR.21,22

Granulomatous rosacea typically is seen in middle-aged women.20,23 Hallmarks of rosacea, such as facial erythema, flushing, telangiectasias, pustules, and rhinophyma, are not always present in GR.5,20,23 Lesions usually are distributed around the central face, although extension to the cheeks, total facial involvement, and extrafacial lesions are possible.5,20 Histologically, perifollicular and follicular-based noncaseating granulomas with dilatation of the dermal papillary vasculature are seen.17,23 As a whole, rosacea is comparatively uncommon in dark-skinned patients; when it does occur, GR is a frequent presentation.24

First-line treatment for GR is tetracycline antibiotics.5 Unresponsive cases have been treated—largely anecdotally—with topical modalities (eg, metronidazole, steroids, immunomodulators), systemic agents (eg, dapsone, erythromycin, isotretinoin), and other therapies.5 Granulomatous rosacea tends to have a chronic course.5,23

Lupus Miliaris Disseminatus Faciei—Classic LMDF demonstrates caseating perifollicular granulomas histologically.6,17,25 Lesions tend to appear on the central face, particularly the eyelids, and can be seen extrafacially.3,6,25,26 Although LMDF originally was categorized as a tuberculid eruption, this no longer is thought to be the case.27 It is now regarded by some as a variant of GR25; however, LMDF responds poorly to tetracyclines, is more common in males, and lacks rosacealike vascular abnormalities, leading some to question this association.3,6,17 In the past 20 years, some have proposed renaming LMDF to better reflect its clinical course and to consider it independent of tuberculosis and GR.28 It usually resolves spontaneously after 1 to 3 years, leaving pitted scars.3,6

 

 

Papular Sarcoidosis—The first potential documented case of sarcoidosis was by Hutchinson29 in 1869 in a patient seen in London. The author labeled purple plaques on the index patient’s legs and hands as “livid papillary psoriasis.” In 1889, Besnier30 described a patient with violaceous swellings on the nose, ears, and fingers, which he called “lupus pernio”; his contemporary, Tenneson,31 published a case of lupus pernio and described its histologic profile as comprising epithelioid cells and giant cells. It was not until 1899 that the term sarkoid was used to describe these cutaneous lesions by Boeck,32 who thought they were reminiscent of sarcoma. In 1915, Kuznitsky and Bittorf33 described a patient with cutaneous lesions histologically consistent with Boeck’s sarkoid but additionally with hilar lymphadenopathy and pulmonary infiltrates. Around 1916 or 1917, Schaumann34 described patients with cutaneous lesions and additionally with involvement of pulmonary, osseous, hepatosplenic, and tonsillar tissue. These reports are among the first to recognize the multisystemic nature of sarcoidosis. The first possible case of childhood sarcoidosis might have been reported by Osler35 in the United States in 1898.

In the past century or so, an ongoing effort by researchers has focused on identifying etiologic triggers for sarcoidosis. Microbial agents have been considered in this role, with Mycobacterium and Propionibacterium organisms the most intensively studied; the possibility that foreign material contributes to the formation of granulomas also has been raised.36 Current models of the pathogenesis of sarcoidosis involve an interplay between the immune system in genetically predisposed patients and an infection that leads to a hyperimmune type 1 T–helper cell response that clears the infection but not antigens generated by the microbes and the acute host response, including proteins such as serum amyloid A and vimentin.36,37 These antigens aggregate and serve as a nidus for granuloma formation and maintenance long after infection has resolved.

Cutaneous lesions of sarcoidosis include macules, papules, plaques, and lupus pernio, as well as lesions arising within scars or tattoos, with many less common presentations.7,38 Papular sarcoidosis is common on the face but also can involve the extremities.4,7 Strictly, at least 2 organ systems must be involved to diagnose sarcoidosis, but this is debatable.4,7 Among 41 patients with cutaneous sarcoidosis, 24 (58.5%) had systemic disease; cutaneous lesions were the presenting sign in 87.5% (21/24) of patients.38 Histologic analysis, regardless of the lesion, usually shows noncaseating so-called “naked” granulomas, which have minimal lymphocytic infiltrate associated with the epithelioid histiocytes.38,39 Perifollicular granulomas are possible but unusual.40

Treatment depends on the extent of cutaneous and systemic involvement. Pharmacotherapeutic modalities include topical steroids, immunomodulators, and retinoids; systemic immunomodulators and immunosuppressants; and biologic agents.7 Isolated cutaneous sarcoidosis, particularly the papular variant, usually is associated with acute disease lasting less than 2 years, with resolution of skin lesions.7,38 That said, a recent report suggested that cutaneous sarcoidosis can progress to multisystemic disease as long as 7 years after the initial diagnosis.41

Clinical and Histologic Overlap—Despite this categorization of noninfectious facial granulomatous conditions, each has some clinical and histologic overlap with the others, which must be considered when encountering a granulomatous facial dermatosis. Both GPD and GR tend to present with lesions near the eyes, mouth, and nose, although GR can extend to lateral aspects of the face, below the mandible, and the forehead and has different demographic features.15,20,23 Granulomas in both GPD and GR generally are noncaseating and form in a follicular or perifollicular distribution within the dermis.2,15,23 Lupus miliaris disseminatus faciei and GR share a similar facial distribution in some cases.17,20 Even papular cutaneous sarcoidosis has masqueraded as GR clinically and histologically.4

 

 

Diagnostic and Treatment Difficulty—Our cases illustrate the range of difficulty in evaluating and managing patients with facial papular granulomas. On one hand, our adult patient’s clinical and histologic findings were highly consistent with GR; on the other hand, our younger patient had clinicopathologic features of both sarcoidosis and GPD at varying times. Both conditions are more common in dark-skinned patients.11,42

Juvenile sarcoidosis is comparatively rare, with a reported annual incidence of 0.22 to 0.27 for every 100,000 children younger than 15 years; however, juvenile sarcoidosis commonly presents around 8 to 15 years of age.43

It is unusual for sarcoid granulomas to be isolated to the skin, much less to the face.4,7,43,44 Patient 1 initially presented in this manner and lacked convincing laboratory or radiographic evidence of systemic sarcoidosis. Bilateral hilar calcifications in sarcoidosis are more typical among adults after 5 to 20 years; there were no signs or symptoms of active infection that could account for the pulmonary and cutaneous lesions.45

The presence of perifollicular granulomas with associated lymphocytic infiltrates on repeat biopsy, coupled with the use of topical steroids, made it difficult to rule out a contribution by GPD to her clinical course. That her lesions resolved with pitted scarring while she was taking methotrexate and after topical steroids had been stopped could be the result of successful management or spontaneous resolution of her dermatosis; both papular sarcoidosis and GPD tend to have a self-limited course.7,13

Conclusion

We present 2 cases of papular facial granulomas in patients with similar skin types who had different clinical courses. Evaluation of such lesions remains challenging given the similarity between specific entities that present in this manner. Certainly, it is reasonable to consider a spectrum upon which all of these conditions fall, in light of the findings of these cases and those reported previously.

References
  1. Beretta-Piccoli BT, Mainetti C, Peeters M-A, et al. Cutaneous granulomatosis: a comprehensive review. Clin Rev Allergy Immunol. 2018;54:131-146. doi:10.1007/s12016-017-8666-8
  2. Lucas CR, Korman NJ, Gilliam AC. Granulomatous periorificial dermatitis: a variant of granulomatous rosacea in children? J Cutan Med Surg. 2009;13:115-118. doi:10.2310/7750.2008.07088
  3. van de Scheur MR, van der Waal RIF, Starink TM. Lupus miliaris disseminatus faciei: a distinctive rosacea-like syndrome and not a granulomatous form of rosacea. Dermatology. 2003;206:120-123. doi:10.1159/000068457
  4. Simonart T, Lowy M, Rasquin F, et al. Overlap of sarcoidosis and rosacea. Dermatology. 1997;194:416-418. doi:10.1159/000246165
  5. Lee GL, Zirwas MJ. Granulomatous rosacea and periorificial dermatitis: controversies and review of management. Dermatol Clin. 2015;33:447-455. doi:10.1016/j.det.2015.03.009
  6. Michaels JD, Cook-Norris RH, Lehman JS, et al. Adult with papular eruption of the central aspect of the face. J Am Acad Dermatol. 2014;71:410-412. doi:10.1016/j.jaad.2012.06.039
  7. Wanat KA, Rosenbach M. Cutaneous sarcoidosis. Clin Chest Med. 2015;38:685-702. doi:10.1016/j.ccm.2015.08.010
  8. Gianotti F, Ermacora E, Benelli MG, et al. Particulière dermatite peri-orale infantile. observations sur 5 cas. Bull Soc Fr Dermatol Syphiligr. 1970;77:341.
  9. Marten RH, Presbury DG, Adamson JE, et al. An unusual papular and acneiform facial eruption in the negro child. Br J Dermatol. 1974;91:435-438. doi:10.1111/j.1365-2133.1974.tb13083.x
  10. Frieden IJ, Prose NS, Fletcher V, et al. Granulomatous perioral dermatitis in children. Arch Dermatol. 1989;125:369-373.
  11. Williams HC, Ashworth J, Pembroke AC, et al. FACE—facial Afro-Caribbean childhood eruption. Clin Exp Dermatol. 1990;15:163-166. doi:10.1111/j.1365-2230.1990.tb02063.x
  12. Knautz MA, Lesher JL Jr. Childhood granulomatous periorificial dermatitis. Pediatr Dermatol. 1996;13:131-134. doi:10.1111/j.1525-1470.1996.tb01419.x
  13. Urbatsch AJ, Frieden I, Williams ML, et al. Extrafacial and generalized granulomatous periorificial dermatitis. Arch Dermatol. 2002;138:1354-1358. doi:10.1001/archderm.138.10.1354
  14. Vincenzi C, Parente G, Tosti A. Perioral granulomatous dermatitis: two cases treated with clarithromycin. J Dermatol Treat. 2000;11:57-61.
  15. Kim YJ, Shin JW, Lee JS, et al. Childhood granulomatous periorificial dermatitis. Ann Dermatol. 2011;23:386-388. doi:10.5021/ad.2011.23.3.386
  16. Snapp RH. Lewandowsky’s rosacea-like eruption; a clinical study. J Invest Dermatol. 1949;13:175-190. doi:10.1038/jid.1949.86
  17. Chougule A, Chatterjee D, Sethi S, et al. Granulomatous rosacea versus lupus miliaris disseminatus faciei—2 faces of facial granulomatous disorder: a clinicohistological and molecular study. Am J Dermatopathol. 2018;40:819-823. doi:10.1097/DAD.0000000000001243
  18. Mullanax MG, Kierland RR. Granulomatous rosacea. Arch Dermatol. 1970;101:206-211.
  19. Sánchez JL, Berlingeri-Ramos AC, Dueño DV. Granulomatous rosacea. Am J Dermatopathol. 2008;30:6-9. doi:10.1097/DAD.0b013e31815bc191
  20. Helm KF, Menz J, Gibson LE, et al. A clinical and histopathologic study of granulomatous rosacea. J Am Acad Dermatol. 1991;25:1038-1043. doi:10.1016/0190-9622(91)70304-k
  21. Kanada KN, Nakatsuji T, Gallo RL. Doxycycline indirectly inhibits proteolytic activation of tryptic kallikrein-related peptidases and activation of cathelicidin. J Invest Dermatol. 2012;132:1435-1442. doi:10.1038/jid.2012.14
  22. Jang YH, Sim JH, Kang HY, et al. Immunohistochemical expression of matrix metalloproteinases in the granulomatous rosacea compared with the non-granulomatous rosacea. J Eur Acad Dermatol Venereol. 2011;25:544-548. doi:10.1111/j.1468-3083.2010.03825.x
  23. Khokhar O, Khachemoune A. A case of granulomatous rosacea: sorting granulomatous rosacea from other granulomatous diseases that affect the face. Dermatol Online J. 2004;10:6.
  24. Rosen T, Stone MS. Acne rosacea in blacks. J Am Acad Dermatol. 1987;17:70-73. doi:10.1016/s0190-9622(87)70173-x
  25. Adams AK, Davis JL, Davis MDP, et al. What is your diagnosis? granulomatous rosacea (lupus miliaris disseminatus faciei, acne agminata). Cutis. 2008;82:103-112.
  26. Shitara A. Lupus miliaris disseminatus faciei. Int J Dermatol. 1984;23:542-544. doi:10.1111/j.1365-4362.1984.tb04206.x
  27. Hodak E, Trattner A, Feuerman H, et al. Lupus miliaris disseminatus faciei—the DNA of Mycobacterium tuberculosis is not detectable in active lesions by polymerase chain reaction. Br J Dermatol. 1997;137:614-619. doi: 10.1111/j.1365-2133.1997.tb03797.x
  28. Skowron F, Causeret AS, Pabion C, et al. F.I.GU.R.E.: facial idiopathic granulomas with regressive evolution. Dermatology. 2000;201:287-289. doi:10.1159/000051539
  29. Hutchinson J. Case of livid papillary psoriasis. In: London J, Churchill A, eds. Illustrations of Clinical Surgery. J&A Churchill; 1877:42-43.
  30. Besnier E. Lupus pernio of the face [in French]. Ann Dermatol Syphiligr (Paris). 1889;10:33-36.
  31. Tenneson H. Lupus pernio. Ann Dermatol Syphiligr (Paris). 1889;10:333-336.
  32. Boeck C. Multiple benign sarkoid of the skin [in Norwegian]. Norsk Mag Laegevidensk. 1899;14:1321-1334.
  33. Kuznitsky E, Bittorf A. Sarkoid mit beteiligung innerer organe. Münch Med Wochenschr. 1915;62:1349-1353.
  34. Schaumann J. Etude sur le lupus pernio et ses rapports avec les sarcoides et la tuberculose. Ann Dermatol Syphiligr. 1916-1917;6:357-373.
  35. Osler W. On chronic symmetrical enlargement of the salivary and lacrimal glands. Am J Med Sci. 1898;115:27-30.
  36. Chen ES, Moller DR. Etiologies of sarcoidosis. Clin Rev Allergy Immunol. 2015;49:6-18. doi:10.1007/s12016-015-8481-z
  37. Eberhardt C, Thillai M, Parker R, et al. Proteomic analysis of Kveim reagent identifies targets of cellular immunity in sarcoidosis. PLoS One. 2017;12:e0170285. doi:10.1371/journal.pone.0170285
  38. Esteves TC, Aparicio G, Ferrer B, et al. Prognostic value of skin lesions in sarcoidosis: clinical and histopathological clues. Eur J Dermatol. 2015;25:556-562. doi:10.1684/ejd.2015.2666
  39. Cardoso JC, Cravo M, Reis JP, et al. Cutaneous sarcoidosis: a histopathological study. J Eur Acad Dermatol Venereol. 2009;23:678-682. doi:10.1111/j.1468-3083.2009.03153.x
  40. Mangas C, Fernández-Figueras M-T, Fité E, et al. Clinical spectrum and histological analysis of 32 cases of specific cutaneous sarcoidosis. J Cutan Pathol. 2006;33:772-777. doi:10.1111/j.1600-0560.2006.00563.x
  41. García-Colmenero L, Sánchez-Schmidt JM, Barranco C, et al. The natural history of cutaneous sarcoidosis. clinical spectrum and histological analysis of 40 cases. Int J Dermatol. 2019;58:178-184. doi: 10.1111/ijd.14218
  42. Shetty AK, Gedalia A. Childhood sarcoidosis: a rare but fascinating disorder. Pediatr Rheumatol Online J. 2008;6:16. doi:10.1186/1546-0096-6-16
  43. Milman N, Hoffmann AL, Byg KE. Sarcoidosis in children. epidemiology in Danes, clinical features, diagnosis, treatment and prognosis. Acta Paediatr. 1998;87:871-878. doi:10.1080/08035259875001366244. S¸ims¸ek A, Çelikten H, Yapıcı I. Isolated cutaneous sarcoidosis. Arch Bronconeumol. 2016;52:220.
  44. Scadding JG. The late stages of pulmonary sarcoidosis. Postgrad Med J. 1970;46:530-536. doi:10.1136/pgmj.46.538.530
References
  1. Beretta-Piccoli BT, Mainetti C, Peeters M-A, et al. Cutaneous granulomatosis: a comprehensive review. Clin Rev Allergy Immunol. 2018;54:131-146. doi:10.1007/s12016-017-8666-8
  2. Lucas CR, Korman NJ, Gilliam AC. Granulomatous periorificial dermatitis: a variant of granulomatous rosacea in children? J Cutan Med Surg. 2009;13:115-118. doi:10.2310/7750.2008.07088
  3. van de Scheur MR, van der Waal RIF, Starink TM. Lupus miliaris disseminatus faciei: a distinctive rosacea-like syndrome and not a granulomatous form of rosacea. Dermatology. 2003;206:120-123. doi:10.1159/000068457
  4. Simonart T, Lowy M, Rasquin F, et al. Overlap of sarcoidosis and rosacea. Dermatology. 1997;194:416-418. doi:10.1159/000246165
  5. Lee GL, Zirwas MJ. Granulomatous rosacea and periorificial dermatitis: controversies and review of management. Dermatol Clin. 2015;33:447-455. doi:10.1016/j.det.2015.03.009
  6. Michaels JD, Cook-Norris RH, Lehman JS, et al. Adult with papular eruption of the central aspect of the face. J Am Acad Dermatol. 2014;71:410-412. doi:10.1016/j.jaad.2012.06.039
  7. Wanat KA, Rosenbach M. Cutaneous sarcoidosis. Clin Chest Med. 2015;38:685-702. doi:10.1016/j.ccm.2015.08.010
  8. Gianotti F, Ermacora E, Benelli MG, et al. Particulière dermatite peri-orale infantile. observations sur 5 cas. Bull Soc Fr Dermatol Syphiligr. 1970;77:341.
  9. Marten RH, Presbury DG, Adamson JE, et al. An unusual papular and acneiform facial eruption in the negro child. Br J Dermatol. 1974;91:435-438. doi:10.1111/j.1365-2133.1974.tb13083.x
  10. Frieden IJ, Prose NS, Fletcher V, et al. Granulomatous perioral dermatitis in children. Arch Dermatol. 1989;125:369-373.
  11. Williams HC, Ashworth J, Pembroke AC, et al. FACE—facial Afro-Caribbean childhood eruption. Clin Exp Dermatol. 1990;15:163-166. doi:10.1111/j.1365-2230.1990.tb02063.x
  12. Knautz MA, Lesher JL Jr. Childhood granulomatous periorificial dermatitis. Pediatr Dermatol. 1996;13:131-134. doi:10.1111/j.1525-1470.1996.tb01419.x
  13. Urbatsch AJ, Frieden I, Williams ML, et al. Extrafacial and generalized granulomatous periorificial dermatitis. Arch Dermatol. 2002;138:1354-1358. doi:10.1001/archderm.138.10.1354
  14. Vincenzi C, Parente G, Tosti A. Perioral granulomatous dermatitis: two cases treated with clarithromycin. J Dermatol Treat. 2000;11:57-61.
  15. Kim YJ, Shin JW, Lee JS, et al. Childhood granulomatous periorificial dermatitis. Ann Dermatol. 2011;23:386-388. doi:10.5021/ad.2011.23.3.386
  16. Snapp RH. Lewandowsky’s rosacea-like eruption; a clinical study. J Invest Dermatol. 1949;13:175-190. doi:10.1038/jid.1949.86
  17. Chougule A, Chatterjee D, Sethi S, et al. Granulomatous rosacea versus lupus miliaris disseminatus faciei—2 faces of facial granulomatous disorder: a clinicohistological and molecular study. Am J Dermatopathol. 2018;40:819-823. doi:10.1097/DAD.0000000000001243
  18. Mullanax MG, Kierland RR. Granulomatous rosacea. Arch Dermatol. 1970;101:206-211.
  19. Sánchez JL, Berlingeri-Ramos AC, Dueño DV. Granulomatous rosacea. Am J Dermatopathol. 2008;30:6-9. doi:10.1097/DAD.0b013e31815bc191
  20. Helm KF, Menz J, Gibson LE, et al. A clinical and histopathologic study of granulomatous rosacea. J Am Acad Dermatol. 1991;25:1038-1043. doi:10.1016/0190-9622(91)70304-k
  21. Kanada KN, Nakatsuji T, Gallo RL. Doxycycline indirectly inhibits proteolytic activation of tryptic kallikrein-related peptidases and activation of cathelicidin. J Invest Dermatol. 2012;132:1435-1442. doi:10.1038/jid.2012.14
  22. Jang YH, Sim JH, Kang HY, et al. Immunohistochemical expression of matrix metalloproteinases in the granulomatous rosacea compared with the non-granulomatous rosacea. J Eur Acad Dermatol Venereol. 2011;25:544-548. doi:10.1111/j.1468-3083.2010.03825.x
  23. Khokhar O, Khachemoune A. A case of granulomatous rosacea: sorting granulomatous rosacea from other granulomatous diseases that affect the face. Dermatol Online J. 2004;10:6.
  24. Rosen T, Stone MS. Acne rosacea in blacks. J Am Acad Dermatol. 1987;17:70-73. doi:10.1016/s0190-9622(87)70173-x
  25. Adams AK, Davis JL, Davis MDP, et al. What is your diagnosis? granulomatous rosacea (lupus miliaris disseminatus faciei, acne agminata). Cutis. 2008;82:103-112.
  26. Shitara A. Lupus miliaris disseminatus faciei. Int J Dermatol. 1984;23:542-544. doi:10.1111/j.1365-4362.1984.tb04206.x
  27. Hodak E, Trattner A, Feuerman H, et al. Lupus miliaris disseminatus faciei—the DNA of Mycobacterium tuberculosis is not detectable in active lesions by polymerase chain reaction. Br J Dermatol. 1997;137:614-619. doi: 10.1111/j.1365-2133.1997.tb03797.x
  28. Skowron F, Causeret AS, Pabion C, et al. F.I.GU.R.E.: facial idiopathic granulomas with regressive evolution. Dermatology. 2000;201:287-289. doi:10.1159/000051539
  29. Hutchinson J. Case of livid papillary psoriasis. In: London J, Churchill A, eds. Illustrations of Clinical Surgery. J&A Churchill; 1877:42-43.
  30. Besnier E. Lupus pernio of the face [in French]. Ann Dermatol Syphiligr (Paris). 1889;10:33-36.
  31. Tenneson H. Lupus pernio. Ann Dermatol Syphiligr (Paris). 1889;10:333-336.
  32. Boeck C. Multiple benign sarkoid of the skin [in Norwegian]. Norsk Mag Laegevidensk. 1899;14:1321-1334.
  33. Kuznitsky E, Bittorf A. Sarkoid mit beteiligung innerer organe. Münch Med Wochenschr. 1915;62:1349-1353.
  34. Schaumann J. Etude sur le lupus pernio et ses rapports avec les sarcoides et la tuberculose. Ann Dermatol Syphiligr. 1916-1917;6:357-373.
  35. Osler W. On chronic symmetrical enlargement of the salivary and lacrimal glands. Am J Med Sci. 1898;115:27-30.
  36. Chen ES, Moller DR. Etiologies of sarcoidosis. Clin Rev Allergy Immunol. 2015;49:6-18. doi:10.1007/s12016-015-8481-z
  37. Eberhardt C, Thillai M, Parker R, et al. Proteomic analysis of Kveim reagent identifies targets of cellular immunity in sarcoidosis. PLoS One. 2017;12:e0170285. doi:10.1371/journal.pone.0170285
  38. Esteves TC, Aparicio G, Ferrer B, et al. Prognostic value of skin lesions in sarcoidosis: clinical and histopathological clues. Eur J Dermatol. 2015;25:556-562. doi:10.1684/ejd.2015.2666
  39. Cardoso JC, Cravo M, Reis JP, et al. Cutaneous sarcoidosis: a histopathological study. J Eur Acad Dermatol Venereol. 2009;23:678-682. doi:10.1111/j.1468-3083.2009.03153.x
  40. Mangas C, Fernández-Figueras M-T, Fité E, et al. Clinical spectrum and histological analysis of 32 cases of specific cutaneous sarcoidosis. J Cutan Pathol. 2006;33:772-777. doi:10.1111/j.1600-0560.2006.00563.x
  41. García-Colmenero L, Sánchez-Schmidt JM, Barranco C, et al. The natural history of cutaneous sarcoidosis. clinical spectrum and histological analysis of 40 cases. Int J Dermatol. 2019;58:178-184. doi: 10.1111/ijd.14218
  42. Shetty AK, Gedalia A. Childhood sarcoidosis: a rare but fascinating disorder. Pediatr Rheumatol Online J. 2008;6:16. doi:10.1186/1546-0096-6-16
  43. Milman N, Hoffmann AL, Byg KE. Sarcoidosis in children. epidemiology in Danes, clinical features, diagnosis, treatment and prognosis. Acta Paediatr. 1998;87:871-878. doi:10.1080/08035259875001366244. S¸ims¸ek A, Çelikten H, Yapıcı I. Isolated cutaneous sarcoidosis. Arch Bronconeumol. 2016;52:220.
  44. Scadding JG. The late stages of pulmonary sarcoidosis. Postgrad Med J. 1970;46:530-536. doi:10.1136/pgmj.46.538.530
Issue
Cutis - 108(4)
Issue
Cutis - 108(4)
Page Number
E5-E10
Page Number
E5-E10
Publications
Publications
Topics
Article Type
Display Headline
Granulomatous Facial Dermatoses
Display Headline
Granulomatous Facial Dermatoses
Sections
Inside the Article

Practice Points

  • Dermatologists should be aware that noninfectious granulomatous dermatosis of the face can be caused by granulomatous periorificial dermatitis, granulomatous rosacea, lupus miliaris disseminatus faciei, and papular sarcoidosis.
  • These conditions lie on a spectrum, suggested by their historical description and clinical and histological features.
  • Because their clinical courses can vary considerably from patient to patient, a thorough effort should be made to differentiate these conditions.
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
Article PDF Media

Unexpected Complications: A Case of Rosacea Fulminans in Pregnancy

Article Type
Changed
Fri, 08/20/2021 - 09:16

Rosacea fulminans (RF) is a rare facial dermatosis characterized by its fulminating course. 1 It presents with superficial and deep-seated papules, pustules, and nodules combined with an intense reddish or cyanotic erythema localized to the face. Furthermore, there is an absence of comedones and involvement of the chest or back. 2 Rosacea fulminans primarily affects women and often is, but not always, proceeded by seborrhea, chronic acne vulgaris, or rosacea. Although the etiology of RF remains unknown, immunologic, hormonal, and vascular factors have been implicated. 3 We report a case of RF in a pregnant patient with a history of mild acne as a teenager that was long ago resolved.

Case Report

A 32-year-old pregnant woman (10 weeks’ gestation) presented with a rapidly progressing inflammatory disorder of the face of 1 month’s duration. The lesions developed 3 weeks after beginning progesterone therapy (200 mg vaginal suppository) for infertility due to polycystic ovary syndrome. Despite discontinuing progesterone for the last month, the patient’s lesions had dramatically worsened (Figure 1). Empiric cephalosporin treatment prescribed by her primary care physician yielded no improvement. Physical examination at the current presentation revealed erythematous nodules and pustules all over the face, coalescing into large thick plaques on the patient’s right cheek and chin. Submental nodes were palpable and tender. Based on the initial clinical findings, acne conglobata secondary to progesterone therapy was considered. The patient was given intralesional triamcinolone (2.5 mg/cc) injections to all larger nodules and several blue light treatments.

Figure 1. Rosacea fulminans in a pregnant woman at presentation (10 weeks’ gestation).

The injected areas had improved 5 days after the initial visit; however, the chin and right paranasal cheek developed even more nodules and papules coalescing into large plaques. After consulting the patient’s obstetrician, prednisone (20 mg once daily) was initiated. Three weeks later, the patient’s nodular lesions had improved, but there was a showering of more than 100 pustules and increased general erythema of the entire face (Figure 2). Crotamiton cream 10% (every day before noon), ivermectin cream 1% (every night at bedtime), and sodium sulfacetamide cleanser 10% once daily were added to the treatment plan.

Figure 2. Three weeks after starting prednisone, there was a showering of more than 100 pustules and increased general erythema of the entire face due to rosacea fulminans.


At 16 weeks’ gestation, there was slight improvement; however, there was still erythema on the entire face with scattered pustules and multiple papules and nodules. Many small ice-pick scars were seen on the cheeks and forehead. No comedones were observed. A punch biopsy of an intact papule showed a prominent inflammatory infiltrate with granulomatous reaction and numerous neutrophils predominantly affecting hair follicles. Based on the clinical presentation and histopathology, a diagnosis of RF was made. Azithromycin (250 mg once daily) and metronidazole cream 0.75% twice daily were added. Two weeks later there were fewer nodules but many papules, edema, and intense erythema. The prednisone dosage was increased to 40 mg once daily. Two weeks later, the patient showed improvement with fewer lesions, less edema, and less erythema. The patient was instructed to finish the azithromycin course and discontinue use. At 28 weeks’ gestation, a prednisone taper was started with the intention to reduce the daily dose by delivery.



The patient delivered a healthy girl (birth weight, 1.985 kg) prematurely at 34 weeks’ gestation. At 2 months postpartum, the patient’s existing lesions continued to spontaneously improve; however, she still had numerous nodules and papules and continued to develop new lesions and form additional scars. Isotretinoin was instituted at 3 months postpartum upon cessation of nursing. Three months later (40 mg/d isotretinoin), the patient was nearly clear. At 8 months postpartum, isotretinoin was discontinued after a course of 150 mg/kg.

Comment

Rosacea fulminans initially was called pyoderma faciale but was later regarded as a severe form of rosacea and was renamed rosacea fulminans.2 According to a PubMed search of articles indexed for MEDLINE using the terms pregnancy and rosacea fulminans or pyoderma faciale, we identified 12 publications reporting 20 cases of RF associated with pregnancy (Table). Although there is no substantial evidence regarding the exact mechanism, these cases indicate that pregnancy can be an exacerbating or causative factor in the pathogenesis of RF.

In addition to pregnancy, RF has been associated with inflammatory bowel disease, thyroid and liver disease, erythema nodosum, and severe emotional trauma. However, no organism has been consistently isolated, and no evidence of family history has been reported.1 Histopathologic findings are dependent on the stage of disease. Massive infiltrates of neutrophils may be observed in early stages. In older lesions, infiltrates take the form of epithelioid cell granulomas.2

Treatment of RF during pregnancy is challenging. Early and aggressive treatment with retinoids, tetracycline antibiotics, antiandrogenic contraceptives, and dapsone is recommended in patients who are not pregnant; these therapies are all contraindicated in pregnancy. Topical steroids can be safely used; however, systemic steroids usually are required to control RF. The use of systemic steroids can only be justified if the risks for intrauterine growth retardation, maternal diabetes mellitus, and hypertension outweigh the benefits of treating this severe disfiguring skin condition.10 A study by Bakar et al13 indicated that azithromycin is an effective and safe alternative in the treatment of RF. It has a superior pharmacokinetic profile compared to other macrolides and does not pose increased risks for congenital malformation or miscarriage. Because of the concomitant use of both azithromycin and prednisone, it is not possible to determine which had the larger role in the patient’s improvement.



Isotretinoin therapy in our patient led to substantial improvement of RF. Time will tell if the response will be durable. Also unknown is the risk for recurrence with subsequent pregnancies, which has not been reported in the literature. Although it is difficult to confidently say that pregnancy was the inciting factor in this patient’s RF, this case certainly provides more evidence for a link between pregnancy and RF.

References
  1. Jarrett R, Gonsalves R, Anstey AV. Differing obstetric outcomes of rosacea fulminans in pregnancy: report of three cases with review of pathogenesis and management. Clin Exp Dermatol. 2010;35:888-891. doi:10.1111/j.1365-2230.2010.03846.x
  2. Ferahbas A, Utas S, Mistik S, et al. Rosacea fulminans in pregnancy: case report and review of the literature. Am J Clin Dermatol. 2006;7:141-144. doi:10.2165/00128071-200607020-00007
  3. Fuentelsaz V, Ara M, Corredera C, et al. Rosacea fulminans in pregnancy: successful treatment with azithromycin. Clin Exp Dermatol. 2011;36:674-676. doi:10.1111/j.1365-2230.2011.04042.x
  4. Garayar Cantero M, Garabito Solovera E, Aguado García Á, et al. Use of permethrin in the treatment of rosacea fulminans during pregnancy: one case report. Dermatol Ther. 2020;33:E13436. doi:10.1111/dth.13436
  5. Demir O, Tas IS, Gunay B, et al. A rare dermatologic disease in pregnancy: rosacea fulminans—case report and review of the literature. Open Access Maced J Med Sci. 2018;6:1438-1441. doi:10.3889/oamjms.2018.267
  6. Markou AG, Alessandrini V, Muray JM, et al. Rosacea fulminans during pregnancy. Clin Exp Obstet Gynecol. 2017;44:157-159.
  7. Haenen CCP, Kouwenhoven STP, van Doorn R. Rosacea fulminans in pregnancy [in Dutch]. Ned Tijdschr Geneeskd. 2015;159:A8334.
  8. de Morais e Silva FA, Bonassi M, Steiner D, et al. Rosacea fulminans in pregnancy with ocular perforation. J Dtsch Dermatol Ges. 2011;9:542-543. doi:10.1111/j.1610-0387.2011.07616.x
  9. Cisse M, Maruani A, Bré C. Rosacea fulminans in the early course of a pregnancy by in vitro fertilization with embryo transfer [in French]. Ann Dermatol Venereol. 2008;135:675-678. doi:10.1016/j.annder.2008.04.015
  10. Lewis VJ, Holme SA, Wright A, et al. Rosacea fulminans in pregnancy. Br J Dermatol. 2004;151:917-919. doi:10.1111/j.1365-2133.2004.06190.x
  11. Plewig G, Jansen T, Kligman AM. Pyoderma faciale. a review and report of 20 additional cases: is it rosacea? Arch Dermatol. 1992;128:1611-1617. doi:10.1001/archderm.128.12.1611
  12.  
  13. Massa MC, Su WP. Pyoderma faciale: a clinical study of twenty-nine patients. J Am Acad Dermatol. 1982;6:84-91. doi:10.1016/s0190-9622(82)70008-8
  14.  
  15. Bakar O, Demirçay Z, Gürbüz O. Therapeutic potential of azithromycin in rosacea. Int J Dermatol. 2004;43:151-154. doi:10.1111/j.1365-4632.2004.01958.x
Article PDF
Author and Disclosure Information

Mr. Ranpariya is from Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey. Dr. Baldwin is from Acne Treatment and Research Center, Morristown, New Jersey.

The authors report no conflict of interest.

Correspondence: Varun Ranpariya, BA, 142 Joralemon St, Brooklyn, NY 11201 (vkr16@rwjms.rutgers.edu).

Issue
cutis - 108(1)
Publications
Topics
Page Number
51-54
Sections
Author and Disclosure Information

Mr. Ranpariya is from Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey. Dr. Baldwin is from Acne Treatment and Research Center, Morristown, New Jersey.

The authors report no conflict of interest.

Correspondence: Varun Ranpariya, BA, 142 Joralemon St, Brooklyn, NY 11201 (vkr16@rwjms.rutgers.edu).

Author and Disclosure Information

Mr. Ranpariya is from Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey. Dr. Baldwin is from Acne Treatment and Research Center, Morristown, New Jersey.

The authors report no conflict of interest.

Correspondence: Varun Ranpariya, BA, 142 Joralemon St, Brooklyn, NY 11201 (vkr16@rwjms.rutgers.edu).

Article PDF
Article PDF

Rosacea fulminans (RF) is a rare facial dermatosis characterized by its fulminating course. 1 It presents with superficial and deep-seated papules, pustules, and nodules combined with an intense reddish or cyanotic erythema localized to the face. Furthermore, there is an absence of comedones and involvement of the chest or back. 2 Rosacea fulminans primarily affects women and often is, but not always, proceeded by seborrhea, chronic acne vulgaris, or rosacea. Although the etiology of RF remains unknown, immunologic, hormonal, and vascular factors have been implicated. 3 We report a case of RF in a pregnant patient with a history of mild acne as a teenager that was long ago resolved.

Case Report

A 32-year-old pregnant woman (10 weeks’ gestation) presented with a rapidly progressing inflammatory disorder of the face of 1 month’s duration. The lesions developed 3 weeks after beginning progesterone therapy (200 mg vaginal suppository) for infertility due to polycystic ovary syndrome. Despite discontinuing progesterone for the last month, the patient’s lesions had dramatically worsened (Figure 1). Empiric cephalosporin treatment prescribed by her primary care physician yielded no improvement. Physical examination at the current presentation revealed erythematous nodules and pustules all over the face, coalescing into large thick plaques on the patient’s right cheek and chin. Submental nodes were palpable and tender. Based on the initial clinical findings, acne conglobata secondary to progesterone therapy was considered. The patient was given intralesional triamcinolone (2.5 mg/cc) injections to all larger nodules and several blue light treatments.

Figure 1. Rosacea fulminans in a pregnant woman at presentation (10 weeks’ gestation).

The injected areas had improved 5 days after the initial visit; however, the chin and right paranasal cheek developed even more nodules and papules coalescing into large plaques. After consulting the patient’s obstetrician, prednisone (20 mg once daily) was initiated. Three weeks later, the patient’s nodular lesions had improved, but there was a showering of more than 100 pustules and increased general erythema of the entire face (Figure 2). Crotamiton cream 10% (every day before noon), ivermectin cream 1% (every night at bedtime), and sodium sulfacetamide cleanser 10% once daily were added to the treatment plan.

Figure 2. Three weeks after starting prednisone, there was a showering of more than 100 pustules and increased general erythema of the entire face due to rosacea fulminans.


At 16 weeks’ gestation, there was slight improvement; however, there was still erythema on the entire face with scattered pustules and multiple papules and nodules. Many small ice-pick scars were seen on the cheeks and forehead. No comedones were observed. A punch biopsy of an intact papule showed a prominent inflammatory infiltrate with granulomatous reaction and numerous neutrophils predominantly affecting hair follicles. Based on the clinical presentation and histopathology, a diagnosis of RF was made. Azithromycin (250 mg once daily) and metronidazole cream 0.75% twice daily were added. Two weeks later there were fewer nodules but many papules, edema, and intense erythema. The prednisone dosage was increased to 40 mg once daily. Two weeks later, the patient showed improvement with fewer lesions, less edema, and less erythema. The patient was instructed to finish the azithromycin course and discontinue use. At 28 weeks’ gestation, a prednisone taper was started with the intention to reduce the daily dose by delivery.



The patient delivered a healthy girl (birth weight, 1.985 kg) prematurely at 34 weeks’ gestation. At 2 months postpartum, the patient’s existing lesions continued to spontaneously improve; however, she still had numerous nodules and papules and continued to develop new lesions and form additional scars. Isotretinoin was instituted at 3 months postpartum upon cessation of nursing. Three months later (40 mg/d isotretinoin), the patient was nearly clear. At 8 months postpartum, isotretinoin was discontinued after a course of 150 mg/kg.

Comment

Rosacea fulminans initially was called pyoderma faciale but was later regarded as a severe form of rosacea and was renamed rosacea fulminans.2 According to a PubMed search of articles indexed for MEDLINE using the terms pregnancy and rosacea fulminans or pyoderma faciale, we identified 12 publications reporting 20 cases of RF associated with pregnancy (Table). Although there is no substantial evidence regarding the exact mechanism, these cases indicate that pregnancy can be an exacerbating or causative factor in the pathogenesis of RF.

In addition to pregnancy, RF has been associated with inflammatory bowel disease, thyroid and liver disease, erythema nodosum, and severe emotional trauma. However, no organism has been consistently isolated, and no evidence of family history has been reported.1 Histopathologic findings are dependent on the stage of disease. Massive infiltrates of neutrophils may be observed in early stages. In older lesions, infiltrates take the form of epithelioid cell granulomas.2

Treatment of RF during pregnancy is challenging. Early and aggressive treatment with retinoids, tetracycline antibiotics, antiandrogenic contraceptives, and dapsone is recommended in patients who are not pregnant; these therapies are all contraindicated in pregnancy. Topical steroids can be safely used; however, systemic steroids usually are required to control RF. The use of systemic steroids can only be justified if the risks for intrauterine growth retardation, maternal diabetes mellitus, and hypertension outweigh the benefits of treating this severe disfiguring skin condition.10 A study by Bakar et al13 indicated that azithromycin is an effective and safe alternative in the treatment of RF. It has a superior pharmacokinetic profile compared to other macrolides and does not pose increased risks for congenital malformation or miscarriage. Because of the concomitant use of both azithromycin and prednisone, it is not possible to determine which had the larger role in the patient’s improvement.



Isotretinoin therapy in our patient led to substantial improvement of RF. Time will tell if the response will be durable. Also unknown is the risk for recurrence with subsequent pregnancies, which has not been reported in the literature. Although it is difficult to confidently say that pregnancy was the inciting factor in this patient’s RF, this case certainly provides more evidence for a link between pregnancy and RF.

Rosacea fulminans (RF) is a rare facial dermatosis characterized by its fulminating course. 1 It presents with superficial and deep-seated papules, pustules, and nodules combined with an intense reddish or cyanotic erythema localized to the face. Furthermore, there is an absence of comedones and involvement of the chest or back. 2 Rosacea fulminans primarily affects women and often is, but not always, proceeded by seborrhea, chronic acne vulgaris, or rosacea. Although the etiology of RF remains unknown, immunologic, hormonal, and vascular factors have been implicated. 3 We report a case of RF in a pregnant patient with a history of mild acne as a teenager that was long ago resolved.

Case Report

A 32-year-old pregnant woman (10 weeks’ gestation) presented with a rapidly progressing inflammatory disorder of the face of 1 month’s duration. The lesions developed 3 weeks after beginning progesterone therapy (200 mg vaginal suppository) for infertility due to polycystic ovary syndrome. Despite discontinuing progesterone for the last month, the patient’s lesions had dramatically worsened (Figure 1). Empiric cephalosporin treatment prescribed by her primary care physician yielded no improvement. Physical examination at the current presentation revealed erythematous nodules and pustules all over the face, coalescing into large thick plaques on the patient’s right cheek and chin. Submental nodes were palpable and tender. Based on the initial clinical findings, acne conglobata secondary to progesterone therapy was considered. The patient was given intralesional triamcinolone (2.5 mg/cc) injections to all larger nodules and several blue light treatments.

Figure 1. Rosacea fulminans in a pregnant woman at presentation (10 weeks’ gestation).

The injected areas had improved 5 days after the initial visit; however, the chin and right paranasal cheek developed even more nodules and papules coalescing into large plaques. After consulting the patient’s obstetrician, prednisone (20 mg once daily) was initiated. Three weeks later, the patient’s nodular lesions had improved, but there was a showering of more than 100 pustules and increased general erythema of the entire face (Figure 2). Crotamiton cream 10% (every day before noon), ivermectin cream 1% (every night at bedtime), and sodium sulfacetamide cleanser 10% once daily were added to the treatment plan.

Figure 2. Three weeks after starting prednisone, there was a showering of more than 100 pustules and increased general erythema of the entire face due to rosacea fulminans.


At 16 weeks’ gestation, there was slight improvement; however, there was still erythema on the entire face with scattered pustules and multiple papules and nodules. Many small ice-pick scars were seen on the cheeks and forehead. No comedones were observed. A punch biopsy of an intact papule showed a prominent inflammatory infiltrate with granulomatous reaction and numerous neutrophils predominantly affecting hair follicles. Based on the clinical presentation and histopathology, a diagnosis of RF was made. Azithromycin (250 mg once daily) and metronidazole cream 0.75% twice daily were added. Two weeks later there were fewer nodules but many papules, edema, and intense erythema. The prednisone dosage was increased to 40 mg once daily. Two weeks later, the patient showed improvement with fewer lesions, less edema, and less erythema. The patient was instructed to finish the azithromycin course and discontinue use. At 28 weeks’ gestation, a prednisone taper was started with the intention to reduce the daily dose by delivery.



The patient delivered a healthy girl (birth weight, 1.985 kg) prematurely at 34 weeks’ gestation. At 2 months postpartum, the patient’s existing lesions continued to spontaneously improve; however, she still had numerous nodules and papules and continued to develop new lesions and form additional scars. Isotretinoin was instituted at 3 months postpartum upon cessation of nursing. Three months later (40 mg/d isotretinoin), the patient was nearly clear. At 8 months postpartum, isotretinoin was discontinued after a course of 150 mg/kg.

Comment

Rosacea fulminans initially was called pyoderma faciale but was later regarded as a severe form of rosacea and was renamed rosacea fulminans.2 According to a PubMed search of articles indexed for MEDLINE using the terms pregnancy and rosacea fulminans or pyoderma faciale, we identified 12 publications reporting 20 cases of RF associated with pregnancy (Table). Although there is no substantial evidence regarding the exact mechanism, these cases indicate that pregnancy can be an exacerbating or causative factor in the pathogenesis of RF.

In addition to pregnancy, RF has been associated with inflammatory bowel disease, thyroid and liver disease, erythema nodosum, and severe emotional trauma. However, no organism has been consistently isolated, and no evidence of family history has been reported.1 Histopathologic findings are dependent on the stage of disease. Massive infiltrates of neutrophils may be observed in early stages. In older lesions, infiltrates take the form of epithelioid cell granulomas.2

Treatment of RF during pregnancy is challenging. Early and aggressive treatment with retinoids, tetracycline antibiotics, antiandrogenic contraceptives, and dapsone is recommended in patients who are not pregnant; these therapies are all contraindicated in pregnancy. Topical steroids can be safely used; however, systemic steroids usually are required to control RF. The use of systemic steroids can only be justified if the risks for intrauterine growth retardation, maternal diabetes mellitus, and hypertension outweigh the benefits of treating this severe disfiguring skin condition.10 A study by Bakar et al13 indicated that azithromycin is an effective and safe alternative in the treatment of RF. It has a superior pharmacokinetic profile compared to other macrolides and does not pose increased risks for congenital malformation or miscarriage. Because of the concomitant use of both azithromycin and prednisone, it is not possible to determine which had the larger role in the patient’s improvement.



Isotretinoin therapy in our patient led to substantial improvement of RF. Time will tell if the response will be durable. Also unknown is the risk for recurrence with subsequent pregnancies, which has not been reported in the literature. Although it is difficult to confidently say that pregnancy was the inciting factor in this patient’s RF, this case certainly provides more evidence for a link between pregnancy and RF.

References
  1. Jarrett R, Gonsalves R, Anstey AV. Differing obstetric outcomes of rosacea fulminans in pregnancy: report of three cases with review of pathogenesis and management. Clin Exp Dermatol. 2010;35:888-891. doi:10.1111/j.1365-2230.2010.03846.x
  2. Ferahbas A, Utas S, Mistik S, et al. Rosacea fulminans in pregnancy: case report and review of the literature. Am J Clin Dermatol. 2006;7:141-144. doi:10.2165/00128071-200607020-00007
  3. Fuentelsaz V, Ara M, Corredera C, et al. Rosacea fulminans in pregnancy: successful treatment with azithromycin. Clin Exp Dermatol. 2011;36:674-676. doi:10.1111/j.1365-2230.2011.04042.x
  4. Garayar Cantero M, Garabito Solovera E, Aguado García Á, et al. Use of permethrin in the treatment of rosacea fulminans during pregnancy: one case report. Dermatol Ther. 2020;33:E13436. doi:10.1111/dth.13436
  5. Demir O, Tas IS, Gunay B, et al. A rare dermatologic disease in pregnancy: rosacea fulminans—case report and review of the literature. Open Access Maced J Med Sci. 2018;6:1438-1441. doi:10.3889/oamjms.2018.267
  6. Markou AG, Alessandrini V, Muray JM, et al. Rosacea fulminans during pregnancy. Clin Exp Obstet Gynecol. 2017;44:157-159.
  7. Haenen CCP, Kouwenhoven STP, van Doorn R. Rosacea fulminans in pregnancy [in Dutch]. Ned Tijdschr Geneeskd. 2015;159:A8334.
  8. de Morais e Silva FA, Bonassi M, Steiner D, et al. Rosacea fulminans in pregnancy with ocular perforation. J Dtsch Dermatol Ges. 2011;9:542-543. doi:10.1111/j.1610-0387.2011.07616.x
  9. Cisse M, Maruani A, Bré C. Rosacea fulminans in the early course of a pregnancy by in vitro fertilization with embryo transfer [in French]. Ann Dermatol Venereol. 2008;135:675-678. doi:10.1016/j.annder.2008.04.015
  10. Lewis VJ, Holme SA, Wright A, et al. Rosacea fulminans in pregnancy. Br J Dermatol. 2004;151:917-919. doi:10.1111/j.1365-2133.2004.06190.x
  11. Plewig G, Jansen T, Kligman AM. Pyoderma faciale. a review and report of 20 additional cases: is it rosacea? Arch Dermatol. 1992;128:1611-1617. doi:10.1001/archderm.128.12.1611
  12.  
  13. Massa MC, Su WP. Pyoderma faciale: a clinical study of twenty-nine patients. J Am Acad Dermatol. 1982;6:84-91. doi:10.1016/s0190-9622(82)70008-8
  14.  
  15. Bakar O, Demirçay Z, Gürbüz O. Therapeutic potential of azithromycin in rosacea. Int J Dermatol. 2004;43:151-154. doi:10.1111/j.1365-4632.2004.01958.x
References
  1. Jarrett R, Gonsalves R, Anstey AV. Differing obstetric outcomes of rosacea fulminans in pregnancy: report of three cases with review of pathogenesis and management. Clin Exp Dermatol. 2010;35:888-891. doi:10.1111/j.1365-2230.2010.03846.x
  2. Ferahbas A, Utas S, Mistik S, et al. Rosacea fulminans in pregnancy: case report and review of the literature. Am J Clin Dermatol. 2006;7:141-144. doi:10.2165/00128071-200607020-00007
  3. Fuentelsaz V, Ara M, Corredera C, et al. Rosacea fulminans in pregnancy: successful treatment with azithromycin. Clin Exp Dermatol. 2011;36:674-676. doi:10.1111/j.1365-2230.2011.04042.x
  4. Garayar Cantero M, Garabito Solovera E, Aguado García Á, et al. Use of permethrin in the treatment of rosacea fulminans during pregnancy: one case report. Dermatol Ther. 2020;33:E13436. doi:10.1111/dth.13436
  5. Demir O, Tas IS, Gunay B, et al. A rare dermatologic disease in pregnancy: rosacea fulminans—case report and review of the literature. Open Access Maced J Med Sci. 2018;6:1438-1441. doi:10.3889/oamjms.2018.267
  6. Markou AG, Alessandrini V, Muray JM, et al. Rosacea fulminans during pregnancy. Clin Exp Obstet Gynecol. 2017;44:157-159.
  7. Haenen CCP, Kouwenhoven STP, van Doorn R. Rosacea fulminans in pregnancy [in Dutch]. Ned Tijdschr Geneeskd. 2015;159:A8334.
  8. de Morais e Silva FA, Bonassi M, Steiner D, et al. Rosacea fulminans in pregnancy with ocular perforation. J Dtsch Dermatol Ges. 2011;9:542-543. doi:10.1111/j.1610-0387.2011.07616.x
  9. Cisse M, Maruani A, Bré C. Rosacea fulminans in the early course of a pregnancy by in vitro fertilization with embryo transfer [in French]. Ann Dermatol Venereol. 2008;135:675-678. doi:10.1016/j.annder.2008.04.015
  10. Lewis VJ, Holme SA, Wright A, et al. Rosacea fulminans in pregnancy. Br J Dermatol. 2004;151:917-919. doi:10.1111/j.1365-2133.2004.06190.x
  11. Plewig G, Jansen T, Kligman AM. Pyoderma faciale. a review and report of 20 additional cases: is it rosacea? Arch Dermatol. 1992;128:1611-1617. doi:10.1001/archderm.128.12.1611
  12.  
  13. Massa MC, Su WP. Pyoderma faciale: a clinical study of twenty-nine patients. J Am Acad Dermatol. 1982;6:84-91. doi:10.1016/s0190-9622(82)70008-8
  14.  
  15. Bakar O, Demirçay Z, Gürbüz O. Therapeutic potential of azithromycin in rosacea. Int J Dermatol. 2004;43:151-154. doi:10.1111/j.1365-4632.2004.01958.x
Issue
cutis - 108(1)
Issue
cutis - 108(1)
Page Number
51-54
Page Number
51-54
Publications
Publications
Topics
Article Type
Sections
Inside the Article

Practice Points

  • Rosacea fulminans (RF) is a rare facial dermatosis that can present in pregnant patients.
  • Treatment of RF in a pregnant patient requires special considerations because typical therapies are contraindicated in pregnancy.
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
Article PDF Media

Acne and Rosacea Supplement

Article Type
Changed
Mon, 09/13/2021 - 12:25
Display Headline
Acne and Rosacea Supplement

Acne & Rosacea

  • Rosacea phenotypes
  • Erythema and flushing
  • Aggressive treatment of acne
  • Energy-based rosacea therapy
  • Diet and acne

 

 

Publications
Topics
Sections

Acne & Rosacea

  • Rosacea phenotypes
  • Erythema and flushing
  • Aggressive treatment of acne
  • Energy-based rosacea therapy
  • Diet and acne

 

 

Acne & Rosacea

  • Rosacea phenotypes
  • Erythema and flushing
  • Aggressive treatment of acne
  • Energy-based rosacea therapy
  • Diet and acne

 

 

Publications
Publications
Topics
Article Type
Display Headline
Acne and Rosacea Supplement
Display Headline
Acne and Rosacea Supplement
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
CFC Schedule Remove Status
Thu, 07/08/2021 - 11:45
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

Ocular Manifestations of Patients With Cutaneous Rosacea With and Without Demodex Infection

Article Type
Changed
Tue, 07/13/2021 - 23:07
Display Headline
Ocular Manifestations of Patients With Cutaneous Rosacea With and Without Demodex Infection

Acne rosacea is a chronic inflammatory disease that may affect the facial skin, eyes, and eyelids.1 It is characterized by transient or persistent flushing, facial erythema, and telangiectases, generally located on the central portion of the face, and may progress to papules and pustules.2,3 At the late stage of the disease, dermal edema or fibroplasia and sebaceous gland hypertrophy may cause phymatous alterations in the skin. In 2004, the National Rosacea Society Expert Committee developed a classification system for rosacea to standardize subtypes and variants that has since been widely accepted and continues to aid in research and epidemiologic studies.4 The committee defined 4 subtypes based on clinical characteristics: erythematotelangiectatic (ETR), papulopustular (PPR), phymatous, and ocular rosacea.2,3

Ocular rosacea may accompany mild, moderate, and severe dermatologic disease or may occur in the absence of diagnostic skin disease.5 Ocular signs include eyelid margin telangiectasia, spade-shaped infiltrates in the cornea, scleritis, and sclerokeratitis. Common symptoms include burning, stinging, light sensitivity, and foreign-body sensation. Ocular signs commonly seen in rosacea are meibomian gland dysfunction characterized by inspissation and inflammation of the meibomian glands (chalazia), conjunctivitis, honey crust and cylindrical collarette accumulation at the base of the eyelashes, irregularity of the eyelid margin architecture, and evaporative tear dysfunction.5,6

The physiopathology of rosacea is still unknown. Potential factors include genetic predisposition, abnormal inflammation, vascular dysfunction, and involvement of several microbial agents, such as commensal Demodex mites. The number of Demodex mites on normal skin flora is less than 5/cm2; however, the increased vascular dilation and capillary permeability associated with rosacea that result from sunlight and heat exposure increase the density of Demodex folliculorum.7 Elevated Demodex mite density has been observed in the lumens of the sebaceous follicles in patients with rosacea. However, because the severity of the clinical manifestations of the disease is not directly associated with the density of D folliculorum, it generally is accepted that D folliculorum is not a pathogenetic but rather an exacerbating factor.8 It has been reported that this species of mite is mostly found on the face and around the eyelashes and scalp of patients and that it can cause ocular surface inflammation.8

Most studies have researched ocular manifestations of rosacea but not ocular involvement in rosacea patients with and without Demodex mite infestation. In our study, we sought to compare the ocular surface, meibomian gland characteristics, and tear film abnormalities among patients with cutaneous rosacea with and without Demodex infestation.

Materials and Methods

We conducted a retrospective study of 60 patients with cutaneous rosacea. This study was approved by the ethics committee of the local hospital (2018/002-003), and all patients provided verbal and written informed consent before participating in the study. The study was carried out according to the guidelines of the Declaration of Helsinki.

Patient Selection and Evaluation
Patients diagnosed with rosacea by a dermatologist within 6 months were included in the study. Diagnosis of the disease was made after a detailed anamnesis and dermatologic examination. Rosacea was diagnosed if patients had an itching sensation, erythema and/or erythema attacks, and papules and pustules, and fulfilled the diagnostic criteria according to the National Rosacea Society. The skin disease was classified according to the subtypes as ETR, PPR, phymatous rosacea, or ocular rosacea.



The standard skin surface biopsy method was used in 60 patients for detecting Demodex density. When more than 5 mites were detected per square centimeter, the result was recorded as positive. Thirty consecutive, newly diagnosed patients with cutaneous acne rosacea with Demodex infestation and 30 consecutive, newly diagnosed sex- and age-matched patients with acne rosacea without Demodex infestation admitted to the dermatology outpatient clinic were included to this study. The patients who did not have any known dermatologic, systemic, or ocular diseases were included in the study. Patients who met any of the following criteria were excluded from the study: prior anti-inflammatory topical and/or systemic treatment for rosacea during the last 3 months, contact lens wear, eyelid surgery, or autoimmune disease requiring treatment.

 

 



Microscopic Demodex Examination
Demodex count was determined using a standardized skin surface biopsy, which is a noninvasive method. Every patient gave samples from the cheeks. This biopsy was repeated from the same site. A drop of cyanoacrylate was placed on a clean slide, pressed against a skin lesion, held in place for 1 minute, and removed. The obtained samples were evaluated under a light microscope (Nikon E200) with oil immersion. When more than 5 mites were detected per square centimeter, the result was recorded as positive.

Ophthalmologic Examination
A complete ophthalmologic examination including visual acuity assessment, standardized slit lamp examination, and fundus examination was done for all patients. Ocular rosacea was diagnosed on detection of 1 or more of the following: watery or bloodshot appearance, foreign-body sensation, burning or stinging, dryness, itching, light sensitivity, blurred vision, telangiectases of the conjunctiva and eyelid margin, eyelid lid and periocular erythema, anterior blepharitis, meibomian gland dysfunction, or irregularity of eyelid margins. All patients were screened for the signs and symptoms of ocular rosacea and underwent other ophthalmologic examinations, including tear function tests. Tear functions were evaluated with Schirmer tests without anesthesia and fluorescein tear breakup time (TBUT). Tear film breakup time was assessed after instillation of 2% fluorescein staining under a cobalt blue filter. The time interval between the last complete blink and the appearance of the first dry spot was recorded. The mean of 3 consecutive measurements was obtained. The Schirmer test was performed without topical anesthesia using a standardized filter strip (Bio-Tech Vision Care). The amount of wetting was measured after 5 minutes. Meibomian gland expressibility was assessed by applying digital pressure to the eyelid margin.



Statistical Analysis
Statistical analysis of the study was performed with SPSS Statistics Version 22.0 (SPSS Inc). Continuous variables were reported as mean (SD), and categorical variables were reported as percentages and counts. Descriptive statistics for numerical variables were created. An independent sample t test was used for normally distributed continuous variables. The Kolmogorov-Smirnov test was used to determine normality. The Schirmer test without anesthesia and TBUT values among groups were compared using one-way analysis of variance. The differences were calculated using the multiple comparison Tukey test. P<.05 was considered statistically significant.

Results

Demographic Characteristics of Rosacea Patients
Sixty eyes of 30 newly diagnosed patients with acne rosacea with Demodex infestation and 60 eyes of 30 newly diagnosed patients with acne rosacea without Demodex infestation were enrolled in this study. The mean age (SD) of the 60 patients was 37.63 (10.01) years. The mean TBUT (SD) of the 120 eyes was 6.65 (3.44) seconds, and the mean Schirmer score (SD) was 12.59 (6.71) mm (Table 1).

Meibomian Gland Dysfunction vs Subgroup of Rosacea Patients
Thirty-four (57%) patients had blepharitis, and 18 (30%) patients had meibomitis. Thirty-five (58.3%) patients had ETR, 5 (8.3%) patients had phymatous rosacea, and 20 (33.4%) patients had PPR (Table 2). Of the Demodex-negative patients, 73.3% (22/30) had ETR, 20% (6/30) had PPR, and 6.7% (2/30) had phymatous rosacea. Of the Demodex-positive patients, 43.3% (13/30) had ETR, 46.7% (14/30) had PPR, and 10% (3/30) had phymatous rosacea (Table 3). Papulopustular rosacea was found to be significantly associated with Demodex positivity (P=.003); neither ETR nor phymatous rosacea was found to be significantly associated with Demodex infestation (P=.66 and P=.13, respectively)(Table 3).



There was no statistically significant difference between the Demodex-negative and Demodex-positive groups for mean age (SD)(37.4 [11.54] years vs 37.87 [8.41] years; P=.85), mean TBUT (SD)(6.73 [3.62] seconds vs 6.57 [3.33] seconds; P=.85), and mean Schirmer score (SD)(13.68 [7.23] mm vs 11.5 [6.08] mm; P=.21)(Table 4).



Fifteen (50%) patients (30 eyes) in the Demodex-negative group and 19 (63.3%) patients (38 eyes) in the Demodex-positive group had blepharitis, with no statistically significant difference between the groups (P=.43). Seven (23.3%) patients (14 eyes) in the Demodex-negative group and 11 (36.7%) patients (22 eyes) in the Demodex-positive group had meibomitis, with no statistically significant difference between the groups (P=.39)(Table 3).

 

 



Sixteen (53.3%) patients (32 eyes) in the Demodex-negative group and 21 (70%) patients (42 eyes) in the Demodex-positive group had TBUT values less than 10 seconds. Eighteen (60%) patients (36 eyes) in the Demodex-negative group and 25 (83.3%) patients (50 eyes) in the Demodex-positive group had Schirmer scores less than 10 mm (Table 3). The 2 groups were not significantly different in dry eye findings (P=.25 and P=.29, respectively).

Comment

Inflammation in Rosacea
It is known that the density of nonfloral bacteria as well as D folliculorum and Demodex brevis increases in skin affected by rosacea compared to normal skin. Vascular dilation associated with rosacea that results from sunlight and heat causes increased capillary permeability and creates the ideal environment for the proliferation of D folliculorum. Demodex is thought to act as a vector for the activity of certain other microorganisms, particularly Bacillus oleronius, and thus initiates the inflammatory response associated with rosacea.9

One study reported that the inflammation associated with rosacea that was caused by Demodex and other environmental stimuli occurred through toll-like receptor 2 and various cytokines.10 It has been reported that the abnormal function of toll-like receptor 2 in the epidermis leads to the increased production of cathelicidin. Cathelicidin is an antimicrobial peptide with both vasoactive and proinflammatory activity and has been used as a basis to explain the pathogenesis of facial erythema, flushing, and telangiectasia in the context of rosacea.11,12 In addition, it has been reported that the increased secretion of proinflammatory cytokines such as IL-1 and gelatinase B in ocular rosacea leads to tearing film abnormalities that result from increased bacterial flora in the eyelids, which subsequently leads to decreased tear drainage and dry eyes.13 In addition, B oleronius isolated from a D folliculorum mite from patients with PPR produced proteins that induced an inflammatory immune response in 73% (16/22) of patients with rosacea.14

Ocular Findings in Rosacea Patients
In our study, PPR was found to be significantly associated with Demodex positivity compared to ETR and phymatous rosacea (P=.003). However, ocular inflammation findings such as blepharitis and meibomitis were not significantly different between Demodex-positive and Demodex-negative patients. Although the mean Schirmer score of Demodex-positive patients was lower than Demodex-negative patients, this difference was not statistically significant. We evaluated a TBUT of less than 10 seconds and a Schirmer score less than 10 mm as dry eye. Accordingly, the number of patients with dry eye was higher in the Demodex-positive group, but this difference was not statistically significant.



Chronic blepharitis, conjunctival inflammation, and meibomian gland dysfunction are among the most common findings of ocular rosacea.15,16 Patients with ocular rosacea commonly have dry eye and abnormal TBUT and Schirmer scores.17 In our study, we found that the fluorescein TBUT and Schirmer scores were more likely to be abnormal in the Demodex-positive group, but the difference between the 2 groups was not statistically significant.

It has been reported that proinflammatory cytokines due to a weakened immune system in rosacea patients were increased. The weakened immune system was further supported by the increased concentrations of proinflammatory cytokines such as IL-1 and matrix metalloproteinase 9 in these patients’ tears and the improvement of symptoms after the inhibition of these cytokines.11 Luo et al18 reported that Demodex inflammation causes dry eye, particularly with D brevis. Ayyildiz and Sezgin19 reported that Schirmer scores were significantly lower and that the Ocular Surface Disease Index had significantly increased in the Demodex-positive group compared to the Demodex-negative group (P=.001 for both). A Korean study reported that Demodex density was correlated with age, sex, and TBUT results, but there was no significant relationship between Demodex density and Schirmer scores.16

Sobolewska et al20 administered ivermectin cream 1% to 10 patients with cutaneous and ocular rosacea, but only to the forehead, chin, nose, cheeks, and regions close to the eyelids, and observed a significant improvement in blepharitis (P=.004). They stated that ivermectin, as applied only to the face, suppressed the proinflammatory cytokines associated with rosacea and showed anti-inflammatory effects by reducing Demodex mites.20Li et al21 demonstrated a strong correlation between ocular Demodex inflammation and serum reactivity to these bacterial proteins in patients with ocular rosacea, and they found that eyelid margin inflammation and facial rosacea correlated with reactivity to these proteins. These studies suggest a possible role for Demodex infestation and bacterial proteins in the etiology of rosacea.

Gonzalez-Hinojosa et al22 demonstrated that even though eyelash blepharitis was more common in PPR than ETR, there was no statistically significant association between rosacea and Demodex blepharitis. In our study, we found a significant correlation between PPR and Demodex positivity. Also, meibomian gland dysfunction was more common in the Demodex-positive group; however, this result was not statistically significant. One study compared patients with primary demodicosis and patients with rosacea with Demodex-induced blepharitis to healthy controls and found that patients with primary demodicosis and patients with rosacea did not have significantly different ocular findings.23 In contrast, Forton and De Maertelaer24 reported that patients with PPR had significantly more severe ocular manifestations compared with patients with demodicosis (P=.004).

Mizuno et al25 compared the normal (nonrosacea) population with and without Demodex-infested eyelashes and found that the 2 groups were not significantly different for meibomian gland dysfunction, fluorescein TBUT, or ocular surface discomfort.

Varying results have been reported regarding the association between Demodex and blepharitis or ocular surface discomfort with or without rosacea. In our study, we found that Demodex did not affect tear function tests or meibomian gland function in patients with rosacea. We believe this study is important because it demonstrates the effects of Demodex on ocular findings in patients with cutaneous rosacea.

Limitations
Our study has some limitations. The number of patients was relatively small, resulting in few significant differences between the comparison groups. A larger prospective research study is required to assess the prevalence of Demodex mites in the ocular rosacea population along with associated symptoms and findings.

Conclusion

Rosacea is a chronic disease associated with skin and ocular manifestations that range from mild to severe, that progresses in the form of attacks, and that requires long-term follow-up and treatment. Rosacea most often presents as a disease that causes ocular surface inflammation of varying degrees. Demodex infestation may increase cutaneous or ocular inflammation in rosacea. Therefore, every patient diagnosed with rosacea should be given a dermatologic examination to determine Demodex positivity and an ophthalmologic examination to determine ocular manifestations.

References
  1. O’Reilly N, Gallagher C, Reddy Katikireddy K, et al. Demodex-associated Bacillus proteins induce an aberrant wound healing response in a corneal epithelial cell line: possible implications for corneal ulcer formation in ocular rosacea. Invest Ophthalmol Vis Sci. 2012;53:3250-3259.
  2. Webster G, Schaller M. Ocular rosacea: a dermatologic perspective. J Am Acad Dermatol. 2013;69(6 suppl 1):S42-S43.
  3. Crawford GH, Pelle MT, James WD. Rosacea: I. etiology, pathogenesis, and subtype classification. J Am Acad Dermatol. 2004;51:327-341.
  4. Wilkin J, Dahl M, Detmar M, et al. Standard grading system for rosacea: report of the National Rosacea Society Expert Committee on the classification and staging of rosacea. J Am Acad Dermatol. 2004;50:907-912.
  5. Gallo RLGranstein RDKang S, et al. Standard classification and pathophysiology of rosacea: the 2017 update by the National Rosacea Society Expert Committee. J Am Acad Dermatol. 2018;78:148-155.
  6. Gao YY, Di Pascuale MA, Li W, et al. High prevalence of Demodex in eyelashes with cylindrical dandruff. Invest Ophthalmol Vis Sci. 2005;46:3089-3094.
  7. Fallen RS, Gooderham M. Rosacea: update on management and emerging therapies. Skin Therapy Lett. 2012;17:1-4.
  8. Erbagcı Z, Ozgoztası O. The significance of Demodex folliculorum density in rosacea. Int J Dermatol. 1998;37:421-425.
  9. Ahn CS, Huang WW. Rosacea pathogenesis. Dermatol Clin. 2018;36:81‐86.
  10. Forton FMN, De Maertelaer V. Two consecutive standardized skin surface biopsies: an improved sampling method to evaluate Demodex density as a diagnostic tool for rosacea and demodicosis. Acta Derm Venereol. 2017;97:242‐248.
  11. Yamasaki K, Kanada K, Macleod DT, et al. TLR2 expression is increased in rosacea and stimulates enhanced serine protease production by keratinocytes. J Invest Dermatol. 2011;131:688-697.
  12. Gold LM, Draelos ZD. New and emerging treatments for rosacea. Am J Clin Dermatol. 2015;16:457-461.
  13. Two AM, Del Rosso JQ. Kallikrein 5-mediated inflammation in rosacea: clinically relevant correlations with acute and chronic manifestations in rosacea and how individual treatments may provide therapeutic benefit. J Clin Aesthet Dermatol. 2014;7:20-25.
  14. Lacey N, Delaney S, Kavanagh K, et al. Mite-related bacterial antigens stimulate inflammatory cells in rosacea. Br J Dermatol. 2007;157:474-481.
  15. Forton F, Germaux MA, Brasseur T, et al. Demodicosis and rosacea: epidemiology and significance in daily dermatologic practice. J Am Acad Dermatol. 2005;52:74-87.
  16. Lee SH, Chun YS, Kim JH, et al. The relationship between Demodex and ocular discomfort. Invest Ophthalmol Vis Sci. 2010;51:2906-2911.
  17. Awais M, Anwar MI, Ilfikhar R, et al. Rosacea—the ophthalmic perspective. Cutan Ocul Toxicol. 2015;34:161-166.
  18. Luo X, Li J, Chen C, et al. Ocular demodicosis as a potential cause of ocular surface inflammation. Cornea. 2017;36(suppl 1):S9-S14.
  19. Ayyildiz T, Sezgin FM. The effect of ocular Demodex colonization on Schirmer test and OSDI scores in newly diagnosed dry eye patients. Eye Contact Lens. 2020;46(suppl 1):S39-S41.
  20. Sobolewska B, Doycheva D, Deuter CM, et al. Efficacy of topical ivermectin for the treatment of cutaneous and ocular rosacea [published online April 7, 2020]. Ocul Immunol Inflamm. doi:10.1080/09273948.2020.1727531
  21. Li J, O‘Reilly N, Sheha H, et al. Correlation between ocular Demodex infestation and serum immunoreactivity to Bacillus proteins in patients with facial rosacea. 2010;117:870-877.
  22. Gonzalez‐Hinojosa D, Jaime‐Villalonga A, Aguilar‐Montes G, et al. Demodex and rosacea: is there a relationship? Indian J Ophthalmol. 2018;66:36‐38.
  23. Sarac G, Cankaya C, Ozcan KN, et al. Increased frequency of Demodex blepharitis in rosacea and facial demodicosis patients. J Cosmet Dermatol. 2020;19:1260-1265.
  24. Forton FMN, De Maertelaer V. Rosacea and demodicosis: little-known diagnostic signs and symptoms. Acta Derm Venereol. 2019;99:47-52.
  25. Mizuno M, Kawashima M, Uchino M, et al. Demodex-mite infestation in cilia and its association with ocular surface parameters in Japanese volunteers. Eye Contact Lens. 2020;46:291-296.
Article PDF
Author and Disclosure Information

Drs. Kara and Çalis¸ are from Koru Hospital, Ankara, Turkey. Dr. Kara is from the Department of Dermatology, and Dr. Çalis¸ is from the Department of Ophthalmology. Dr. Gürel is from the Department of Ophthalmology, 29 Mayıs State Hospital, Ankara.

The authors report no conflict of interest.

Correspondence: Yesim Akpinar Kara, MD, 1428.sk No:16/8, Cukurambar, Ankara, Turkey 06520 (yesim_akpinar@yahoo.com).

Issue
cutis - 108(1)
Publications
Topics
Page Number
46-50
Sections
Author and Disclosure Information

Drs. Kara and Çalis¸ are from Koru Hospital, Ankara, Turkey. Dr. Kara is from the Department of Dermatology, and Dr. Çalis¸ is from the Department of Ophthalmology. Dr. Gürel is from the Department of Ophthalmology, 29 Mayıs State Hospital, Ankara.

The authors report no conflict of interest.

Correspondence: Yesim Akpinar Kara, MD, 1428.sk No:16/8, Cukurambar, Ankara, Turkey 06520 (yesim_akpinar@yahoo.com).

Author and Disclosure Information

Drs. Kara and Çalis¸ are from Koru Hospital, Ankara, Turkey. Dr. Kara is from the Department of Dermatology, and Dr. Çalis¸ is from the Department of Ophthalmology. Dr. Gürel is from the Department of Ophthalmology, 29 Mayıs State Hospital, Ankara.

The authors report no conflict of interest.

Correspondence: Yesim Akpinar Kara, MD, 1428.sk No:16/8, Cukurambar, Ankara, Turkey 06520 (yesim_akpinar@yahoo.com).

Article PDF
Article PDF

Acne rosacea is a chronic inflammatory disease that may affect the facial skin, eyes, and eyelids.1 It is characterized by transient or persistent flushing, facial erythema, and telangiectases, generally located on the central portion of the face, and may progress to papules and pustules.2,3 At the late stage of the disease, dermal edema or fibroplasia and sebaceous gland hypertrophy may cause phymatous alterations in the skin. In 2004, the National Rosacea Society Expert Committee developed a classification system for rosacea to standardize subtypes and variants that has since been widely accepted and continues to aid in research and epidemiologic studies.4 The committee defined 4 subtypes based on clinical characteristics: erythematotelangiectatic (ETR), papulopustular (PPR), phymatous, and ocular rosacea.2,3

Ocular rosacea may accompany mild, moderate, and severe dermatologic disease or may occur in the absence of diagnostic skin disease.5 Ocular signs include eyelid margin telangiectasia, spade-shaped infiltrates in the cornea, scleritis, and sclerokeratitis. Common symptoms include burning, stinging, light sensitivity, and foreign-body sensation. Ocular signs commonly seen in rosacea are meibomian gland dysfunction characterized by inspissation and inflammation of the meibomian glands (chalazia), conjunctivitis, honey crust and cylindrical collarette accumulation at the base of the eyelashes, irregularity of the eyelid margin architecture, and evaporative tear dysfunction.5,6

The physiopathology of rosacea is still unknown. Potential factors include genetic predisposition, abnormal inflammation, vascular dysfunction, and involvement of several microbial agents, such as commensal Demodex mites. The number of Demodex mites on normal skin flora is less than 5/cm2; however, the increased vascular dilation and capillary permeability associated with rosacea that result from sunlight and heat exposure increase the density of Demodex folliculorum.7 Elevated Demodex mite density has been observed in the lumens of the sebaceous follicles in patients with rosacea. However, because the severity of the clinical manifestations of the disease is not directly associated with the density of D folliculorum, it generally is accepted that D folliculorum is not a pathogenetic but rather an exacerbating factor.8 It has been reported that this species of mite is mostly found on the face and around the eyelashes and scalp of patients and that it can cause ocular surface inflammation.8

Most studies have researched ocular manifestations of rosacea but not ocular involvement in rosacea patients with and without Demodex mite infestation. In our study, we sought to compare the ocular surface, meibomian gland characteristics, and tear film abnormalities among patients with cutaneous rosacea with and without Demodex infestation.

Materials and Methods

We conducted a retrospective study of 60 patients with cutaneous rosacea. This study was approved by the ethics committee of the local hospital (2018/002-003), and all patients provided verbal and written informed consent before participating in the study. The study was carried out according to the guidelines of the Declaration of Helsinki.

Patient Selection and Evaluation
Patients diagnosed with rosacea by a dermatologist within 6 months were included in the study. Diagnosis of the disease was made after a detailed anamnesis and dermatologic examination. Rosacea was diagnosed if patients had an itching sensation, erythema and/or erythema attacks, and papules and pustules, and fulfilled the diagnostic criteria according to the National Rosacea Society. The skin disease was classified according to the subtypes as ETR, PPR, phymatous rosacea, or ocular rosacea.



The standard skin surface biopsy method was used in 60 patients for detecting Demodex density. When more than 5 mites were detected per square centimeter, the result was recorded as positive. Thirty consecutive, newly diagnosed patients with cutaneous acne rosacea with Demodex infestation and 30 consecutive, newly diagnosed sex- and age-matched patients with acne rosacea without Demodex infestation admitted to the dermatology outpatient clinic were included to this study. The patients who did not have any known dermatologic, systemic, or ocular diseases were included in the study. Patients who met any of the following criteria were excluded from the study: prior anti-inflammatory topical and/or systemic treatment for rosacea during the last 3 months, contact lens wear, eyelid surgery, or autoimmune disease requiring treatment.

 

 



Microscopic Demodex Examination
Demodex count was determined using a standardized skin surface biopsy, which is a noninvasive method. Every patient gave samples from the cheeks. This biopsy was repeated from the same site. A drop of cyanoacrylate was placed on a clean slide, pressed against a skin lesion, held in place for 1 minute, and removed. The obtained samples were evaluated under a light microscope (Nikon E200) with oil immersion. When more than 5 mites were detected per square centimeter, the result was recorded as positive.

Ophthalmologic Examination
A complete ophthalmologic examination including visual acuity assessment, standardized slit lamp examination, and fundus examination was done for all patients. Ocular rosacea was diagnosed on detection of 1 or more of the following: watery or bloodshot appearance, foreign-body sensation, burning or stinging, dryness, itching, light sensitivity, blurred vision, telangiectases of the conjunctiva and eyelid margin, eyelid lid and periocular erythema, anterior blepharitis, meibomian gland dysfunction, or irregularity of eyelid margins. All patients were screened for the signs and symptoms of ocular rosacea and underwent other ophthalmologic examinations, including tear function tests. Tear functions were evaluated with Schirmer tests without anesthesia and fluorescein tear breakup time (TBUT). Tear film breakup time was assessed after instillation of 2% fluorescein staining under a cobalt blue filter. The time interval between the last complete blink and the appearance of the first dry spot was recorded. The mean of 3 consecutive measurements was obtained. The Schirmer test was performed without topical anesthesia using a standardized filter strip (Bio-Tech Vision Care). The amount of wetting was measured after 5 minutes. Meibomian gland expressibility was assessed by applying digital pressure to the eyelid margin.



Statistical Analysis
Statistical analysis of the study was performed with SPSS Statistics Version 22.0 (SPSS Inc). Continuous variables were reported as mean (SD), and categorical variables were reported as percentages and counts. Descriptive statistics for numerical variables were created. An independent sample t test was used for normally distributed continuous variables. The Kolmogorov-Smirnov test was used to determine normality. The Schirmer test without anesthesia and TBUT values among groups were compared using one-way analysis of variance. The differences were calculated using the multiple comparison Tukey test. P<.05 was considered statistically significant.

Results

Demographic Characteristics of Rosacea Patients
Sixty eyes of 30 newly diagnosed patients with acne rosacea with Demodex infestation and 60 eyes of 30 newly diagnosed patients with acne rosacea without Demodex infestation were enrolled in this study. The mean age (SD) of the 60 patients was 37.63 (10.01) years. The mean TBUT (SD) of the 120 eyes was 6.65 (3.44) seconds, and the mean Schirmer score (SD) was 12.59 (6.71) mm (Table 1).

Meibomian Gland Dysfunction vs Subgroup of Rosacea Patients
Thirty-four (57%) patients had blepharitis, and 18 (30%) patients had meibomitis. Thirty-five (58.3%) patients had ETR, 5 (8.3%) patients had phymatous rosacea, and 20 (33.4%) patients had PPR (Table 2). Of the Demodex-negative patients, 73.3% (22/30) had ETR, 20% (6/30) had PPR, and 6.7% (2/30) had phymatous rosacea. Of the Demodex-positive patients, 43.3% (13/30) had ETR, 46.7% (14/30) had PPR, and 10% (3/30) had phymatous rosacea (Table 3). Papulopustular rosacea was found to be significantly associated with Demodex positivity (P=.003); neither ETR nor phymatous rosacea was found to be significantly associated with Demodex infestation (P=.66 and P=.13, respectively)(Table 3).



There was no statistically significant difference between the Demodex-negative and Demodex-positive groups for mean age (SD)(37.4 [11.54] years vs 37.87 [8.41] years; P=.85), mean TBUT (SD)(6.73 [3.62] seconds vs 6.57 [3.33] seconds; P=.85), and mean Schirmer score (SD)(13.68 [7.23] mm vs 11.5 [6.08] mm; P=.21)(Table 4).



Fifteen (50%) patients (30 eyes) in the Demodex-negative group and 19 (63.3%) patients (38 eyes) in the Demodex-positive group had blepharitis, with no statistically significant difference between the groups (P=.43). Seven (23.3%) patients (14 eyes) in the Demodex-negative group and 11 (36.7%) patients (22 eyes) in the Demodex-positive group had meibomitis, with no statistically significant difference between the groups (P=.39)(Table 3).

 

 



Sixteen (53.3%) patients (32 eyes) in the Demodex-negative group and 21 (70%) patients (42 eyes) in the Demodex-positive group had TBUT values less than 10 seconds. Eighteen (60%) patients (36 eyes) in the Demodex-negative group and 25 (83.3%) patients (50 eyes) in the Demodex-positive group had Schirmer scores less than 10 mm (Table 3). The 2 groups were not significantly different in dry eye findings (P=.25 and P=.29, respectively).

Comment

Inflammation in Rosacea
It is known that the density of nonfloral bacteria as well as D folliculorum and Demodex brevis increases in skin affected by rosacea compared to normal skin. Vascular dilation associated with rosacea that results from sunlight and heat causes increased capillary permeability and creates the ideal environment for the proliferation of D folliculorum. Demodex is thought to act as a vector for the activity of certain other microorganisms, particularly Bacillus oleronius, and thus initiates the inflammatory response associated with rosacea.9

One study reported that the inflammation associated with rosacea that was caused by Demodex and other environmental stimuli occurred through toll-like receptor 2 and various cytokines.10 It has been reported that the abnormal function of toll-like receptor 2 in the epidermis leads to the increased production of cathelicidin. Cathelicidin is an antimicrobial peptide with both vasoactive and proinflammatory activity and has been used as a basis to explain the pathogenesis of facial erythema, flushing, and telangiectasia in the context of rosacea.11,12 In addition, it has been reported that the increased secretion of proinflammatory cytokines such as IL-1 and gelatinase B in ocular rosacea leads to tearing film abnormalities that result from increased bacterial flora in the eyelids, which subsequently leads to decreased tear drainage and dry eyes.13 In addition, B oleronius isolated from a D folliculorum mite from patients with PPR produced proteins that induced an inflammatory immune response in 73% (16/22) of patients with rosacea.14

Ocular Findings in Rosacea Patients
In our study, PPR was found to be significantly associated with Demodex positivity compared to ETR and phymatous rosacea (P=.003). However, ocular inflammation findings such as blepharitis and meibomitis were not significantly different between Demodex-positive and Demodex-negative patients. Although the mean Schirmer score of Demodex-positive patients was lower than Demodex-negative patients, this difference was not statistically significant. We evaluated a TBUT of less than 10 seconds and a Schirmer score less than 10 mm as dry eye. Accordingly, the number of patients with dry eye was higher in the Demodex-positive group, but this difference was not statistically significant.



Chronic blepharitis, conjunctival inflammation, and meibomian gland dysfunction are among the most common findings of ocular rosacea.15,16 Patients with ocular rosacea commonly have dry eye and abnormal TBUT and Schirmer scores.17 In our study, we found that the fluorescein TBUT and Schirmer scores were more likely to be abnormal in the Demodex-positive group, but the difference between the 2 groups was not statistically significant.

It has been reported that proinflammatory cytokines due to a weakened immune system in rosacea patients were increased. The weakened immune system was further supported by the increased concentrations of proinflammatory cytokines such as IL-1 and matrix metalloproteinase 9 in these patients’ tears and the improvement of symptoms after the inhibition of these cytokines.11 Luo et al18 reported that Demodex inflammation causes dry eye, particularly with D brevis. Ayyildiz and Sezgin19 reported that Schirmer scores were significantly lower and that the Ocular Surface Disease Index had significantly increased in the Demodex-positive group compared to the Demodex-negative group (P=.001 for both). A Korean study reported that Demodex density was correlated with age, sex, and TBUT results, but there was no significant relationship between Demodex density and Schirmer scores.16

Sobolewska et al20 administered ivermectin cream 1% to 10 patients with cutaneous and ocular rosacea, but only to the forehead, chin, nose, cheeks, and regions close to the eyelids, and observed a significant improvement in blepharitis (P=.004). They stated that ivermectin, as applied only to the face, suppressed the proinflammatory cytokines associated with rosacea and showed anti-inflammatory effects by reducing Demodex mites.20Li et al21 demonstrated a strong correlation between ocular Demodex inflammation and serum reactivity to these bacterial proteins in patients with ocular rosacea, and they found that eyelid margin inflammation and facial rosacea correlated with reactivity to these proteins. These studies suggest a possible role for Demodex infestation and bacterial proteins in the etiology of rosacea.

Gonzalez-Hinojosa et al22 demonstrated that even though eyelash blepharitis was more common in PPR than ETR, there was no statistically significant association between rosacea and Demodex blepharitis. In our study, we found a significant correlation between PPR and Demodex positivity. Also, meibomian gland dysfunction was more common in the Demodex-positive group; however, this result was not statistically significant. One study compared patients with primary demodicosis and patients with rosacea with Demodex-induced blepharitis to healthy controls and found that patients with primary demodicosis and patients with rosacea did not have significantly different ocular findings.23 In contrast, Forton and De Maertelaer24 reported that patients with PPR had significantly more severe ocular manifestations compared with patients with demodicosis (P=.004).

Mizuno et al25 compared the normal (nonrosacea) population with and without Demodex-infested eyelashes and found that the 2 groups were not significantly different for meibomian gland dysfunction, fluorescein TBUT, or ocular surface discomfort.

Varying results have been reported regarding the association between Demodex and blepharitis or ocular surface discomfort with or without rosacea. In our study, we found that Demodex did not affect tear function tests or meibomian gland function in patients with rosacea. We believe this study is important because it demonstrates the effects of Demodex on ocular findings in patients with cutaneous rosacea.

Limitations
Our study has some limitations. The number of patients was relatively small, resulting in few significant differences between the comparison groups. A larger prospective research study is required to assess the prevalence of Demodex mites in the ocular rosacea population along with associated symptoms and findings.

Conclusion

Rosacea is a chronic disease associated with skin and ocular manifestations that range from mild to severe, that progresses in the form of attacks, and that requires long-term follow-up and treatment. Rosacea most often presents as a disease that causes ocular surface inflammation of varying degrees. Demodex infestation may increase cutaneous or ocular inflammation in rosacea. Therefore, every patient diagnosed with rosacea should be given a dermatologic examination to determine Demodex positivity and an ophthalmologic examination to determine ocular manifestations.

Acne rosacea is a chronic inflammatory disease that may affect the facial skin, eyes, and eyelids.1 It is characterized by transient or persistent flushing, facial erythema, and telangiectases, generally located on the central portion of the face, and may progress to papules and pustules.2,3 At the late stage of the disease, dermal edema or fibroplasia and sebaceous gland hypertrophy may cause phymatous alterations in the skin. In 2004, the National Rosacea Society Expert Committee developed a classification system for rosacea to standardize subtypes and variants that has since been widely accepted and continues to aid in research and epidemiologic studies.4 The committee defined 4 subtypes based on clinical characteristics: erythematotelangiectatic (ETR), papulopustular (PPR), phymatous, and ocular rosacea.2,3

Ocular rosacea may accompany mild, moderate, and severe dermatologic disease or may occur in the absence of diagnostic skin disease.5 Ocular signs include eyelid margin telangiectasia, spade-shaped infiltrates in the cornea, scleritis, and sclerokeratitis. Common symptoms include burning, stinging, light sensitivity, and foreign-body sensation. Ocular signs commonly seen in rosacea are meibomian gland dysfunction characterized by inspissation and inflammation of the meibomian glands (chalazia), conjunctivitis, honey crust and cylindrical collarette accumulation at the base of the eyelashes, irregularity of the eyelid margin architecture, and evaporative tear dysfunction.5,6

The physiopathology of rosacea is still unknown. Potential factors include genetic predisposition, abnormal inflammation, vascular dysfunction, and involvement of several microbial agents, such as commensal Demodex mites. The number of Demodex mites on normal skin flora is less than 5/cm2; however, the increased vascular dilation and capillary permeability associated with rosacea that result from sunlight and heat exposure increase the density of Demodex folliculorum.7 Elevated Demodex mite density has been observed in the lumens of the sebaceous follicles in patients with rosacea. However, because the severity of the clinical manifestations of the disease is not directly associated with the density of D folliculorum, it generally is accepted that D folliculorum is not a pathogenetic but rather an exacerbating factor.8 It has been reported that this species of mite is mostly found on the face and around the eyelashes and scalp of patients and that it can cause ocular surface inflammation.8

Most studies have researched ocular manifestations of rosacea but not ocular involvement in rosacea patients with and without Demodex mite infestation. In our study, we sought to compare the ocular surface, meibomian gland characteristics, and tear film abnormalities among patients with cutaneous rosacea with and without Demodex infestation.

Materials and Methods

We conducted a retrospective study of 60 patients with cutaneous rosacea. This study was approved by the ethics committee of the local hospital (2018/002-003), and all patients provided verbal and written informed consent before participating in the study. The study was carried out according to the guidelines of the Declaration of Helsinki.

Patient Selection and Evaluation
Patients diagnosed with rosacea by a dermatologist within 6 months were included in the study. Diagnosis of the disease was made after a detailed anamnesis and dermatologic examination. Rosacea was diagnosed if patients had an itching sensation, erythema and/or erythema attacks, and papules and pustules, and fulfilled the diagnostic criteria according to the National Rosacea Society. The skin disease was classified according to the subtypes as ETR, PPR, phymatous rosacea, or ocular rosacea.



The standard skin surface biopsy method was used in 60 patients for detecting Demodex density. When more than 5 mites were detected per square centimeter, the result was recorded as positive. Thirty consecutive, newly diagnosed patients with cutaneous acne rosacea with Demodex infestation and 30 consecutive, newly diagnosed sex- and age-matched patients with acne rosacea without Demodex infestation admitted to the dermatology outpatient clinic were included to this study. The patients who did not have any known dermatologic, systemic, or ocular diseases were included in the study. Patients who met any of the following criteria were excluded from the study: prior anti-inflammatory topical and/or systemic treatment for rosacea during the last 3 months, contact lens wear, eyelid surgery, or autoimmune disease requiring treatment.

 

 



Microscopic Demodex Examination
Demodex count was determined using a standardized skin surface biopsy, which is a noninvasive method. Every patient gave samples from the cheeks. This biopsy was repeated from the same site. A drop of cyanoacrylate was placed on a clean slide, pressed against a skin lesion, held in place for 1 minute, and removed. The obtained samples were evaluated under a light microscope (Nikon E200) with oil immersion. When more than 5 mites were detected per square centimeter, the result was recorded as positive.

Ophthalmologic Examination
A complete ophthalmologic examination including visual acuity assessment, standardized slit lamp examination, and fundus examination was done for all patients. Ocular rosacea was diagnosed on detection of 1 or more of the following: watery or bloodshot appearance, foreign-body sensation, burning or stinging, dryness, itching, light sensitivity, blurred vision, telangiectases of the conjunctiva and eyelid margin, eyelid lid and periocular erythema, anterior blepharitis, meibomian gland dysfunction, or irregularity of eyelid margins. All patients were screened for the signs and symptoms of ocular rosacea and underwent other ophthalmologic examinations, including tear function tests. Tear functions were evaluated with Schirmer tests without anesthesia and fluorescein tear breakup time (TBUT). Tear film breakup time was assessed after instillation of 2% fluorescein staining under a cobalt blue filter. The time interval between the last complete blink and the appearance of the first dry spot was recorded. The mean of 3 consecutive measurements was obtained. The Schirmer test was performed without topical anesthesia using a standardized filter strip (Bio-Tech Vision Care). The amount of wetting was measured after 5 minutes. Meibomian gland expressibility was assessed by applying digital pressure to the eyelid margin.



Statistical Analysis
Statistical analysis of the study was performed with SPSS Statistics Version 22.0 (SPSS Inc). Continuous variables were reported as mean (SD), and categorical variables were reported as percentages and counts. Descriptive statistics for numerical variables were created. An independent sample t test was used for normally distributed continuous variables. The Kolmogorov-Smirnov test was used to determine normality. The Schirmer test without anesthesia and TBUT values among groups were compared using one-way analysis of variance. The differences were calculated using the multiple comparison Tukey test. P<.05 was considered statistically significant.

Results

Demographic Characteristics of Rosacea Patients
Sixty eyes of 30 newly diagnosed patients with acne rosacea with Demodex infestation and 60 eyes of 30 newly diagnosed patients with acne rosacea without Demodex infestation were enrolled in this study. The mean age (SD) of the 60 patients was 37.63 (10.01) years. The mean TBUT (SD) of the 120 eyes was 6.65 (3.44) seconds, and the mean Schirmer score (SD) was 12.59 (6.71) mm (Table 1).

Meibomian Gland Dysfunction vs Subgroup of Rosacea Patients
Thirty-four (57%) patients had blepharitis, and 18 (30%) patients had meibomitis. Thirty-five (58.3%) patients had ETR, 5 (8.3%) patients had phymatous rosacea, and 20 (33.4%) patients had PPR (Table 2). Of the Demodex-negative patients, 73.3% (22/30) had ETR, 20% (6/30) had PPR, and 6.7% (2/30) had phymatous rosacea. Of the Demodex-positive patients, 43.3% (13/30) had ETR, 46.7% (14/30) had PPR, and 10% (3/30) had phymatous rosacea (Table 3). Papulopustular rosacea was found to be significantly associated with Demodex positivity (P=.003); neither ETR nor phymatous rosacea was found to be significantly associated with Demodex infestation (P=.66 and P=.13, respectively)(Table 3).



There was no statistically significant difference between the Demodex-negative and Demodex-positive groups for mean age (SD)(37.4 [11.54] years vs 37.87 [8.41] years; P=.85), mean TBUT (SD)(6.73 [3.62] seconds vs 6.57 [3.33] seconds; P=.85), and mean Schirmer score (SD)(13.68 [7.23] mm vs 11.5 [6.08] mm; P=.21)(Table 4).



Fifteen (50%) patients (30 eyes) in the Demodex-negative group and 19 (63.3%) patients (38 eyes) in the Demodex-positive group had blepharitis, with no statistically significant difference between the groups (P=.43). Seven (23.3%) patients (14 eyes) in the Demodex-negative group and 11 (36.7%) patients (22 eyes) in the Demodex-positive group had meibomitis, with no statistically significant difference between the groups (P=.39)(Table 3).

 

 



Sixteen (53.3%) patients (32 eyes) in the Demodex-negative group and 21 (70%) patients (42 eyes) in the Demodex-positive group had TBUT values less than 10 seconds. Eighteen (60%) patients (36 eyes) in the Demodex-negative group and 25 (83.3%) patients (50 eyes) in the Demodex-positive group had Schirmer scores less than 10 mm (Table 3). The 2 groups were not significantly different in dry eye findings (P=.25 and P=.29, respectively).

Comment

Inflammation in Rosacea
It is known that the density of nonfloral bacteria as well as D folliculorum and Demodex brevis increases in skin affected by rosacea compared to normal skin. Vascular dilation associated with rosacea that results from sunlight and heat causes increased capillary permeability and creates the ideal environment for the proliferation of D folliculorum. Demodex is thought to act as a vector for the activity of certain other microorganisms, particularly Bacillus oleronius, and thus initiates the inflammatory response associated with rosacea.9

One study reported that the inflammation associated with rosacea that was caused by Demodex and other environmental stimuli occurred through toll-like receptor 2 and various cytokines.10 It has been reported that the abnormal function of toll-like receptor 2 in the epidermis leads to the increased production of cathelicidin. Cathelicidin is an antimicrobial peptide with both vasoactive and proinflammatory activity and has been used as a basis to explain the pathogenesis of facial erythema, flushing, and telangiectasia in the context of rosacea.11,12 In addition, it has been reported that the increased secretion of proinflammatory cytokines such as IL-1 and gelatinase B in ocular rosacea leads to tearing film abnormalities that result from increased bacterial flora in the eyelids, which subsequently leads to decreased tear drainage and dry eyes.13 In addition, B oleronius isolated from a D folliculorum mite from patients with PPR produced proteins that induced an inflammatory immune response in 73% (16/22) of patients with rosacea.14

Ocular Findings in Rosacea Patients
In our study, PPR was found to be significantly associated with Demodex positivity compared to ETR and phymatous rosacea (P=.003). However, ocular inflammation findings such as blepharitis and meibomitis were not significantly different between Demodex-positive and Demodex-negative patients. Although the mean Schirmer score of Demodex-positive patients was lower than Demodex-negative patients, this difference was not statistically significant. We evaluated a TBUT of less than 10 seconds and a Schirmer score less than 10 mm as dry eye. Accordingly, the number of patients with dry eye was higher in the Demodex-positive group, but this difference was not statistically significant.



Chronic blepharitis, conjunctival inflammation, and meibomian gland dysfunction are among the most common findings of ocular rosacea.15,16 Patients with ocular rosacea commonly have dry eye and abnormal TBUT and Schirmer scores.17 In our study, we found that the fluorescein TBUT and Schirmer scores were more likely to be abnormal in the Demodex-positive group, but the difference between the 2 groups was not statistically significant.

It has been reported that proinflammatory cytokines due to a weakened immune system in rosacea patients were increased. The weakened immune system was further supported by the increased concentrations of proinflammatory cytokines such as IL-1 and matrix metalloproteinase 9 in these patients’ tears and the improvement of symptoms after the inhibition of these cytokines.11 Luo et al18 reported that Demodex inflammation causes dry eye, particularly with D brevis. Ayyildiz and Sezgin19 reported that Schirmer scores were significantly lower and that the Ocular Surface Disease Index had significantly increased in the Demodex-positive group compared to the Demodex-negative group (P=.001 for both). A Korean study reported that Demodex density was correlated with age, sex, and TBUT results, but there was no significant relationship between Demodex density and Schirmer scores.16

Sobolewska et al20 administered ivermectin cream 1% to 10 patients with cutaneous and ocular rosacea, but only to the forehead, chin, nose, cheeks, and regions close to the eyelids, and observed a significant improvement in blepharitis (P=.004). They stated that ivermectin, as applied only to the face, suppressed the proinflammatory cytokines associated with rosacea and showed anti-inflammatory effects by reducing Demodex mites.20Li et al21 demonstrated a strong correlation between ocular Demodex inflammation and serum reactivity to these bacterial proteins in patients with ocular rosacea, and they found that eyelid margin inflammation and facial rosacea correlated with reactivity to these proteins. These studies suggest a possible role for Demodex infestation and bacterial proteins in the etiology of rosacea.

Gonzalez-Hinojosa et al22 demonstrated that even though eyelash blepharitis was more common in PPR than ETR, there was no statistically significant association between rosacea and Demodex blepharitis. In our study, we found a significant correlation between PPR and Demodex positivity. Also, meibomian gland dysfunction was more common in the Demodex-positive group; however, this result was not statistically significant. One study compared patients with primary demodicosis and patients with rosacea with Demodex-induced blepharitis to healthy controls and found that patients with primary demodicosis and patients with rosacea did not have significantly different ocular findings.23 In contrast, Forton and De Maertelaer24 reported that patients with PPR had significantly more severe ocular manifestations compared with patients with demodicosis (P=.004).

Mizuno et al25 compared the normal (nonrosacea) population with and without Demodex-infested eyelashes and found that the 2 groups were not significantly different for meibomian gland dysfunction, fluorescein TBUT, or ocular surface discomfort.

Varying results have been reported regarding the association between Demodex and blepharitis or ocular surface discomfort with or without rosacea. In our study, we found that Demodex did not affect tear function tests or meibomian gland function in patients with rosacea. We believe this study is important because it demonstrates the effects of Demodex on ocular findings in patients with cutaneous rosacea.

Limitations
Our study has some limitations. The number of patients was relatively small, resulting in few significant differences between the comparison groups. A larger prospective research study is required to assess the prevalence of Demodex mites in the ocular rosacea population along with associated symptoms and findings.

Conclusion

Rosacea is a chronic disease associated with skin and ocular manifestations that range from mild to severe, that progresses in the form of attacks, and that requires long-term follow-up and treatment. Rosacea most often presents as a disease that causes ocular surface inflammation of varying degrees. Demodex infestation may increase cutaneous or ocular inflammation in rosacea. Therefore, every patient diagnosed with rosacea should be given a dermatologic examination to determine Demodex positivity and an ophthalmologic examination to determine ocular manifestations.

References
  1. O’Reilly N, Gallagher C, Reddy Katikireddy K, et al. Demodex-associated Bacillus proteins induce an aberrant wound healing response in a corneal epithelial cell line: possible implications for corneal ulcer formation in ocular rosacea. Invest Ophthalmol Vis Sci. 2012;53:3250-3259.
  2. Webster G, Schaller M. Ocular rosacea: a dermatologic perspective. J Am Acad Dermatol. 2013;69(6 suppl 1):S42-S43.
  3. Crawford GH, Pelle MT, James WD. Rosacea: I. etiology, pathogenesis, and subtype classification. J Am Acad Dermatol. 2004;51:327-341.
  4. Wilkin J, Dahl M, Detmar M, et al. Standard grading system for rosacea: report of the National Rosacea Society Expert Committee on the classification and staging of rosacea. J Am Acad Dermatol. 2004;50:907-912.
  5. Gallo RLGranstein RDKang S, et al. Standard classification and pathophysiology of rosacea: the 2017 update by the National Rosacea Society Expert Committee. J Am Acad Dermatol. 2018;78:148-155.
  6. Gao YY, Di Pascuale MA, Li W, et al. High prevalence of Demodex in eyelashes with cylindrical dandruff. Invest Ophthalmol Vis Sci. 2005;46:3089-3094.
  7. Fallen RS, Gooderham M. Rosacea: update on management and emerging therapies. Skin Therapy Lett. 2012;17:1-4.
  8. Erbagcı Z, Ozgoztası O. The significance of Demodex folliculorum density in rosacea. Int J Dermatol. 1998;37:421-425.
  9. Ahn CS, Huang WW. Rosacea pathogenesis. Dermatol Clin. 2018;36:81‐86.
  10. Forton FMN, De Maertelaer V. Two consecutive standardized skin surface biopsies: an improved sampling method to evaluate Demodex density as a diagnostic tool for rosacea and demodicosis. Acta Derm Venereol. 2017;97:242‐248.
  11. Yamasaki K, Kanada K, Macleod DT, et al. TLR2 expression is increased in rosacea and stimulates enhanced serine protease production by keratinocytes. J Invest Dermatol. 2011;131:688-697.
  12. Gold LM, Draelos ZD. New and emerging treatments for rosacea. Am J Clin Dermatol. 2015;16:457-461.
  13. Two AM, Del Rosso JQ. Kallikrein 5-mediated inflammation in rosacea: clinically relevant correlations with acute and chronic manifestations in rosacea and how individual treatments may provide therapeutic benefit. J Clin Aesthet Dermatol. 2014;7:20-25.
  14. Lacey N, Delaney S, Kavanagh K, et al. Mite-related bacterial antigens stimulate inflammatory cells in rosacea. Br J Dermatol. 2007;157:474-481.
  15. Forton F, Germaux MA, Brasseur T, et al. Demodicosis and rosacea: epidemiology and significance in daily dermatologic practice. J Am Acad Dermatol. 2005;52:74-87.
  16. Lee SH, Chun YS, Kim JH, et al. The relationship between Demodex and ocular discomfort. Invest Ophthalmol Vis Sci. 2010;51:2906-2911.
  17. Awais M, Anwar MI, Ilfikhar R, et al. Rosacea—the ophthalmic perspective. Cutan Ocul Toxicol. 2015;34:161-166.
  18. Luo X, Li J, Chen C, et al. Ocular demodicosis as a potential cause of ocular surface inflammation. Cornea. 2017;36(suppl 1):S9-S14.
  19. Ayyildiz T, Sezgin FM. The effect of ocular Demodex colonization on Schirmer test and OSDI scores in newly diagnosed dry eye patients. Eye Contact Lens. 2020;46(suppl 1):S39-S41.
  20. Sobolewska B, Doycheva D, Deuter CM, et al. Efficacy of topical ivermectin for the treatment of cutaneous and ocular rosacea [published online April 7, 2020]. Ocul Immunol Inflamm. doi:10.1080/09273948.2020.1727531
  21. Li J, O‘Reilly N, Sheha H, et al. Correlation between ocular Demodex infestation and serum immunoreactivity to Bacillus proteins in patients with facial rosacea. 2010;117:870-877.
  22. Gonzalez‐Hinojosa D, Jaime‐Villalonga A, Aguilar‐Montes G, et al. Demodex and rosacea: is there a relationship? Indian J Ophthalmol. 2018;66:36‐38.
  23. Sarac G, Cankaya C, Ozcan KN, et al. Increased frequency of Demodex blepharitis in rosacea and facial demodicosis patients. J Cosmet Dermatol. 2020;19:1260-1265.
  24. Forton FMN, De Maertelaer V. Rosacea and demodicosis: little-known diagnostic signs and symptoms. Acta Derm Venereol. 2019;99:47-52.
  25. Mizuno M, Kawashima M, Uchino M, et al. Demodex-mite infestation in cilia and its association with ocular surface parameters in Japanese volunteers. Eye Contact Lens. 2020;46:291-296.
References
  1. O’Reilly N, Gallagher C, Reddy Katikireddy K, et al. Demodex-associated Bacillus proteins induce an aberrant wound healing response in a corneal epithelial cell line: possible implications for corneal ulcer formation in ocular rosacea. Invest Ophthalmol Vis Sci. 2012;53:3250-3259.
  2. Webster G, Schaller M. Ocular rosacea: a dermatologic perspective. J Am Acad Dermatol. 2013;69(6 suppl 1):S42-S43.
  3. Crawford GH, Pelle MT, James WD. Rosacea: I. etiology, pathogenesis, and subtype classification. J Am Acad Dermatol. 2004;51:327-341.
  4. Wilkin J, Dahl M, Detmar M, et al. Standard grading system for rosacea: report of the National Rosacea Society Expert Committee on the classification and staging of rosacea. J Am Acad Dermatol. 2004;50:907-912.
  5. Gallo RLGranstein RDKang S, et al. Standard classification and pathophysiology of rosacea: the 2017 update by the National Rosacea Society Expert Committee. J Am Acad Dermatol. 2018;78:148-155.
  6. Gao YY, Di Pascuale MA, Li W, et al. High prevalence of Demodex in eyelashes with cylindrical dandruff. Invest Ophthalmol Vis Sci. 2005;46:3089-3094.
  7. Fallen RS, Gooderham M. Rosacea: update on management and emerging therapies. Skin Therapy Lett. 2012;17:1-4.
  8. Erbagcı Z, Ozgoztası O. The significance of Demodex folliculorum density in rosacea. Int J Dermatol. 1998;37:421-425.
  9. Ahn CS, Huang WW. Rosacea pathogenesis. Dermatol Clin. 2018;36:81‐86.
  10. Forton FMN, De Maertelaer V. Two consecutive standardized skin surface biopsies: an improved sampling method to evaluate Demodex density as a diagnostic tool for rosacea and demodicosis. Acta Derm Venereol. 2017;97:242‐248.
  11. Yamasaki K, Kanada K, Macleod DT, et al. TLR2 expression is increased in rosacea and stimulates enhanced serine protease production by keratinocytes. J Invest Dermatol. 2011;131:688-697.
  12. Gold LM, Draelos ZD. New and emerging treatments for rosacea. Am J Clin Dermatol. 2015;16:457-461.
  13. Two AM, Del Rosso JQ. Kallikrein 5-mediated inflammation in rosacea: clinically relevant correlations with acute and chronic manifestations in rosacea and how individual treatments may provide therapeutic benefit. J Clin Aesthet Dermatol. 2014;7:20-25.
  14. Lacey N, Delaney S, Kavanagh K, et al. Mite-related bacterial antigens stimulate inflammatory cells in rosacea. Br J Dermatol. 2007;157:474-481.
  15. Forton F, Germaux MA, Brasseur T, et al. Demodicosis and rosacea: epidemiology and significance in daily dermatologic practice. J Am Acad Dermatol. 2005;52:74-87.
  16. Lee SH, Chun YS, Kim JH, et al. The relationship between Demodex and ocular discomfort. Invest Ophthalmol Vis Sci. 2010;51:2906-2911.
  17. Awais M, Anwar MI, Ilfikhar R, et al. Rosacea—the ophthalmic perspective. Cutan Ocul Toxicol. 2015;34:161-166.
  18. Luo X, Li J, Chen C, et al. Ocular demodicosis as a potential cause of ocular surface inflammation. Cornea. 2017;36(suppl 1):S9-S14.
  19. Ayyildiz T, Sezgin FM. The effect of ocular Demodex colonization on Schirmer test and OSDI scores in newly diagnosed dry eye patients. Eye Contact Lens. 2020;46(suppl 1):S39-S41.
  20. Sobolewska B, Doycheva D, Deuter CM, et al. Efficacy of topical ivermectin for the treatment of cutaneous and ocular rosacea [published online April 7, 2020]. Ocul Immunol Inflamm. doi:10.1080/09273948.2020.1727531
  21. Li J, O‘Reilly N, Sheha H, et al. Correlation between ocular Demodex infestation and serum immunoreactivity to Bacillus proteins in patients with facial rosacea. 2010;117:870-877.
  22. Gonzalez‐Hinojosa D, Jaime‐Villalonga A, Aguilar‐Montes G, et al. Demodex and rosacea: is there a relationship? Indian J Ophthalmol. 2018;66:36‐38.
  23. Sarac G, Cankaya C, Ozcan KN, et al. Increased frequency of Demodex blepharitis in rosacea and facial demodicosis patients. J Cosmet Dermatol. 2020;19:1260-1265.
  24. Forton FMN, De Maertelaer V. Rosacea and demodicosis: little-known diagnostic signs and symptoms. Acta Derm Venereol. 2019;99:47-52.
  25. Mizuno M, Kawashima M, Uchino M, et al. Demodex-mite infestation in cilia and its association with ocular surface parameters in Japanese volunteers. Eye Contact Lens. 2020;46:291-296.
Issue
cutis - 108(1)
Issue
cutis - 108(1)
Page Number
46-50
Page Number
46-50
Publications
Publications
Topics
Article Type
Display Headline
Ocular Manifestations of Patients With Cutaneous Rosacea With and Without Demodex Infection
Display Headline
Ocular Manifestations of Patients With Cutaneous Rosacea With and Without Demodex Infection
Sections
Inside the Article

Practice Points

  • Rosacea is a common chronic inflammatory skin disease of the central facial skin and is of unknown origin. Patients with ocular rosacea may report dryness, itching, and photophobia.
  • Demodex infestation may increase cutaneous or ocular inflammation in rosacea.
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
Article PDF Media

Reexamining the Role of Diet in Dermatology

Article Type
Changed
Tue, 08/09/2022 - 09:46

Within the last decade, almost 3000 articles have been published on the role of diet in the prevention and management of dermatologic conditions. Patients are increasingly interested in—and employing—dietary modifications that may influence skin appearance and aid in the treatment of cutaneous disease.1 It is essential that dermatologists are familiar with existing evidence on the role of diet in dermatology to counsel patients appropriately. Herein, we discuss the compositions of several popular diets and their proposed utility for dermatologic purposes. We highlight the limited literature that exists surrounding this topic and emphasize the need for future, well-designed clinical trials that study the impact of diet on skin disease.

Ketogenic Diet

The ketogenic diet has a macronutrient profile composed of high fat, low to moderate protein, and very low carbohydrates. Nutritional ketosis occurs as the body begins to use free fatty acids (via beta oxidation) as the primary metabolite driving cellular metabolism. It has been suggested that the ketogenic diet may impart beneficial effects on skin disease; however, limited literature exists on the role of nutritional ketosis in the treatment of dermatologic conditions.

Mechanistically, the ketogenic diet decreases the secretion of insulin and insulinlike growth factor 1, resulting in a reduction of circulating androgens and increased activity of the retinoid X receptor.2 In acne vulgaris, it has been suggested that the ketogenic diet may be beneficial in decreasing androgen-induced sebum production and the overproliferation of keratinocytes.2-7 The ketogenic diet is one of the most rapidly effective dietary strategies for normalizing both insulin and androgens, thus it may theoretically be useful for other metabolic and hormone-dependent skin diseases, such as hidradenitis suppurativa.8,9

The cutaneous manifestations associated with chronic hyperinsulinemia and hyperglycemia are numerous and include acanthosis nigricans, acrochordons, diabetic dermopathy, scleredema diabeticorum, bullosis diabeticorum, keratosis pilaris, and generalized granuloma annulare. There also is an increased risk for bacterial and fungal skin infections associated with hyperglycemic states.10 The ketogenic diet is an effective nonpharmacologic tool for normalizing serum insulin and glucose levels in most patients and may have utility in the aforementioned conditions.11,12 In addition to improving insulin sensitivity, it has been used as a dietary strategy for weight loss.11-15 Because obesity and metabolic syndrome are highly correlated with common skin conditions such as psoriasis, hidradenitis suppurativa, and androgenetic alopecia, there may be a role for employing the ketogenic diet in these patient populations.16,17

Although robust clinical studies on ketogenic diets in skin disease are lacking, a recent single-arm, open-label clinical trial observed benefit in all 37 drug-naïve, overweight patients with chronic plaque psoriasis who underwent a ketogenic weight loss protocol. Significant reductions in psoriasis area and severity index (PASI) score and dermatology life quality index score were reported (P<.001).18 Another study of 30 patients with psoriasis found that a 4-week, low-calorie, ketogenic diet resulted in 50% improvement of PASI scores, 10% weight loss, and a reduction in the proinflammatory cytokines IL-1β and IL-2.19 Despite these results, it is a challenge to tease out if the specific dietary intervention or its associated weight loss was the main driver in these reported improvements in skin disease.

There is mixed evidence on the anti-inflammatory nature of the ketogenic diet, likely due to wide variation in the composition of foods included in individual diets. In many instances, the ketogenic diet is thought to possess considerable antioxidant and anti-inflammatory capabilities. Ketones are known activators of the nuclear factor erythroid 2–related factor 2 pathway, which upregulates the production of glutathione, a major endogenous intracellular antioxidant.20 Additionally, dietary compounds from foods that are encouraged while on the ketogenic diet, such as sulforaphane from broccoli, also are independent activators of nuclear factor erythroid 2–related factor 2.21 Ketones are efficiently utilized by mitochondria, which also may result in the decreased production of reactive oxygen species and lower oxidative stress.22 Moreover, the ketone body β-hydroxybutyrate has demonstrated the ability to reduce proinflammatory IL-1β levels via suppression of nucleotide-binding domain-like receptor protein 3 inflammasome activity.23,24 The activity of IL-1β is known to be elevated in many dermatologic conditions, including juvenile idiopathic arthritis, relapsing polychondritis, Schnitzler syndrome, hidradenitis suppurativa, Behçet disease, and other autoinflammatory syndromes.25 Ketones also have been shown to inhibit the nuclear factor–κB proinflammatory signaling pathway.22,26,27 Overexpression of IL-1β and aberrant activation of nuclear factor–κB are implicated in a variety of inflammatory, autoimmune, and oncologic cutaneous pathologies. The ketogenic diet may prove to be an effective adjunctive treatment for dermatologists to consider in select patient populations.23,24,28-30



For patients with keratinocyte carcinomas, the ketogenic diet may offer the aforementioned anti-inflammatory and antioxidant effects, as well as suppression of the mechanistic target of rapamycin, a major regulator of cell metabolism and proliferation.31,32 Inhibition of mechanistic target of rapamycin activity has been shown to slow tumor growth and reduce the development of squamous cell carcinoma.25,33,34 The ketogenic diet also may exploit the preferential utilization of glucose exhibited by many types of cancer cells, thereby “starving” the tumor of its primary fuel source.35,36 In vitro and animal studies in a variety of cancer types have demonstrated that a ketogenic metabolic state—achieved through the ketogenic diet or fasting—can sensitize tumor cells to chemotherapy and radiation while conferring a protective effect to normal cells.37-40 This recently described phenomenon is known as differential stress resistance, but it has not been studied in keratinocyte malignancies or melanoma to date. Importantly, some basal cell carcinomas and BRAF V600E–mutated melanomas have worsened while on the ketogenic diet, suggesting more data is needed before it can be recommended for all cancer patients.41,42 Furthermore, other skin conditions such as prurigo pigmentosa have been associated with initiation of the ketogenic diet.43

 

 

Low FODMAP Diet

Fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) are short-chain carbohydrates that are poorly absorbed, osmotically active, and rapidly fermented by intestinal bacteria.44 The low FODMAP diet has been shown to be efficacious for treatment of irritable bowel syndrome, small intestinal bacterial overgrowth (SIBO), and some cases of inflammatory bowel disease (IBD).44-49 A low FODMAP diet may have potential implications for several dermatologic conditions.

Rosacea has been associated with various gastrointestinal tract disorders including irritable bowel syndrome, SIBO, and IBD.50-54 A single study found that patients with rosacea had a 13-fold increased risk for SIBO.55,56 Treatment of 40 patients with SIBO using rifaximin resulted in complete resolution of rosacea in all patients, with no relapse after a 3-year follow-up period.55 Psoriasis also has been associated with SIBO and IBD.57,58 One small study found that eradication of SIBO in psoriatic patients resulted in improved PASI scores and colorimetric values.59

Although the long-term health consequences of the low FODMAP diet are unknown, further research on such dietary interventions for inflammatory skin conditions is warranted given the mounting evidence of a gut-skin connection and the role of the intestinal microbiome in skin health.50,51

Gluten-Free Diet

Gluten is a protein found in a variety of grains. Although the role of gluten in the pathogenesis of celiac disease and dermatitis herpetiformis is indisputable, the deleterious effects of gluten outside of the context of these diseases remain controversial. There may be a compelling case for eliminating gluten in psoriasis patients with seropositivity for celiac disease. A recent systematic review found a 2.2-fold increased risk for celiac disease in psoriasis patients.60 Antigliadin antibody titers also were found to be positively correlated with psoriatic disease severity.61 In addition, one open-label study found a reduction in PASI scores in 73% of patients with antigliadin antibodies after 3 months on a gluten-free diet compared to those without antibodies; however, the study only included 22 patients.62 Several other small studies have yielded similar results63,64; however, antigliadin antibodies are neither the most sensitive nor specific markers of celiac disease, and additional testing should be completed in any patient who may carry this diagnosis. A survey study by the National Psoriasis Foundation found that the dietary change associated with the greatest skin improvement was removal of gluten and nightshade vegetables in approximately 50% of the 1200 psoriasis patients that responded.65 Case reports of various dermatologic conditions including sarcoidosis, vitiligo, alopecia areata, lichen planus, dermatomyositis, pyoderma gangrenosum, erythema nodosum, leukocytoclastic vasculitis, linear IgA bullous dermatosis, and aphthous ulcerations have reportedly improved with a gluten-free diet; however, this should not be used as primary therapy in patients without celiac disease.66-71 Because gluten-free diets can be expensive and challenging to follow, a formal assessment for celiac disease should be considered before recommendation of this dietary intervention.

Low Histamine Diet

Histamine is a biogenic amine produced by the decarboxylation of the amino acid histidine.72 It is found in several foods in varying amounts. Because bacteria can convert histidine into histamine, many fermented and aged foods such as kimchi, sauerkraut, cheese, and red wine contain high levels of histamine. Individuals who have decreased activity of diamine oxidase (DAO), an enzyme that degrades histamine, may be more susceptible to histamine intolerance.72 The symptoms of histamine intolerance are numerous and include gastrointestinal tract distress, rhinorrhea and nasal congestion, headache, urticaria, flushing, and pruritus. Histamine intolerance can mimic an IgE-mediated food allergy; however, allergy testing is negative in these patients. Unfortunately, there is no laboratory test for histamine intolerance; a double-blind, placebo-controlled food challenge is considered the gold-standard test.72

As it pertains to dermatology, a low histamine diet may play a role in the treatment of certain patients with atopic dermatitis and chronic spontaneous urticaria. One study reported that 17 of 54 (31.5%) atopic patients had higher basal levels of serum histamine compared to controls.73 Another study found that a histamine-free diet led to improvement in both histamine intolerance symptoms and atopic dermatitis disease severity (SCORing atopic dermatitis) in patients with low DAO activity.74 In chronic spontaneous urticaria, a recent systematic review found that in 223 patients placed on a low histamine diet for 3 to 4 weeks, 12% and 44% achieved complete and partial remission, respectively.75 Although treatment response based on a patient’s DAO activity level has not been correlated, a diet low in histamine may prove useful for patients with persistent atopic dermatitis and chronic spontaneous urticaria who have negative food allergy tests and report exacerbation of symptoms after ingestion of histamine-rich foods.76,77

Mediterranean Diet

The Mediterranean diet has been touted as one of the healthiest diets to date, and large randomized clinical trials have demonstrated its effectiveness in weight loss, improving insulin sensitivity, and reducing inflammatory cytokine profiles.78,79 A major criticism of the Mediterranean diet is that it has considerable ambiguity and lacks a precise definition due to the variability of what is consumed in different Mediterranean regions. Generally, the diet emphasizes high consumption of colorful fruits and vegetables, aromatic herbs and spices, olive oil, nuts, and seafood, as well as modest amounts of dairy, eggs, and red meat.80 The anti-inflammatory effects of this diet largely have been attributed to its abundance of polyphenols, carotenoids, monounsaturated fatty acids, and omega-3 polyunsaturated fatty acids (PUFAs).80,81 Examples of polyphenols include resveratrol in red grapes, quercetin in apples and red onions, and curcumin in turmeric, while examples of carotenoids include lycopene in tomatoes and zeaxanthin in dark leafy greens. Oleic acid is a monounsaturated fatty acid present in high concentrations in olive oil, while eicosapentaenoic acid and docosahexaenoic acid are omega-3 PUFAs predominantly found in fish.82

Unfortunately, rigorous clinical trials regarding the Mediterranean diet as it pertains to dermatology have not been undertaken. Numerous observational studies in patients with psoriasis have suggested that close adherence to the Mediterranean diet was associated with improvement in PASI scores.83-86 The National Psoriasis Foundation now recommends a trial of the Mediterranean diet in some patients with psoriasis, emphasizing increased dietary intake of olive oil, fish, and vegetables.87 Adherence to a Mediterranean diet also has been inversely correlated to the severity of acne vulgaris and hidradenitis suppurativa88,89; however, these studies failed to account for the multifactorial risk factors associated with these conditions. Mediterranean diets also may impart a chemopreventive effect, supported by a number of in vivo and in vitro studies demonstrating the inhibition and/or reversal of cutaneous DNA damage induced by UV radiation through supplementation with various phytonutrients and omega-3 PUFAs.81,90-92 Although small case-control studies have found a decreased risk of basal cell carcinoma in those who closely adhered to a Mediterranean diet, more rigorous clinical research is needed.93

 

 

Whole-Food, Plant-Based Diet

A whole-food, plant-based (WFPB) diet is another popular dietary approach that consists of eating fruits, vegetables, legumes, nuts, seeds, and grains in their whole natural form.94 This diet discourages all animal products, including red meat, seafood, dairy, and eggs. It is similar to a vegan diet except that it eliminates all highly refined carbohydrates, vegetable oils, and other processed foods.94 Randomized clinical studies have demonstrated the WFPB diet to be effective in the treatment of obesity and metabolic syndrome.95,96

A WFPB diet has been shown to increase the antioxidant capacity of cells, lengthen telomeres, and reduce formation of advanced glycation end products.94,97,98 These benefits may help combat accelerated skin aging, including increased skin permeability, reduced elasticity and hydration, decreased angiogenesis, impaired immune function, and decreased vitamin D synthesis. Accelerated skin aging can result in delayed wound healing and susceptibility to skin tears and ecchymoses and also may promote the development of cutaneous malignancies.99 There remains a lack of clinical data studying a properly formulated WFPB diet in the dermatologic setting.

Paleolithic Diet

The paleolithic (Paleo) diet is an increasingly popular way of eating that attempts to mirror what our ancestors may have consumed between 10,000 and 2.5 million years ago.100 It is similar to the Mediterranean diet but excludes grains, dairy, legumes, and nightshade vegetables. It also calls for elimination of highly processed sugars and oils as well as chemical food additives and preservatives. There is a strict variation of the diet for individuals with autoimmune disease that also excludes eggs, nuts, and seeds, as these can be inflammatory or immunogenic in some patients.100-106 Other variations of the diet exist, including the ketogenic Paleo diet, pegan (Paleo vegan) diet, and lacto-Paleo diet.100 An often cited criticism of the Paleo diet is the low intake of calcium and risk for osteoporosis; however, consumption of calcium-rich foods or a calcium supplement can address this concern.107

Although small clinical studies have found the Paleo diet to be beneficial for various autoimmune diseases, clinical data evaluating the utility of the diet for cutaneous disease is lacking.108,109 Numerous randomized trials have demonstrated the Paleo diet to be effective for weight loss and improving insulin sensitivity and lipid levels.110-116 Thus, the Paleo diet may theoretically serve as a viable adjunct dietary approach to the treatment of cutaneous diseases associated with obesity and metabolic derangement.117

Carnivore Diet

Arguably the most controversial and radical diet is the carnivore diet. As the name implies, the carnivore diet is based on consuming solely animal products. A properly structured carnivore diet emphasizes a “nose-to-tail” eating approach where all parts of the animal including the muscle meats, organs, and fat are consumed. Proponents of the diet cite anthropologic evidence from fossil-stable carbon-13/carbon-12 isotope analyses, craniodental features, and numerous other adaptations that indicate increased consumption of meat during human evolution.118-122 Notably, many early humans ate a carnivore diet, but life span was very short at this time, suggesting the diet may not be as beneficial as has been suggested.

Despite the abundance of anecdotal evidence supporting its use for a variety of chronic conditions, including cutaneous autoimmune disease, there is a virtual absence of high-quality research on the carnivore diet.123-125



The purported benefits of the carnivore diet may be attributed to the consumption of organ meats that contain highly bioavailable essential vitamins and minerals, such as iron, zinc, copper, selenium, thiamine, niacin, folate, vitamin B6, vitamin B12, vitamin A, vitamin D, vitamin K, and choline.126-128 Other dietary compounds that have demonstrated benefit for skin health and are predominantly found in animal foods include carnosine, carnitine, creatine, taurine, coenzyme Q10, and collagen.129-134 Nevertheless, there is no data to recommend the elimination of antioxidant- and micronutrient-dense plant-based foods. Rigorous clinical research evaluating the efficacy and safety of the carnivore diet in dermatologic patients is needed. A carnivore diet should not be undertaken without the assistance of a dietician who can ensure adequate micronutrient and macronutrient support.

Final Thoughts

The adjunctive role of diet in the treatment of skin disease is expanding and becoming more widely accepted among dermatologists. Unfortunately, there remains a lack of randomized controlled trials confirming the efficacy of various dietary interventions in the dermatologic setting. Although evidence-based dietary recommendations currently are limited, it is important for dermatologists to be aware of the varied and nuanced dietary interventions employed by patients.

Ultimately, dietary recommendations must be personalized, considering a patient’s comorbidities, personal beliefs and preferences, and nutrigenetics. The emerging field of dermatonutrigenomics—the study of how dietary compounds interact with one’s genes to influence skin health—may allow for precise dietary recommendations to be made in dermatologic practice. Direct-to-consumer genetic tests targeted toward dermatology patients are already on the market, but their clinical utility awaits validation.1 Because nutritional science is a constantly evolving field, becoming familiar with these popular diets will serve both dermatologists and their patients well.

References
  1. Jaros J, Katta R, Shi VY. Dermatonutrigenomics: past, present, and future. Dermatology. 2019;235:164-166. 
  2. Paoli A, Grimaldi K, Toniolo L, et al. Nutrition and acne: therapeutic potential of ketogenic diets. Skin Pharmacol Physiol. 2012;25:111-117. 
  3. Melnik BC, Schmitz G. Role of insulin, insulin-like growth factor-1, hyperglycaemic food and milk consumption in the pathogenesis of acne vulgaris. Exp Dermatol. 2009;18:833-841. 
  4. Smith RN, Mann NJ, Braue A, et al. The effect of a high-protein, low glycemic-load diet versus a conventional, high glycemic-load diet on biochemical parameters associated with acne vulgaris: a randomized, investigator-masked, controlled trial. J Am Acad Dermatol. 2007;57:247-256. 
  5. Smith R, Mann N, Mäkeläinen H, et al. A pilot study to determine the short-term effects of a low glycemic load diet on hormonal markers of acne: a nonrandomized, parallel, controlled feeding trial. Mol Nutr Food Res. 2008;52:718-726. 
  6. Smith RN, Braue A, Varigos GA, et al. The effect of a low glycemic load diet on acne vulgaris and the fatty acid composition of skin surface triglycerides. J Dermatol Sci. 2008;50:41-52. 
  7. Kwon HH, Yoon JY, Hong JS, et al. Clinical and histological effect of a low glycaemic load diet in treatment of acne vulgaris in Korean patients: a randomized, controlled trial. Acta Derm Venereol. 2012;92:241-246. 
  8. Khandalavala BN, Do MV. Finasteride in hidradenitis suppurativa: a "male" therapy for a predominantly "female" disease. J Clin Aesthet Dermatol. 2016;9:44. 
  9. Nikolakis G, Karagiannidis I, Vaiopoulos AG, et al. Endocrinological mechanisms in the pathophysiology of hidradenitis suppurativa [in German]. Hautarzt. 2020;71:762-771. 
  10. Karadag AS, Ozlu E, Lavery MJ. Cutaneous manifestations of diabetes mellitus and the metabolic syndrome. Clin Dermatology. 2018;36:89-93. 
  11. Gardner CD, Kiazand A, Alhassan S, et al. Comparison of the Atkins, Zone, Ornish, and LEARN diets for change in weight and related risk factors among overweight premenopausal women: the A TO Z Weight Loss Study: a randomized trial. JAMA. 2007;297:969-977. 
  12. Anton SD, Hida A, Heekin K, et al. Effects of popular diets without specific calorie targets on weight loss outcomes: systematic review of findings from clinical trials. Nutrients. 2017;9:822. 
  13. Castellana M, Conte E, Cignarelli A, et al. Efficacy and safety of very low calorie ketogenic diet (VLCKD) in patients with overweight and obesity: a systematic review and meta-analysis. Rev Endocr Metab Disord. 2020;21:5-16. 
  14. Paoli A, Mancin L, Giacona MC, et al. Effects of a ketogenic diet in overweight women with polycystic ovary syndrome. J Transl Med. 2020;18:104. 
  15. Dashti HM, Mathew TC, Hussein T, et al. Long-term effects of a ketogenic diet in obese patients. Exp Clin Cardiol. 2004;9:200-205. 
  16. Lian N, Chen M. Metabolic syndrome and skin disease: potential connection and risk. Int J Dermatol Venereol. 2019;2:89-93. 
  17. Hu Y, Zhu Y, Lian N, et al. Metabolic syndrome and skin diseases. Front Endocrinol (Lausanne). 2019;10:788. 
  18. Castaldo G, Rastrelli L, Galdo G, et al. Aggressive weight-loss program with a ketogenic induction phase for the treatment of chronic plaque psoriasis: a proof-of-concept, single-arm, open-label clinical trial. Nutrition. 2020;74:110757. 
  19. Castaldo G, Pagano I, Grimaldi M, et al. Effect of very-low-calorie ketogenic diet on psoriasis patients: a nuclear magnetic resonance-based metabolomic study. J Proteome Res. 2021;20:1509-1521. 
  20. Milder J, Liang L-P, Patel M. Acute oxidative stress and systemic Nrf2 activation by the ketogenic diet. Neurobiol Dis. 2010;40:238-244. 
  21. Kubo E, Chhunchha B, Singh P, et al. Sulforaphane reactivates cellular antioxidant defense by inducing Nrf2/ARE/Prdx6 activity during aging and oxidative stress. Sci Rep. 2017;7:14130. 
  22. Pinto A, Bonucci A, Maggi E, et al. Anti-oxidant and anti-inflammatory activity of ketogenic diet: new perspectives for neuroprotection in Alzheimer's disease. Antioxidants (Basel). 2018;7:63. 
  23. Youm Y-H, Nguyen KY, Grant RW, et al. The ketone metabolite &#946;-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21:263-269. 
  24. Kelley N, Jeltema D, Duan Y, et al. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20:3328. 
  25. Fomin DA, McDaniel B, Crane J. The promising potential role of ketones in inflammatory dermatologic disease: a new frontier in treatment research. J Dermatol Treat. 2017;28:484-487. 
  26. Rahman M, Muhammad S, Khan MA, et al. The β-hydroxybutyrate receptor HCA 2 activates a neuroprotective subset of macrophages. Nat Commun. 2014;5:1-11. 
  27. Lu Y, Yang YY, Zhou MW, et al. Ketogenic diet attenuates oxidative stress and inflammation after spinal cord injury by activating Nrf2 and suppressing the NF-&#954;B signaling pathways. Neurosci Lett. 2018;683:13-18. 
  28.  Hamarsheh S, Zeiser R. NLRP3 inflammasome activation in cancer: a double-edged sword. Front Immunol. 2020;11:1444. 
  29. Bell S, Degitz K, Quirling M, et al. Involvement of NF-&#954;B signalling in skin physiology and disease. Cell Signal. 2003;15:1-7. 
  30. Goldminz AM, Au SC, Kim N, et al. NF-κB: an essential transcription factor in psoriasis. J Dermatol Sci. 2013;69:89-94. 
  31. Laplante M, Sabatini DM. mTOR signaling at a glance. J Cell Sci. 2009;122:3589. 
  32. McDaniel S, Rensing N, Yamada K, et al. The ketogenic diet inhibits the mammalian target of rapamycin (mTOR) pathway. Epilepsia. 2011;52:E7-E11. 
  33. Alter M, Satzger I, Schrem H, et al. Non-melanoma skin cancer is reduced after switch of immunosuppression to mTOR-inhibitors in organ transplant recipients. J Dtsch Dermatol Ges. 2014;12:480-488. 
  34. Feldmeyer L, Hofbauer GF, Böni T, et al. Mammalian target of rapamycin (mTOR) inhibitors slow skin carcinogenesis, but impair wound healing. Br J Dermatol. 2012;166:422-424. 
  35. Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016;41:211-218. 
  36. Li W. "Warburg effect" and mitochondrial metabolism in skin cancer.J Carcinogene Mutagene. 2012:S4. 
  37. Naveed S, Aslam M, Ahmad A. Starvation based differential chemotherapy: a novel approach for cancer treatment. Oman Med J. 2014;29:391-398. 
  38. Raffaghello L, Lee C, Safdie FM, et al. Starvation-dependent differential stress resistance protects normal but not cancer cells against high-dose chemotherapy. Proc Natl Acad Sci U S A. 2008;105:8215-8220. 
  39. Buono R, Longo VD. Starvation, stress resistance, and cancer. Trends Endocrinol Metab. 2018;29:271-280. 
  40. de Groot S, Pijl H, van der Hoeven JJM, et al. Effects of short-term fasting on cancer treatment. J Exp Clin Cancer Res. 2019;38:209. 
  41. Hosseini M, Kasraian Z, Rezvani HR. Energy metabolism in skin cancers: a therapeutic perspective. Biochim Biophys Acta Bioenerg. 2017;1858:712-722. 
  42. Feichtinger RG, Lang R, Geilberger R, et al. Melanoma tumors exhibit a variable but distinct metabolic signature. Exp Dermatol. 2018;27:204-207. 
  43. Alshaya MA, Turkmani MG, Alissa AM. Prurigo pigmentosa following ketogenic diet and bariatric surgery: a growing association. JAAD Case Rep. 2019;5:504-507. 
  44. Bellini M, Tonarelli S, Nagy AG, et al. Low FODMAP diet: evidence, doubts, and hopes. Nutrients. 2020;12:148. 
  45. Kwiatkowski L, Rice E, Langland J. Integrative treatment of chronic abdominal bloating and pain associated with overgrowth of small intestinal bacteria: a case report. Altern Ther Health Med. 2017;23:56-61. 
  46. Hubkova T. No more pain in the gut: lifestyle medicine approach to irritable bowel syndrome. Am J Lifestyle Med. 2017;11:223-226. 
  47. Schumann D, Klose P, Lauche R, et al. Low fermentable, oligo-, di-, mono-saccharides and polyol diet in the treatment of irritable bowel syndrome: a systematic review and meta-analysis. Nutrition. 2018;45:24-31. 
  48. Cox SR, Prince AC, Myers CE, et al. Fermentable carbohydrates [FODMAPs] exacerbate functional gastrointestinal symptoms in patients with inflammatory bowel disease: a randomised, double-blind, placebo-controlled, cross-over, re-challenge trial. J Crohns Colitis. 2017;11:1420-1429. 
  49. Damas OM, Garces L, Abreu MT. Diet as adjunctive treatment for inflammatory bowel disease: review and update of the latest literature. Curr Treat Options Gastroenterol. 2019;17:313-325. 
  50. Wang FY, Chi CC. Rosacea, germs, and bowels: a review on gastrointestinal comorbidities and gut-skin axis of rosacea. Adv Ther. 2021;38:1415-1424. 
  51. Daou H, Paradiso M, Hennessy K, et al. Rosacea and the microbiome: a systematic review. Dermatol Ther (Heidelb). 2021;11:1-12. 
  52. Weinstock LB, Steinhoff M. Rosacea and small intestinal bacterial overgrowth: prevalence and response to rifaximin. J Am Acad Dermatol. 2013;68:875-876. 
  53. Wu CY, Chang YT, Juan CK, et al. Risk of inflammatory bowel disease in patients with rosacea: results from a nationwide cohort study in Taiwan. J Am Acad Dermatol. 2017;76:911-917. 
  54. Egeberg A, Weinstock LB, Thyssen EP, et al. Rosacea and gastrointestinal disorders: a population-based cohort study. Br J Dermatol. 2017;176:100-106. 
  55. Drago F, De Col E, Agnoletti AF, et al. The role of small intestinal bacterial overgrowth in rosacea: a 3-year follow-up. J Am Acad Dermatol. 2016;75:E113-E115. 
  56. Parodi A, Paolino S, Greco A, et al. Small intestinal bacterial overgrowth in rosacea: clinical effectiveness of its eradication. Clin Gastroenterol Hepatol. 2008;6:759-764. 
  57. Ojetti V, De Simone C, Aguilar Sanchez J, et al. Malabsorption in psoriatic patients: cause or consequence? Scand J Gastroenterol. 2006;41:1267-1271. 
  58. Kim M, Choi KH, Hwang SW, et al. Inflammatory bowel disease is associated with an increased risk of inflammatory skin diseases: a population-based cross-sectional study. J Am Acad Dermatol. 2017;76:40-48. 
  59. Drago F, Ciccarese G, Indemini E, et al. Psoriasis and small intestine bacterial overgrowth. Int J Dermatol. 2018;57:112-113. 
  60. Acharya P, Mathur M. Association between psoriasis and celiac disease: a systematic review and meta-analysis. J Am Acad Dermatol. 2020;82:1376-1385. 
  61. Bhatia BK, Millsop JW, Debbaneh M, et al. Diet and psoriasis, part II: celiac disease and role of a gluten-free diet. J Am Acad Dermatol. 2014;71:350-358. 
  62. Michaëlsson G, Gerdén B, Hagforsen E, et al. Psoriasis patients with antibodies to gliadin can be improved by a gluten-free diet. Br J Dermatol. 2000;142:44-51. 
  63. Kolchak NA, Tetarnikova MK, Theodoropoulou MS, et al. Prevalence of antigliadin IgA antibodies in psoriasis vulgaris and response of seropositive patients to a gluten-free diet. J Multidiscip Healthc. 2018;11:13-19. 
  64. De Bastiani R, Gabrielli M, Lora L, et al. Association between coeliac disease and psoriasis: Italian primary care multicentre study. Dermatology. 2015;230:156-160. 
  65. Afifi L, Danesh MJ, Lee KM, et al. Dietary behaviors in psoriasis: patient-reported outcomes from a U.S. national survey. Dermatol Ther (Heidelb). 2017;7:227-242. 
  66. Loche F, Bazex J. Celiac disease associated with cutaneous sarcoidosic granuloma [in French]. Rev Med Interne. 1997;18:975-978. 
  67. Rodríguez-García C, González-Hernández S, Pérez-Robayna N, et al. Repigmentation of vitiligo lesions in a child with celiac disease after a gluten-free diet. Pediatr Dermatol. 2011;28:209-210. 
  68. Wijarnpreecha K, Panjawatanan P, Corral JE, et al. Celiac disease and risk of sarcoidosis: a systematic review and meta-analysis. J Evid Based Med. 2019;12:194-199. 
  69. Rodrigo L, Beteta-Gorriti V, Alvarez N, et al. Cutaneous and mucosal manifestations associated with celiac disease. Nutrients. 2018;10:800. 
  70. Song MS, Farber D, Bitton A, et al. Dermatomyositis associated with celiac disease: response to a gluten-free diet. Can J Gastroenterol. 2006;20:433-435. 
  71. Egan CA, Smith EP, Taylor TB, et al. Linear IgA bullous dermatosis responsive to a gluten-free diet. Am J Gastroenterol. 2001;96:1927-1929. 
  72. Comas-Basté O, Sánchez-Pérez S, Veciana-Nogués MT, et al. Histamine intolerance: the current state of the art. Biomolecules. 2020;10:1181. 
  73. Ring J. Plasma histamine concentrations in atopic eczema. Clin Allergy. 1983;13:545-552. 
  74. Maintz L, Benfadal S, Allam JP, et al. Evidence for a reduced histamine degradation capacity in a subgroup of patients with atopic eczema. J Allergy Clin Immunol. 2006;117:1106-1112. 
  75. Cornillier H, Giraudeau B, Samimi M, et al. Effect of diet in chronic spontaneous urticaria: a systematic review. Acta Derm Venereol. 2019;99:127-132. 
  76. Son JH, Chung BY, Kim HO, et al. A histamine-free diet is helpful for treatment of adult patients with chronic spontaneous urticaria. Ann Dermatol. 2018;30:164-172. 
  77. Wagner N, Dirk D, Peveling-Oberhag A, et al. A popular myth - low-histamine diet improves chronic spontaneous urticaria - fact or fiction? J Eur Acad Dermatol Venereol. 2017;31:650-655. 
  78. Esposito K, Marfella R, Ciotola M, et al. Effect of a Mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: a randomized trial. JAMA. 2004;292:1440-1446. 
  79. Steffen LM, Van Horn L, Daviglus ML, et al. A modified Mediterranean diet score is associated with a lower risk of incident metabolic syndrome over 25 years among young adults: the CARDIA (coronary artery risk development in young adults) study. Br J Nutr. 2014;112:1654-1661. 
  80. Bower A, Marquez S, de Mejia EG. The health benefits of selected culinary herbs and spices found in the traditional Mediterranean diet. Crit Rev Food Sci Nutr. 2016;56:2728-2746. 
  81. Bosch R, Philips N, Suárez-Pérez JA, et al. Mechanisms of photoaging and cutaneous photocarcinogenesis, and photoprotective strategies with phytochemicals. Antioxidants (Basel). 2015;4:248-268. 
  82. Katsimbri P, Korakas E, Kountouri A, et al. The effect of antioxidant and anti-inflammatory capacity of diet on psoriasis and psoriatic arthritis phenotype: nutrition as therapeutic tool? Antioxidants. 2021;10:157. 
  83. Molina-Leyva A, Cuenca-Barrales C, Vega-Castillo JJ, et al. Adherence to Mediterranean diet in Spanish patients with psoriasis: cardiovascular benefits? Dermatol Ther. 2019;32:E12810. 
  84. Barrea L, Balato N, Di Somma C, et al. Nutrition and psoriasis: is there any association between the severity of the disease and adherence to the Mediterranean diet? J Transl Med. 2015;13:1-10. 
  85. Phan C, Touvier M, Kesse-Guyot E, et al. Association between Mediterranean anti-inflammatory dietary profile and severity of psoriasis: results from the NutriNet-Santé cohort. JAMA Dermatol. 2018;154:1017-1024. 
  86. Korovesi A, Dalamaga M, Kotopouli M, et al. Adherence to the Mediterranean diet is independently associated with psoriasis risk, severity, and quality of life: a cross-sectional observational study. Int J Dermatol. 2019;58:E164-E165. 
  87. Ford AR, Siegel M, Bagel J, et al. Dietary recommendations for adults with psoriasis or psoriatic arthritis from the medical board of the National Psoriasis Foundation: a systematic review. JAMA Dermatol. 2018;154:934-950. 
  88. Skroza N, Tolino E, Semyonov L, et al. Mediterranean diet and familial dysmetabolism as factors influencing the development of acne. Scand J Public Health. 2012;40:466-474. 
  89. Barrea L, Fabbrocini G, Annunziata G, et al. Role of nutrition and adherence to the Mediterranean diet in the multidisciplinary approach of hidradenitis suppurativa: evaluation of nutritional status and its association with severity of disease. Nutrients. 2018;11:57. 
  90. Nichols JA, Katiyar SK. Skin photoprotection by natural polyphenols: anti-inflammatory, antioxidant and DNA repair mechanisms. Arch Dermatol Res. 2010;302:71-83. 
  91. Huang T-H, Wang P-W, Yang S-C, et al. Cosmetic and therapeutic applications of fish oil's fatty acids on the skin. Mar Drugs. 2018;16:256. 
  92. Rizwan M, Rodriguez-Blanco I, Harbottle A, et al. Tomato paste rich in lycopene protects against cutaneous photodamage in humans in vivo: a randomized controlled trial. Br J Dermatol. 2011;164:154-162. 
  93. Leone A, Martínez-González M, Martin-Gorgojo A, et al. Mediterranean diet, dietary approaches to stop hypertension, and pro-vegetarian dietary pattern in relation to the risk of basal cell carcinoma: a nested case-control study within the Seguimiento Universidad de Navarra (SUN) cohort. Am J Clin Nutr. 2020;112:364-372. 
  94. Solway J, McBride M, Haq F, et al. Diet and dermatology: the role of a whole-food, plant-based diet in preventing and reversing skin aging--a review. J Clin Aesthet Dermatol. 2020;13:38-43. 
  95. Greger M. A whole food plant-based diet is effective for weight loss: the evidence. Am J Lifestyle Med. 2020;14:500-510. 
  96. Wright N, Wilson L, Smith M, et al. The BROAD study: a randomised controlled trial using a whole food plant-based diet in the community for obesity, ischaemic heart disease or diabetes. Nutr Diabetes. 2017;7:E256. 
  97. Ornish D, Lin J, Chan JM, et al. Effect of comprehensive lifestyle changes on telomerase activity and telomere length in men with biopsy-proven low-risk prostate cancer: 5-year follow-up of a descriptive pilot study. Lancet Oncol. 2013;14:1112-1120. 
  98. Ornish D, Lin J, Daubenmier J, et al. Increased telomerase activity and comprehensive lifestyle changes: a pilot study. Lancet Oncol. 2008;9:1048-1057. 
  99. Zouboulis CC, Makrantonaki E. Clinical aspects and molecular diagnostics of skin aging. Clin Dermatol. 2011;29:3-14. 
  100. Gupta L, Khandelwal D, Lal PR, et al. Palaeolithic diet in diabesity and endocrinopathies--a vegan's perspective. Eur Endocrinol. 2019;15:77-82. 
  101. Chassaing B, Van de Wiele T, De Bodt J, et al. Dietary emulsifiers directly alter human microbiota composition and gene expression ex vivo potentiating intestinal inflammation. Gut. 2017;66:1414-1427. 
  102. Thorburn Alison N, Macia L, Mackay Charles R. Diet, metabolites, and "Western lifestyle" inflammatory diseases. Immunity. 2014;40:833-842. 
  103. Katta R, Schlichte M. Diet and dermatitis: food triggers. J Clin Aesthet Dermatol. 2014;7:30-36. 
  104. Dhar S, Srinivas SM. Food allergy in atopic dermatitis. Indian J Dermatol. 2016;61:645-648. 
  105. Birmingham N, Thanesvorakul S, Gangur V. Relative immunogenicity of commonly allergenic foods versus rarely allergenic and nonallergenic foods in mice. J Food Prot. 2002;65:1988-1991. 
  106. Yu W, Freeland DMH, Nadeau KC. Food allergy: immune mechanisms, diagnosis and immunotherapy. Nat Rev Immunol. 2016;16:751-765. 
  107. Kowalski LM, Bujko J. Evaluation of biological and clinical potential of paleolithic diet [in Polish]. Rocz Panstw Zakl Hig. 2012;63:9-15. 
  108. Lee JE, Titcomb TJ, Bisht B, et al. A modified MCT-based ketogenic diet increases plasma β-hydroxybutyrate but has less effect on fatigue and quality of life in people with multiple sclerosis compared to a modified paleolithic diet: a waitlist-controlled, randomized pilot study. J Am Coll Nutr. 2021;40:13-25. 
  109. Abbott RD, Sadowski A, Alt AG. Efficacy of the autoimmune protocol diet as part of a multi-disciplinary, supported lifestyle intervention for Hashimoto's thyroiditis. Cureus. 2019;11:E4556. 
  110. Lindeberg S, Jönsson T, Granfeldt Y, et al. A palaeolithic diet improves glucose tolerance more than a Mediterranean-like diet in individuals with ischaemic heart disease. Diabetologia. 2007;50:1795-1807. 
  111. Jönsson T, Granfeldt Y, Ahrén B, et al. Beneficial effects of a paleolithic diet on cardiovascular risk factors in type 2 diabetes: a randomized cross-over pilot study. Cardiovasc Diabetol. 2009;8:35. 
  112. Boers I, Muskiet FAJ, Berkelaar E, et al. Favourable effects of consuming a palaeolithic-type diet on characteristics of the metabolic syndrome: a randomized controlled pilot-study. Lipids Health Dis. 2014;13:160. 
  113. Ghaedi E, Mohammadi M, Mohammadi H, et al. Effects of a paleolithic diet on cardiovascular disease risk factors: a systematic review and meta-analysis of randomized controlled trials. Adv Nutr. 2019;10:634-646. 
  114. Mellberg C, Sandberg S, Ryberg M, et al. Long-term effects of a palaeolithic-type diet in obese postmenopausal women: a 2-year randomized trial. Eur J Clin Nutr. 2014;68:350-357. 
  115. Pastore RL, Brooks JT, Carbone JW. Paleolithic nutrition improves plasma lipid concentrations of hypercholesterolemic adults to a greater extent than traditional heart-healthy dietary recommendations. Nutr Res. 2015;35:474-479. 
  116. Otten J, Stomby A, Waling M, et al. Benefits of a paleolithic diet with and without supervised exercise on fat mass, insulin sensitivity, and glycemic control: a randomized controlled trial in individuals with type 2 diabetes. Diabetes Metab Res Rev. 2017;33:E2828. 
  117. Stefanadi EC, Dimitrakakis G, Antoniou C-K, et al. Metabolic syndrome and the skin: a more than superficial association. reviewing the association between skin diseases and metabolic syndrome and a clinical decision algorithm for high risk patients. Diabetol Metab Syndr. 2018;10:9. 
  118. Mann N. Meat in the human diet: an anthropological perspective. Nutr Dietetics. 2007;64(suppl 4):S102-S107. 
  119. Bramble DM, Lieberman DE. Endurance running and the evolution of Homo. Nature. 2004;432:345-352. 
  120. Kuhn JE. Throwing, the shoulder, and human evolution. Am J Orthop (Belle Mead NJ). 2016;45:110-114. 
  121. Kobayashi H, Kohshima S. Unique morphology of the human eye and its adaptive meaning: comparative studies on external morphology of the primate eye. J Hum Evol. 2001;40:419-435. 
  122. Cordain L, Eaton SB, Miller JB, et al. The paradoxical nature of hunter-gatherer diets: meat-based, yet non-atherogenic. Eur J Clin Nutr. 2002;56(suppl 1):S42-S52. 
  123. McClellan WS, Du Bois EF. Clinical calorimetry: XLV. prolonged meat diets with a study of kidney function and ketosis. J Biol Chem. 1930;87:651-668. 
  124. O'Hearn A. Can a carnivore diet provide all essential nutrients? Curr Opin Endocrinol Diabetes Obes. 2020;27:312-316. 
  125. O'Hearn LA. A survey of improvements experienced on a carnivore diet compared to only carbohydrate restriction. Open Science Forum website. Published February 12, 2019. Accessed May 17, 2021. doi:10.17605/OSF.IO/5FU4D 
  126. Williams P. Nutritional composition of red meat. Nutrition & Dietetics. 2007;64(suppl 4):S113-S119. 
  127. Biel W, Czerniawska-Piątkowska E, Kowalczyk A. Offal chemical composition from veal, beef, and lamb maintained in organic production systems. Animals (Basel). 2019;9:489. 
  128. Elmadfa I, Meyer AL. The role of the status of selected micronutrients in shaping the immune function. Endocr Metab Immune Disord Drug Targets. 2019;19:1100-1115. 
  129. Babizhayev M. Treatment of skin aging and photoaging with innovative oral dosage forms of nonhydrolized carnosine and carcinine. Int J Clin Derm Res. 2017;5:116-143. 
  130. Danby FW. Nutrition and aging skin: sugar and glycation. Clin Dermatol. 2010;28:409-411. 
  131. Siefken W, Carstensen S, Springmann G, et al. Role of taurine accumulation in keratinocyte hydration. J Invest Dermatol. 2003;121:354-361. 
  132. Vollmer DL, West VA, Lephart ED. Enhancing skin health: by oral administration of natural compounds and minerals with implications to the dermal microbiome. Int J Mol Sci. 2018;19:3059. 
  133. Fischer F, Achterberg V, März A, et al. Folic acid and creatineimprove the firmness of human skin in vivo. J Cosmet Dermatol. 2011;10:15-23. 
  134. Blatt T, Lenz H, Weber T. Topical application of creatine is multibeneficial for human skin. J Am Acad Dermatol. 2005;52:P32.
Article PDF
Author and Disclosure Information

Mr. Svoboda is from the Virginia Tech Carilion School of Medicine, Roanoke. Dr. Christopher is from Ironwood Dermatology and Aesthetic Services, Tucson, Arizona. Dr. Shields is from the Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison.

The authors report no conflicts of interest.

Correspondence: Bridget E. Shields, MD, 1 S Park St, University of Wisconsin School of Medicine and Public Health, Department of Dermatology, Madison, WI 53711 (bshields@dermatology.wisc.edu).

Issue
cutis - 107(6)
Publications
Topics
Page Number
308-314
Sections
Author and Disclosure Information

Mr. Svoboda is from the Virginia Tech Carilion School of Medicine, Roanoke. Dr. Christopher is from Ironwood Dermatology and Aesthetic Services, Tucson, Arizona. Dr. Shields is from the Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison.

The authors report no conflicts of interest.

Correspondence: Bridget E. Shields, MD, 1 S Park St, University of Wisconsin School of Medicine and Public Health, Department of Dermatology, Madison, WI 53711 (bshields@dermatology.wisc.edu).

Author and Disclosure Information

Mr. Svoboda is from the Virginia Tech Carilion School of Medicine, Roanoke. Dr. Christopher is from Ironwood Dermatology and Aesthetic Services, Tucson, Arizona. Dr. Shields is from the Department of Dermatology, University of Wisconsin School of Medicine and Public Health, Madison.

The authors report no conflicts of interest.

Correspondence: Bridget E. Shields, MD, 1 S Park St, University of Wisconsin School of Medicine and Public Health, Department of Dermatology, Madison, WI 53711 (bshields@dermatology.wisc.edu).

Article PDF
Article PDF

Within the last decade, almost 3000 articles have been published on the role of diet in the prevention and management of dermatologic conditions. Patients are increasingly interested in—and employing—dietary modifications that may influence skin appearance and aid in the treatment of cutaneous disease.1 It is essential that dermatologists are familiar with existing evidence on the role of diet in dermatology to counsel patients appropriately. Herein, we discuss the compositions of several popular diets and their proposed utility for dermatologic purposes. We highlight the limited literature that exists surrounding this topic and emphasize the need for future, well-designed clinical trials that study the impact of diet on skin disease.

Ketogenic Diet

The ketogenic diet has a macronutrient profile composed of high fat, low to moderate protein, and very low carbohydrates. Nutritional ketosis occurs as the body begins to use free fatty acids (via beta oxidation) as the primary metabolite driving cellular metabolism. It has been suggested that the ketogenic diet may impart beneficial effects on skin disease; however, limited literature exists on the role of nutritional ketosis in the treatment of dermatologic conditions.

Mechanistically, the ketogenic diet decreases the secretion of insulin and insulinlike growth factor 1, resulting in a reduction of circulating androgens and increased activity of the retinoid X receptor.2 In acne vulgaris, it has been suggested that the ketogenic diet may be beneficial in decreasing androgen-induced sebum production and the overproliferation of keratinocytes.2-7 The ketogenic diet is one of the most rapidly effective dietary strategies for normalizing both insulin and androgens, thus it may theoretically be useful for other metabolic and hormone-dependent skin diseases, such as hidradenitis suppurativa.8,9

The cutaneous manifestations associated with chronic hyperinsulinemia and hyperglycemia are numerous and include acanthosis nigricans, acrochordons, diabetic dermopathy, scleredema diabeticorum, bullosis diabeticorum, keratosis pilaris, and generalized granuloma annulare. There also is an increased risk for bacterial and fungal skin infections associated with hyperglycemic states.10 The ketogenic diet is an effective nonpharmacologic tool for normalizing serum insulin and glucose levels in most patients and may have utility in the aforementioned conditions.11,12 In addition to improving insulin sensitivity, it has been used as a dietary strategy for weight loss.11-15 Because obesity and metabolic syndrome are highly correlated with common skin conditions such as psoriasis, hidradenitis suppurativa, and androgenetic alopecia, there may be a role for employing the ketogenic diet in these patient populations.16,17

Although robust clinical studies on ketogenic diets in skin disease are lacking, a recent single-arm, open-label clinical trial observed benefit in all 37 drug-naïve, overweight patients with chronic plaque psoriasis who underwent a ketogenic weight loss protocol. Significant reductions in psoriasis area and severity index (PASI) score and dermatology life quality index score were reported (P<.001).18 Another study of 30 patients with psoriasis found that a 4-week, low-calorie, ketogenic diet resulted in 50% improvement of PASI scores, 10% weight loss, and a reduction in the proinflammatory cytokines IL-1β and IL-2.19 Despite these results, it is a challenge to tease out if the specific dietary intervention or its associated weight loss was the main driver in these reported improvements in skin disease.

There is mixed evidence on the anti-inflammatory nature of the ketogenic diet, likely due to wide variation in the composition of foods included in individual diets. In many instances, the ketogenic diet is thought to possess considerable antioxidant and anti-inflammatory capabilities. Ketones are known activators of the nuclear factor erythroid 2–related factor 2 pathway, which upregulates the production of glutathione, a major endogenous intracellular antioxidant.20 Additionally, dietary compounds from foods that are encouraged while on the ketogenic diet, such as sulforaphane from broccoli, also are independent activators of nuclear factor erythroid 2–related factor 2.21 Ketones are efficiently utilized by mitochondria, which also may result in the decreased production of reactive oxygen species and lower oxidative stress.22 Moreover, the ketone body β-hydroxybutyrate has demonstrated the ability to reduce proinflammatory IL-1β levels via suppression of nucleotide-binding domain-like receptor protein 3 inflammasome activity.23,24 The activity of IL-1β is known to be elevated in many dermatologic conditions, including juvenile idiopathic arthritis, relapsing polychondritis, Schnitzler syndrome, hidradenitis suppurativa, Behçet disease, and other autoinflammatory syndromes.25 Ketones also have been shown to inhibit the nuclear factor–κB proinflammatory signaling pathway.22,26,27 Overexpression of IL-1β and aberrant activation of nuclear factor–κB are implicated in a variety of inflammatory, autoimmune, and oncologic cutaneous pathologies. The ketogenic diet may prove to be an effective adjunctive treatment for dermatologists to consider in select patient populations.23,24,28-30



For patients with keratinocyte carcinomas, the ketogenic diet may offer the aforementioned anti-inflammatory and antioxidant effects, as well as suppression of the mechanistic target of rapamycin, a major regulator of cell metabolism and proliferation.31,32 Inhibition of mechanistic target of rapamycin activity has been shown to slow tumor growth and reduce the development of squamous cell carcinoma.25,33,34 The ketogenic diet also may exploit the preferential utilization of glucose exhibited by many types of cancer cells, thereby “starving” the tumor of its primary fuel source.35,36 In vitro and animal studies in a variety of cancer types have demonstrated that a ketogenic metabolic state—achieved through the ketogenic diet or fasting—can sensitize tumor cells to chemotherapy and radiation while conferring a protective effect to normal cells.37-40 This recently described phenomenon is known as differential stress resistance, but it has not been studied in keratinocyte malignancies or melanoma to date. Importantly, some basal cell carcinomas and BRAF V600E–mutated melanomas have worsened while on the ketogenic diet, suggesting more data is needed before it can be recommended for all cancer patients.41,42 Furthermore, other skin conditions such as prurigo pigmentosa have been associated with initiation of the ketogenic diet.43

 

 

Low FODMAP Diet

Fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) are short-chain carbohydrates that are poorly absorbed, osmotically active, and rapidly fermented by intestinal bacteria.44 The low FODMAP diet has been shown to be efficacious for treatment of irritable bowel syndrome, small intestinal bacterial overgrowth (SIBO), and some cases of inflammatory bowel disease (IBD).44-49 A low FODMAP diet may have potential implications for several dermatologic conditions.

Rosacea has been associated with various gastrointestinal tract disorders including irritable bowel syndrome, SIBO, and IBD.50-54 A single study found that patients with rosacea had a 13-fold increased risk for SIBO.55,56 Treatment of 40 patients with SIBO using rifaximin resulted in complete resolution of rosacea in all patients, with no relapse after a 3-year follow-up period.55 Psoriasis also has been associated with SIBO and IBD.57,58 One small study found that eradication of SIBO in psoriatic patients resulted in improved PASI scores and colorimetric values.59

Although the long-term health consequences of the low FODMAP diet are unknown, further research on such dietary interventions for inflammatory skin conditions is warranted given the mounting evidence of a gut-skin connection and the role of the intestinal microbiome in skin health.50,51

Gluten-Free Diet

Gluten is a protein found in a variety of grains. Although the role of gluten in the pathogenesis of celiac disease and dermatitis herpetiformis is indisputable, the deleterious effects of gluten outside of the context of these diseases remain controversial. There may be a compelling case for eliminating gluten in psoriasis patients with seropositivity for celiac disease. A recent systematic review found a 2.2-fold increased risk for celiac disease in psoriasis patients.60 Antigliadin antibody titers also were found to be positively correlated with psoriatic disease severity.61 In addition, one open-label study found a reduction in PASI scores in 73% of patients with antigliadin antibodies after 3 months on a gluten-free diet compared to those without antibodies; however, the study only included 22 patients.62 Several other small studies have yielded similar results63,64; however, antigliadin antibodies are neither the most sensitive nor specific markers of celiac disease, and additional testing should be completed in any patient who may carry this diagnosis. A survey study by the National Psoriasis Foundation found that the dietary change associated with the greatest skin improvement was removal of gluten and nightshade vegetables in approximately 50% of the 1200 psoriasis patients that responded.65 Case reports of various dermatologic conditions including sarcoidosis, vitiligo, alopecia areata, lichen planus, dermatomyositis, pyoderma gangrenosum, erythema nodosum, leukocytoclastic vasculitis, linear IgA bullous dermatosis, and aphthous ulcerations have reportedly improved with a gluten-free diet; however, this should not be used as primary therapy in patients without celiac disease.66-71 Because gluten-free diets can be expensive and challenging to follow, a formal assessment for celiac disease should be considered before recommendation of this dietary intervention.

Low Histamine Diet

Histamine is a biogenic amine produced by the decarboxylation of the amino acid histidine.72 It is found in several foods in varying amounts. Because bacteria can convert histidine into histamine, many fermented and aged foods such as kimchi, sauerkraut, cheese, and red wine contain high levels of histamine. Individuals who have decreased activity of diamine oxidase (DAO), an enzyme that degrades histamine, may be more susceptible to histamine intolerance.72 The symptoms of histamine intolerance are numerous and include gastrointestinal tract distress, rhinorrhea and nasal congestion, headache, urticaria, flushing, and pruritus. Histamine intolerance can mimic an IgE-mediated food allergy; however, allergy testing is negative in these patients. Unfortunately, there is no laboratory test for histamine intolerance; a double-blind, placebo-controlled food challenge is considered the gold-standard test.72

As it pertains to dermatology, a low histamine diet may play a role in the treatment of certain patients with atopic dermatitis and chronic spontaneous urticaria. One study reported that 17 of 54 (31.5%) atopic patients had higher basal levels of serum histamine compared to controls.73 Another study found that a histamine-free diet led to improvement in both histamine intolerance symptoms and atopic dermatitis disease severity (SCORing atopic dermatitis) in patients with low DAO activity.74 In chronic spontaneous urticaria, a recent systematic review found that in 223 patients placed on a low histamine diet for 3 to 4 weeks, 12% and 44% achieved complete and partial remission, respectively.75 Although treatment response based on a patient’s DAO activity level has not been correlated, a diet low in histamine may prove useful for patients with persistent atopic dermatitis and chronic spontaneous urticaria who have negative food allergy tests and report exacerbation of symptoms after ingestion of histamine-rich foods.76,77

Mediterranean Diet

The Mediterranean diet has been touted as one of the healthiest diets to date, and large randomized clinical trials have demonstrated its effectiveness in weight loss, improving insulin sensitivity, and reducing inflammatory cytokine profiles.78,79 A major criticism of the Mediterranean diet is that it has considerable ambiguity and lacks a precise definition due to the variability of what is consumed in different Mediterranean regions. Generally, the diet emphasizes high consumption of colorful fruits and vegetables, aromatic herbs and spices, olive oil, nuts, and seafood, as well as modest amounts of dairy, eggs, and red meat.80 The anti-inflammatory effects of this diet largely have been attributed to its abundance of polyphenols, carotenoids, monounsaturated fatty acids, and omega-3 polyunsaturated fatty acids (PUFAs).80,81 Examples of polyphenols include resveratrol in red grapes, quercetin in apples and red onions, and curcumin in turmeric, while examples of carotenoids include lycopene in tomatoes and zeaxanthin in dark leafy greens. Oleic acid is a monounsaturated fatty acid present in high concentrations in olive oil, while eicosapentaenoic acid and docosahexaenoic acid are omega-3 PUFAs predominantly found in fish.82

Unfortunately, rigorous clinical trials regarding the Mediterranean diet as it pertains to dermatology have not been undertaken. Numerous observational studies in patients with psoriasis have suggested that close adherence to the Mediterranean diet was associated with improvement in PASI scores.83-86 The National Psoriasis Foundation now recommends a trial of the Mediterranean diet in some patients with psoriasis, emphasizing increased dietary intake of olive oil, fish, and vegetables.87 Adherence to a Mediterranean diet also has been inversely correlated to the severity of acne vulgaris and hidradenitis suppurativa88,89; however, these studies failed to account for the multifactorial risk factors associated with these conditions. Mediterranean diets also may impart a chemopreventive effect, supported by a number of in vivo and in vitro studies demonstrating the inhibition and/or reversal of cutaneous DNA damage induced by UV radiation through supplementation with various phytonutrients and omega-3 PUFAs.81,90-92 Although small case-control studies have found a decreased risk of basal cell carcinoma in those who closely adhered to a Mediterranean diet, more rigorous clinical research is needed.93

 

 

Whole-Food, Plant-Based Diet

A whole-food, plant-based (WFPB) diet is another popular dietary approach that consists of eating fruits, vegetables, legumes, nuts, seeds, and grains in their whole natural form.94 This diet discourages all animal products, including red meat, seafood, dairy, and eggs. It is similar to a vegan diet except that it eliminates all highly refined carbohydrates, vegetable oils, and other processed foods.94 Randomized clinical studies have demonstrated the WFPB diet to be effective in the treatment of obesity and metabolic syndrome.95,96

A WFPB diet has been shown to increase the antioxidant capacity of cells, lengthen telomeres, and reduce formation of advanced glycation end products.94,97,98 These benefits may help combat accelerated skin aging, including increased skin permeability, reduced elasticity and hydration, decreased angiogenesis, impaired immune function, and decreased vitamin D synthesis. Accelerated skin aging can result in delayed wound healing and susceptibility to skin tears and ecchymoses and also may promote the development of cutaneous malignancies.99 There remains a lack of clinical data studying a properly formulated WFPB diet in the dermatologic setting.

Paleolithic Diet

The paleolithic (Paleo) diet is an increasingly popular way of eating that attempts to mirror what our ancestors may have consumed between 10,000 and 2.5 million years ago.100 It is similar to the Mediterranean diet but excludes grains, dairy, legumes, and nightshade vegetables. It also calls for elimination of highly processed sugars and oils as well as chemical food additives and preservatives. There is a strict variation of the diet for individuals with autoimmune disease that also excludes eggs, nuts, and seeds, as these can be inflammatory or immunogenic in some patients.100-106 Other variations of the diet exist, including the ketogenic Paleo diet, pegan (Paleo vegan) diet, and lacto-Paleo diet.100 An often cited criticism of the Paleo diet is the low intake of calcium and risk for osteoporosis; however, consumption of calcium-rich foods or a calcium supplement can address this concern.107

Although small clinical studies have found the Paleo diet to be beneficial for various autoimmune diseases, clinical data evaluating the utility of the diet for cutaneous disease is lacking.108,109 Numerous randomized trials have demonstrated the Paleo diet to be effective for weight loss and improving insulin sensitivity and lipid levels.110-116 Thus, the Paleo diet may theoretically serve as a viable adjunct dietary approach to the treatment of cutaneous diseases associated with obesity and metabolic derangement.117

Carnivore Diet

Arguably the most controversial and radical diet is the carnivore diet. As the name implies, the carnivore diet is based on consuming solely animal products. A properly structured carnivore diet emphasizes a “nose-to-tail” eating approach where all parts of the animal including the muscle meats, organs, and fat are consumed. Proponents of the diet cite anthropologic evidence from fossil-stable carbon-13/carbon-12 isotope analyses, craniodental features, and numerous other adaptations that indicate increased consumption of meat during human evolution.118-122 Notably, many early humans ate a carnivore diet, but life span was very short at this time, suggesting the diet may not be as beneficial as has been suggested.

Despite the abundance of anecdotal evidence supporting its use for a variety of chronic conditions, including cutaneous autoimmune disease, there is a virtual absence of high-quality research on the carnivore diet.123-125



The purported benefits of the carnivore diet may be attributed to the consumption of organ meats that contain highly bioavailable essential vitamins and minerals, such as iron, zinc, copper, selenium, thiamine, niacin, folate, vitamin B6, vitamin B12, vitamin A, vitamin D, vitamin K, and choline.126-128 Other dietary compounds that have demonstrated benefit for skin health and are predominantly found in animal foods include carnosine, carnitine, creatine, taurine, coenzyme Q10, and collagen.129-134 Nevertheless, there is no data to recommend the elimination of antioxidant- and micronutrient-dense plant-based foods. Rigorous clinical research evaluating the efficacy and safety of the carnivore diet in dermatologic patients is needed. A carnivore diet should not be undertaken without the assistance of a dietician who can ensure adequate micronutrient and macronutrient support.

Final Thoughts

The adjunctive role of diet in the treatment of skin disease is expanding and becoming more widely accepted among dermatologists. Unfortunately, there remains a lack of randomized controlled trials confirming the efficacy of various dietary interventions in the dermatologic setting. Although evidence-based dietary recommendations currently are limited, it is important for dermatologists to be aware of the varied and nuanced dietary interventions employed by patients.

Ultimately, dietary recommendations must be personalized, considering a patient’s comorbidities, personal beliefs and preferences, and nutrigenetics. The emerging field of dermatonutrigenomics—the study of how dietary compounds interact with one’s genes to influence skin health—may allow for precise dietary recommendations to be made in dermatologic practice. Direct-to-consumer genetic tests targeted toward dermatology patients are already on the market, but their clinical utility awaits validation.1 Because nutritional science is a constantly evolving field, becoming familiar with these popular diets will serve both dermatologists and their patients well.

Within the last decade, almost 3000 articles have been published on the role of diet in the prevention and management of dermatologic conditions. Patients are increasingly interested in—and employing—dietary modifications that may influence skin appearance and aid in the treatment of cutaneous disease.1 It is essential that dermatologists are familiar with existing evidence on the role of diet in dermatology to counsel patients appropriately. Herein, we discuss the compositions of several popular diets and their proposed utility for dermatologic purposes. We highlight the limited literature that exists surrounding this topic and emphasize the need for future, well-designed clinical trials that study the impact of diet on skin disease.

Ketogenic Diet

The ketogenic diet has a macronutrient profile composed of high fat, low to moderate protein, and very low carbohydrates. Nutritional ketosis occurs as the body begins to use free fatty acids (via beta oxidation) as the primary metabolite driving cellular metabolism. It has been suggested that the ketogenic diet may impart beneficial effects on skin disease; however, limited literature exists on the role of nutritional ketosis in the treatment of dermatologic conditions.

Mechanistically, the ketogenic diet decreases the secretion of insulin and insulinlike growth factor 1, resulting in a reduction of circulating androgens and increased activity of the retinoid X receptor.2 In acne vulgaris, it has been suggested that the ketogenic diet may be beneficial in decreasing androgen-induced sebum production and the overproliferation of keratinocytes.2-7 The ketogenic diet is one of the most rapidly effective dietary strategies for normalizing both insulin and androgens, thus it may theoretically be useful for other metabolic and hormone-dependent skin diseases, such as hidradenitis suppurativa.8,9

The cutaneous manifestations associated with chronic hyperinsulinemia and hyperglycemia are numerous and include acanthosis nigricans, acrochordons, diabetic dermopathy, scleredema diabeticorum, bullosis diabeticorum, keratosis pilaris, and generalized granuloma annulare. There also is an increased risk for bacterial and fungal skin infections associated with hyperglycemic states.10 The ketogenic diet is an effective nonpharmacologic tool for normalizing serum insulin and glucose levels in most patients and may have utility in the aforementioned conditions.11,12 In addition to improving insulin sensitivity, it has been used as a dietary strategy for weight loss.11-15 Because obesity and metabolic syndrome are highly correlated with common skin conditions such as psoriasis, hidradenitis suppurativa, and androgenetic alopecia, there may be a role for employing the ketogenic diet in these patient populations.16,17

Although robust clinical studies on ketogenic diets in skin disease are lacking, a recent single-arm, open-label clinical trial observed benefit in all 37 drug-naïve, overweight patients with chronic plaque psoriasis who underwent a ketogenic weight loss protocol. Significant reductions in psoriasis area and severity index (PASI) score and dermatology life quality index score were reported (P<.001).18 Another study of 30 patients with psoriasis found that a 4-week, low-calorie, ketogenic diet resulted in 50% improvement of PASI scores, 10% weight loss, and a reduction in the proinflammatory cytokines IL-1β and IL-2.19 Despite these results, it is a challenge to tease out if the specific dietary intervention or its associated weight loss was the main driver in these reported improvements in skin disease.

There is mixed evidence on the anti-inflammatory nature of the ketogenic diet, likely due to wide variation in the composition of foods included in individual diets. In many instances, the ketogenic diet is thought to possess considerable antioxidant and anti-inflammatory capabilities. Ketones are known activators of the nuclear factor erythroid 2–related factor 2 pathway, which upregulates the production of glutathione, a major endogenous intracellular antioxidant.20 Additionally, dietary compounds from foods that are encouraged while on the ketogenic diet, such as sulforaphane from broccoli, also are independent activators of nuclear factor erythroid 2–related factor 2.21 Ketones are efficiently utilized by mitochondria, which also may result in the decreased production of reactive oxygen species and lower oxidative stress.22 Moreover, the ketone body β-hydroxybutyrate has demonstrated the ability to reduce proinflammatory IL-1β levels via suppression of nucleotide-binding domain-like receptor protein 3 inflammasome activity.23,24 The activity of IL-1β is known to be elevated in many dermatologic conditions, including juvenile idiopathic arthritis, relapsing polychondritis, Schnitzler syndrome, hidradenitis suppurativa, Behçet disease, and other autoinflammatory syndromes.25 Ketones also have been shown to inhibit the nuclear factor–κB proinflammatory signaling pathway.22,26,27 Overexpression of IL-1β and aberrant activation of nuclear factor–κB are implicated in a variety of inflammatory, autoimmune, and oncologic cutaneous pathologies. The ketogenic diet may prove to be an effective adjunctive treatment for dermatologists to consider in select patient populations.23,24,28-30



For patients with keratinocyte carcinomas, the ketogenic diet may offer the aforementioned anti-inflammatory and antioxidant effects, as well as suppression of the mechanistic target of rapamycin, a major regulator of cell metabolism and proliferation.31,32 Inhibition of mechanistic target of rapamycin activity has been shown to slow tumor growth and reduce the development of squamous cell carcinoma.25,33,34 The ketogenic diet also may exploit the preferential utilization of glucose exhibited by many types of cancer cells, thereby “starving” the tumor of its primary fuel source.35,36 In vitro and animal studies in a variety of cancer types have demonstrated that a ketogenic metabolic state—achieved through the ketogenic diet or fasting—can sensitize tumor cells to chemotherapy and radiation while conferring a protective effect to normal cells.37-40 This recently described phenomenon is known as differential stress resistance, but it has not been studied in keratinocyte malignancies or melanoma to date. Importantly, some basal cell carcinomas and BRAF V600E–mutated melanomas have worsened while on the ketogenic diet, suggesting more data is needed before it can be recommended for all cancer patients.41,42 Furthermore, other skin conditions such as prurigo pigmentosa have been associated with initiation of the ketogenic diet.43

 

 

Low FODMAP Diet

Fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) are short-chain carbohydrates that are poorly absorbed, osmotically active, and rapidly fermented by intestinal bacteria.44 The low FODMAP diet has been shown to be efficacious for treatment of irritable bowel syndrome, small intestinal bacterial overgrowth (SIBO), and some cases of inflammatory bowel disease (IBD).44-49 A low FODMAP diet may have potential implications for several dermatologic conditions.

Rosacea has been associated with various gastrointestinal tract disorders including irritable bowel syndrome, SIBO, and IBD.50-54 A single study found that patients with rosacea had a 13-fold increased risk for SIBO.55,56 Treatment of 40 patients with SIBO using rifaximin resulted in complete resolution of rosacea in all patients, with no relapse after a 3-year follow-up period.55 Psoriasis also has been associated with SIBO and IBD.57,58 One small study found that eradication of SIBO in psoriatic patients resulted in improved PASI scores and colorimetric values.59

Although the long-term health consequences of the low FODMAP diet are unknown, further research on such dietary interventions for inflammatory skin conditions is warranted given the mounting evidence of a gut-skin connection and the role of the intestinal microbiome in skin health.50,51

Gluten-Free Diet

Gluten is a protein found in a variety of grains. Although the role of gluten in the pathogenesis of celiac disease and dermatitis herpetiformis is indisputable, the deleterious effects of gluten outside of the context of these diseases remain controversial. There may be a compelling case for eliminating gluten in psoriasis patients with seropositivity for celiac disease. A recent systematic review found a 2.2-fold increased risk for celiac disease in psoriasis patients.60 Antigliadin antibody titers also were found to be positively correlated with psoriatic disease severity.61 In addition, one open-label study found a reduction in PASI scores in 73% of patients with antigliadin antibodies after 3 months on a gluten-free diet compared to those without antibodies; however, the study only included 22 patients.62 Several other small studies have yielded similar results63,64; however, antigliadin antibodies are neither the most sensitive nor specific markers of celiac disease, and additional testing should be completed in any patient who may carry this diagnosis. A survey study by the National Psoriasis Foundation found that the dietary change associated with the greatest skin improvement was removal of gluten and nightshade vegetables in approximately 50% of the 1200 psoriasis patients that responded.65 Case reports of various dermatologic conditions including sarcoidosis, vitiligo, alopecia areata, lichen planus, dermatomyositis, pyoderma gangrenosum, erythema nodosum, leukocytoclastic vasculitis, linear IgA bullous dermatosis, and aphthous ulcerations have reportedly improved with a gluten-free diet; however, this should not be used as primary therapy in patients without celiac disease.66-71 Because gluten-free diets can be expensive and challenging to follow, a formal assessment for celiac disease should be considered before recommendation of this dietary intervention.

Low Histamine Diet

Histamine is a biogenic amine produced by the decarboxylation of the amino acid histidine.72 It is found in several foods in varying amounts. Because bacteria can convert histidine into histamine, many fermented and aged foods such as kimchi, sauerkraut, cheese, and red wine contain high levels of histamine. Individuals who have decreased activity of diamine oxidase (DAO), an enzyme that degrades histamine, may be more susceptible to histamine intolerance.72 The symptoms of histamine intolerance are numerous and include gastrointestinal tract distress, rhinorrhea and nasal congestion, headache, urticaria, flushing, and pruritus. Histamine intolerance can mimic an IgE-mediated food allergy; however, allergy testing is negative in these patients. Unfortunately, there is no laboratory test for histamine intolerance; a double-blind, placebo-controlled food challenge is considered the gold-standard test.72

As it pertains to dermatology, a low histamine diet may play a role in the treatment of certain patients with atopic dermatitis and chronic spontaneous urticaria. One study reported that 17 of 54 (31.5%) atopic patients had higher basal levels of serum histamine compared to controls.73 Another study found that a histamine-free diet led to improvement in both histamine intolerance symptoms and atopic dermatitis disease severity (SCORing atopic dermatitis) in patients with low DAO activity.74 In chronic spontaneous urticaria, a recent systematic review found that in 223 patients placed on a low histamine diet for 3 to 4 weeks, 12% and 44% achieved complete and partial remission, respectively.75 Although treatment response based on a patient’s DAO activity level has not been correlated, a diet low in histamine may prove useful for patients with persistent atopic dermatitis and chronic spontaneous urticaria who have negative food allergy tests and report exacerbation of symptoms after ingestion of histamine-rich foods.76,77

Mediterranean Diet

The Mediterranean diet has been touted as one of the healthiest diets to date, and large randomized clinical trials have demonstrated its effectiveness in weight loss, improving insulin sensitivity, and reducing inflammatory cytokine profiles.78,79 A major criticism of the Mediterranean diet is that it has considerable ambiguity and lacks a precise definition due to the variability of what is consumed in different Mediterranean regions. Generally, the diet emphasizes high consumption of colorful fruits and vegetables, aromatic herbs and spices, olive oil, nuts, and seafood, as well as modest amounts of dairy, eggs, and red meat.80 The anti-inflammatory effects of this diet largely have been attributed to its abundance of polyphenols, carotenoids, monounsaturated fatty acids, and omega-3 polyunsaturated fatty acids (PUFAs).80,81 Examples of polyphenols include resveratrol in red grapes, quercetin in apples and red onions, and curcumin in turmeric, while examples of carotenoids include lycopene in tomatoes and zeaxanthin in dark leafy greens. Oleic acid is a monounsaturated fatty acid present in high concentrations in olive oil, while eicosapentaenoic acid and docosahexaenoic acid are omega-3 PUFAs predominantly found in fish.82

Unfortunately, rigorous clinical trials regarding the Mediterranean diet as it pertains to dermatology have not been undertaken. Numerous observational studies in patients with psoriasis have suggested that close adherence to the Mediterranean diet was associated with improvement in PASI scores.83-86 The National Psoriasis Foundation now recommends a trial of the Mediterranean diet in some patients with psoriasis, emphasizing increased dietary intake of olive oil, fish, and vegetables.87 Adherence to a Mediterranean diet also has been inversely correlated to the severity of acne vulgaris and hidradenitis suppurativa88,89; however, these studies failed to account for the multifactorial risk factors associated with these conditions. Mediterranean diets also may impart a chemopreventive effect, supported by a number of in vivo and in vitro studies demonstrating the inhibition and/or reversal of cutaneous DNA damage induced by UV radiation through supplementation with various phytonutrients and omega-3 PUFAs.81,90-92 Although small case-control studies have found a decreased risk of basal cell carcinoma in those who closely adhered to a Mediterranean diet, more rigorous clinical research is needed.93

 

 

Whole-Food, Plant-Based Diet

A whole-food, plant-based (WFPB) diet is another popular dietary approach that consists of eating fruits, vegetables, legumes, nuts, seeds, and grains in their whole natural form.94 This diet discourages all animal products, including red meat, seafood, dairy, and eggs. It is similar to a vegan diet except that it eliminates all highly refined carbohydrates, vegetable oils, and other processed foods.94 Randomized clinical studies have demonstrated the WFPB diet to be effective in the treatment of obesity and metabolic syndrome.95,96

A WFPB diet has been shown to increase the antioxidant capacity of cells, lengthen telomeres, and reduce formation of advanced glycation end products.94,97,98 These benefits may help combat accelerated skin aging, including increased skin permeability, reduced elasticity and hydration, decreased angiogenesis, impaired immune function, and decreased vitamin D synthesis. Accelerated skin aging can result in delayed wound healing and susceptibility to skin tears and ecchymoses and also may promote the development of cutaneous malignancies.99 There remains a lack of clinical data studying a properly formulated WFPB diet in the dermatologic setting.

Paleolithic Diet

The paleolithic (Paleo) diet is an increasingly popular way of eating that attempts to mirror what our ancestors may have consumed between 10,000 and 2.5 million years ago.100 It is similar to the Mediterranean diet but excludes grains, dairy, legumes, and nightshade vegetables. It also calls for elimination of highly processed sugars and oils as well as chemical food additives and preservatives. There is a strict variation of the diet for individuals with autoimmune disease that also excludes eggs, nuts, and seeds, as these can be inflammatory or immunogenic in some patients.100-106 Other variations of the diet exist, including the ketogenic Paleo diet, pegan (Paleo vegan) diet, and lacto-Paleo diet.100 An often cited criticism of the Paleo diet is the low intake of calcium and risk for osteoporosis; however, consumption of calcium-rich foods or a calcium supplement can address this concern.107

Although small clinical studies have found the Paleo diet to be beneficial for various autoimmune diseases, clinical data evaluating the utility of the diet for cutaneous disease is lacking.108,109 Numerous randomized trials have demonstrated the Paleo diet to be effective for weight loss and improving insulin sensitivity and lipid levels.110-116 Thus, the Paleo diet may theoretically serve as a viable adjunct dietary approach to the treatment of cutaneous diseases associated with obesity and metabolic derangement.117

Carnivore Diet

Arguably the most controversial and radical diet is the carnivore diet. As the name implies, the carnivore diet is based on consuming solely animal products. A properly structured carnivore diet emphasizes a “nose-to-tail” eating approach where all parts of the animal including the muscle meats, organs, and fat are consumed. Proponents of the diet cite anthropologic evidence from fossil-stable carbon-13/carbon-12 isotope analyses, craniodental features, and numerous other adaptations that indicate increased consumption of meat during human evolution.118-122 Notably, many early humans ate a carnivore diet, but life span was very short at this time, suggesting the diet may not be as beneficial as has been suggested.

Despite the abundance of anecdotal evidence supporting its use for a variety of chronic conditions, including cutaneous autoimmune disease, there is a virtual absence of high-quality research on the carnivore diet.123-125



The purported benefits of the carnivore diet may be attributed to the consumption of organ meats that contain highly bioavailable essential vitamins and minerals, such as iron, zinc, copper, selenium, thiamine, niacin, folate, vitamin B6, vitamin B12, vitamin A, vitamin D, vitamin K, and choline.126-128 Other dietary compounds that have demonstrated benefit for skin health and are predominantly found in animal foods include carnosine, carnitine, creatine, taurine, coenzyme Q10, and collagen.129-134 Nevertheless, there is no data to recommend the elimination of antioxidant- and micronutrient-dense plant-based foods. Rigorous clinical research evaluating the efficacy and safety of the carnivore diet in dermatologic patients is needed. A carnivore diet should not be undertaken without the assistance of a dietician who can ensure adequate micronutrient and macronutrient support.

Final Thoughts

The adjunctive role of diet in the treatment of skin disease is expanding and becoming more widely accepted among dermatologists. Unfortunately, there remains a lack of randomized controlled trials confirming the efficacy of various dietary interventions in the dermatologic setting. Although evidence-based dietary recommendations currently are limited, it is important for dermatologists to be aware of the varied and nuanced dietary interventions employed by patients.

Ultimately, dietary recommendations must be personalized, considering a patient’s comorbidities, personal beliefs and preferences, and nutrigenetics. The emerging field of dermatonutrigenomics—the study of how dietary compounds interact with one’s genes to influence skin health—may allow for precise dietary recommendations to be made in dermatologic practice. Direct-to-consumer genetic tests targeted toward dermatology patients are already on the market, but their clinical utility awaits validation.1 Because nutritional science is a constantly evolving field, becoming familiar with these popular diets will serve both dermatologists and their patients well.

References
  1. Jaros J, Katta R, Shi VY. Dermatonutrigenomics: past, present, and future. Dermatology. 2019;235:164-166. 
  2. Paoli A, Grimaldi K, Toniolo L, et al. Nutrition and acne: therapeutic potential of ketogenic diets. Skin Pharmacol Physiol. 2012;25:111-117. 
  3. Melnik BC, Schmitz G. Role of insulin, insulin-like growth factor-1, hyperglycaemic food and milk consumption in the pathogenesis of acne vulgaris. Exp Dermatol. 2009;18:833-841. 
  4. Smith RN, Mann NJ, Braue A, et al. The effect of a high-protein, low glycemic-load diet versus a conventional, high glycemic-load diet on biochemical parameters associated with acne vulgaris: a randomized, investigator-masked, controlled trial. J Am Acad Dermatol. 2007;57:247-256. 
  5. Smith R, Mann N, Mäkeläinen H, et al. A pilot study to determine the short-term effects of a low glycemic load diet on hormonal markers of acne: a nonrandomized, parallel, controlled feeding trial. Mol Nutr Food Res. 2008;52:718-726. 
  6. Smith RN, Braue A, Varigos GA, et al. The effect of a low glycemic load diet on acne vulgaris and the fatty acid composition of skin surface triglycerides. J Dermatol Sci. 2008;50:41-52. 
  7. Kwon HH, Yoon JY, Hong JS, et al. Clinical and histological effect of a low glycaemic load diet in treatment of acne vulgaris in Korean patients: a randomized, controlled trial. Acta Derm Venereol. 2012;92:241-246. 
  8. Khandalavala BN, Do MV. Finasteride in hidradenitis suppurativa: a "male" therapy for a predominantly "female" disease. J Clin Aesthet Dermatol. 2016;9:44. 
  9. Nikolakis G, Karagiannidis I, Vaiopoulos AG, et al. Endocrinological mechanisms in the pathophysiology of hidradenitis suppurativa [in German]. Hautarzt. 2020;71:762-771. 
  10. Karadag AS, Ozlu E, Lavery MJ. Cutaneous manifestations of diabetes mellitus and the metabolic syndrome. Clin Dermatology. 2018;36:89-93. 
  11. Gardner CD, Kiazand A, Alhassan S, et al. Comparison of the Atkins, Zone, Ornish, and LEARN diets for change in weight and related risk factors among overweight premenopausal women: the A TO Z Weight Loss Study: a randomized trial. JAMA. 2007;297:969-977. 
  12. Anton SD, Hida A, Heekin K, et al. Effects of popular diets without specific calorie targets on weight loss outcomes: systematic review of findings from clinical trials. Nutrients. 2017;9:822. 
  13. Castellana M, Conte E, Cignarelli A, et al. Efficacy and safety of very low calorie ketogenic diet (VLCKD) in patients with overweight and obesity: a systematic review and meta-analysis. Rev Endocr Metab Disord. 2020;21:5-16. 
  14. Paoli A, Mancin L, Giacona MC, et al. Effects of a ketogenic diet in overweight women with polycystic ovary syndrome. J Transl Med. 2020;18:104. 
  15. Dashti HM, Mathew TC, Hussein T, et al. Long-term effects of a ketogenic diet in obese patients. Exp Clin Cardiol. 2004;9:200-205. 
  16. Lian N, Chen M. Metabolic syndrome and skin disease: potential connection and risk. Int J Dermatol Venereol. 2019;2:89-93. 
  17. Hu Y, Zhu Y, Lian N, et al. Metabolic syndrome and skin diseases. Front Endocrinol (Lausanne). 2019;10:788. 
  18. Castaldo G, Rastrelli L, Galdo G, et al. Aggressive weight-loss program with a ketogenic induction phase for the treatment of chronic plaque psoriasis: a proof-of-concept, single-arm, open-label clinical trial. Nutrition. 2020;74:110757. 
  19. Castaldo G, Pagano I, Grimaldi M, et al. Effect of very-low-calorie ketogenic diet on psoriasis patients: a nuclear magnetic resonance-based metabolomic study. J Proteome Res. 2021;20:1509-1521. 
  20. Milder J, Liang L-P, Patel M. Acute oxidative stress and systemic Nrf2 activation by the ketogenic diet. Neurobiol Dis. 2010;40:238-244. 
  21. Kubo E, Chhunchha B, Singh P, et al. Sulforaphane reactivates cellular antioxidant defense by inducing Nrf2/ARE/Prdx6 activity during aging and oxidative stress. Sci Rep. 2017;7:14130. 
  22. Pinto A, Bonucci A, Maggi E, et al. Anti-oxidant and anti-inflammatory activity of ketogenic diet: new perspectives for neuroprotection in Alzheimer's disease. Antioxidants (Basel). 2018;7:63. 
  23. Youm Y-H, Nguyen KY, Grant RW, et al. The ketone metabolite &#946;-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21:263-269. 
  24. Kelley N, Jeltema D, Duan Y, et al. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20:3328. 
  25. Fomin DA, McDaniel B, Crane J. The promising potential role of ketones in inflammatory dermatologic disease: a new frontier in treatment research. J Dermatol Treat. 2017;28:484-487. 
  26. Rahman M, Muhammad S, Khan MA, et al. The β-hydroxybutyrate receptor HCA 2 activates a neuroprotective subset of macrophages. Nat Commun. 2014;5:1-11. 
  27. Lu Y, Yang YY, Zhou MW, et al. Ketogenic diet attenuates oxidative stress and inflammation after spinal cord injury by activating Nrf2 and suppressing the NF-&#954;B signaling pathways. Neurosci Lett. 2018;683:13-18. 
  28.  Hamarsheh S, Zeiser R. NLRP3 inflammasome activation in cancer: a double-edged sword. Front Immunol. 2020;11:1444. 
  29. Bell S, Degitz K, Quirling M, et al. Involvement of NF-&#954;B signalling in skin physiology and disease. Cell Signal. 2003;15:1-7. 
  30. Goldminz AM, Au SC, Kim N, et al. NF-κB: an essential transcription factor in psoriasis. J Dermatol Sci. 2013;69:89-94. 
  31. Laplante M, Sabatini DM. mTOR signaling at a glance. J Cell Sci. 2009;122:3589. 
  32. McDaniel S, Rensing N, Yamada K, et al. The ketogenic diet inhibits the mammalian target of rapamycin (mTOR) pathway. Epilepsia. 2011;52:E7-E11. 
  33. Alter M, Satzger I, Schrem H, et al. Non-melanoma skin cancer is reduced after switch of immunosuppression to mTOR-inhibitors in organ transplant recipients. J Dtsch Dermatol Ges. 2014;12:480-488. 
  34. Feldmeyer L, Hofbauer GF, Böni T, et al. Mammalian target of rapamycin (mTOR) inhibitors slow skin carcinogenesis, but impair wound healing. Br J Dermatol. 2012;166:422-424. 
  35. Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016;41:211-218. 
  36. Li W. "Warburg effect" and mitochondrial metabolism in skin cancer.J Carcinogene Mutagene. 2012:S4. 
  37. Naveed S, Aslam M, Ahmad A. Starvation based differential chemotherapy: a novel approach for cancer treatment. Oman Med J. 2014;29:391-398. 
  38. Raffaghello L, Lee C, Safdie FM, et al. Starvation-dependent differential stress resistance protects normal but not cancer cells against high-dose chemotherapy. Proc Natl Acad Sci U S A. 2008;105:8215-8220. 
  39. Buono R, Longo VD. Starvation, stress resistance, and cancer. Trends Endocrinol Metab. 2018;29:271-280. 
  40. de Groot S, Pijl H, van der Hoeven JJM, et al. Effects of short-term fasting on cancer treatment. J Exp Clin Cancer Res. 2019;38:209. 
  41. Hosseini M, Kasraian Z, Rezvani HR. Energy metabolism in skin cancers: a therapeutic perspective. Biochim Biophys Acta Bioenerg. 2017;1858:712-722. 
  42. Feichtinger RG, Lang R, Geilberger R, et al. Melanoma tumors exhibit a variable but distinct metabolic signature. Exp Dermatol. 2018;27:204-207. 
  43. Alshaya MA, Turkmani MG, Alissa AM. Prurigo pigmentosa following ketogenic diet and bariatric surgery: a growing association. JAAD Case Rep. 2019;5:504-507. 
  44. Bellini M, Tonarelli S, Nagy AG, et al. Low FODMAP diet: evidence, doubts, and hopes. Nutrients. 2020;12:148. 
  45. Kwiatkowski L, Rice E, Langland J. Integrative treatment of chronic abdominal bloating and pain associated with overgrowth of small intestinal bacteria: a case report. Altern Ther Health Med. 2017;23:56-61. 
  46. Hubkova T. No more pain in the gut: lifestyle medicine approach to irritable bowel syndrome. Am J Lifestyle Med. 2017;11:223-226. 
  47. Schumann D, Klose P, Lauche R, et al. Low fermentable, oligo-, di-, mono-saccharides and polyol diet in the treatment of irritable bowel syndrome: a systematic review and meta-analysis. Nutrition. 2018;45:24-31. 
  48. Cox SR, Prince AC, Myers CE, et al. Fermentable carbohydrates [FODMAPs] exacerbate functional gastrointestinal symptoms in patients with inflammatory bowel disease: a randomised, double-blind, placebo-controlled, cross-over, re-challenge trial. J Crohns Colitis. 2017;11:1420-1429. 
  49. Damas OM, Garces L, Abreu MT. Diet as adjunctive treatment for inflammatory bowel disease: review and update of the latest literature. Curr Treat Options Gastroenterol. 2019;17:313-325. 
  50. Wang FY, Chi CC. Rosacea, germs, and bowels: a review on gastrointestinal comorbidities and gut-skin axis of rosacea. Adv Ther. 2021;38:1415-1424. 
  51. Daou H, Paradiso M, Hennessy K, et al. Rosacea and the microbiome: a systematic review. Dermatol Ther (Heidelb). 2021;11:1-12. 
  52. Weinstock LB, Steinhoff M. Rosacea and small intestinal bacterial overgrowth: prevalence and response to rifaximin. J Am Acad Dermatol. 2013;68:875-876. 
  53. Wu CY, Chang YT, Juan CK, et al. Risk of inflammatory bowel disease in patients with rosacea: results from a nationwide cohort study in Taiwan. J Am Acad Dermatol. 2017;76:911-917. 
  54. Egeberg A, Weinstock LB, Thyssen EP, et al. Rosacea and gastrointestinal disorders: a population-based cohort study. Br J Dermatol. 2017;176:100-106. 
  55. Drago F, De Col E, Agnoletti AF, et al. The role of small intestinal bacterial overgrowth in rosacea: a 3-year follow-up. J Am Acad Dermatol. 2016;75:E113-E115. 
  56. Parodi A, Paolino S, Greco A, et al. Small intestinal bacterial overgrowth in rosacea: clinical effectiveness of its eradication. Clin Gastroenterol Hepatol. 2008;6:759-764. 
  57. Ojetti V, De Simone C, Aguilar Sanchez J, et al. Malabsorption in psoriatic patients: cause or consequence? Scand J Gastroenterol. 2006;41:1267-1271. 
  58. Kim M, Choi KH, Hwang SW, et al. Inflammatory bowel disease is associated with an increased risk of inflammatory skin diseases: a population-based cross-sectional study. J Am Acad Dermatol. 2017;76:40-48. 
  59. Drago F, Ciccarese G, Indemini E, et al. Psoriasis and small intestine bacterial overgrowth. Int J Dermatol. 2018;57:112-113. 
  60. Acharya P, Mathur M. Association between psoriasis and celiac disease: a systematic review and meta-analysis. J Am Acad Dermatol. 2020;82:1376-1385. 
  61. Bhatia BK, Millsop JW, Debbaneh M, et al. Diet and psoriasis, part II: celiac disease and role of a gluten-free diet. J Am Acad Dermatol. 2014;71:350-358. 
  62. Michaëlsson G, Gerdén B, Hagforsen E, et al. Psoriasis patients with antibodies to gliadin can be improved by a gluten-free diet. Br J Dermatol. 2000;142:44-51. 
  63. Kolchak NA, Tetarnikova MK, Theodoropoulou MS, et al. Prevalence of antigliadin IgA antibodies in psoriasis vulgaris and response of seropositive patients to a gluten-free diet. J Multidiscip Healthc. 2018;11:13-19. 
  64. De Bastiani R, Gabrielli M, Lora L, et al. Association between coeliac disease and psoriasis: Italian primary care multicentre study. Dermatology. 2015;230:156-160. 
  65. Afifi L, Danesh MJ, Lee KM, et al. Dietary behaviors in psoriasis: patient-reported outcomes from a U.S. national survey. Dermatol Ther (Heidelb). 2017;7:227-242. 
  66. Loche F, Bazex J. Celiac disease associated with cutaneous sarcoidosic granuloma [in French]. Rev Med Interne. 1997;18:975-978. 
  67. Rodríguez-García C, González-Hernández S, Pérez-Robayna N, et al. Repigmentation of vitiligo lesions in a child with celiac disease after a gluten-free diet. Pediatr Dermatol. 2011;28:209-210. 
  68. Wijarnpreecha K, Panjawatanan P, Corral JE, et al. Celiac disease and risk of sarcoidosis: a systematic review and meta-analysis. J Evid Based Med. 2019;12:194-199. 
  69. Rodrigo L, Beteta-Gorriti V, Alvarez N, et al. Cutaneous and mucosal manifestations associated with celiac disease. Nutrients. 2018;10:800. 
  70. Song MS, Farber D, Bitton A, et al. Dermatomyositis associated with celiac disease: response to a gluten-free diet. Can J Gastroenterol. 2006;20:433-435. 
  71. Egan CA, Smith EP, Taylor TB, et al. Linear IgA bullous dermatosis responsive to a gluten-free diet. Am J Gastroenterol. 2001;96:1927-1929. 
  72. Comas-Basté O, Sánchez-Pérez S, Veciana-Nogués MT, et al. Histamine intolerance: the current state of the art. Biomolecules. 2020;10:1181. 
  73. Ring J. Plasma histamine concentrations in atopic eczema. Clin Allergy. 1983;13:545-552. 
  74. Maintz L, Benfadal S, Allam JP, et al. Evidence for a reduced histamine degradation capacity in a subgroup of patients with atopic eczema. J Allergy Clin Immunol. 2006;117:1106-1112. 
  75. Cornillier H, Giraudeau B, Samimi M, et al. Effect of diet in chronic spontaneous urticaria: a systematic review. Acta Derm Venereol. 2019;99:127-132. 
  76. Son JH, Chung BY, Kim HO, et al. A histamine-free diet is helpful for treatment of adult patients with chronic spontaneous urticaria. Ann Dermatol. 2018;30:164-172. 
  77. Wagner N, Dirk D, Peveling-Oberhag A, et al. A popular myth - low-histamine diet improves chronic spontaneous urticaria - fact or fiction? J Eur Acad Dermatol Venereol. 2017;31:650-655. 
  78. Esposito K, Marfella R, Ciotola M, et al. Effect of a Mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: a randomized trial. JAMA. 2004;292:1440-1446. 
  79. Steffen LM, Van Horn L, Daviglus ML, et al. A modified Mediterranean diet score is associated with a lower risk of incident metabolic syndrome over 25 years among young adults: the CARDIA (coronary artery risk development in young adults) study. Br J Nutr. 2014;112:1654-1661. 
  80. Bower A, Marquez S, de Mejia EG. The health benefits of selected culinary herbs and spices found in the traditional Mediterranean diet. Crit Rev Food Sci Nutr. 2016;56:2728-2746. 
  81. Bosch R, Philips N, Suárez-Pérez JA, et al. Mechanisms of photoaging and cutaneous photocarcinogenesis, and photoprotective strategies with phytochemicals. Antioxidants (Basel). 2015;4:248-268. 
  82. Katsimbri P, Korakas E, Kountouri A, et al. The effect of antioxidant and anti-inflammatory capacity of diet on psoriasis and psoriatic arthritis phenotype: nutrition as therapeutic tool? Antioxidants. 2021;10:157. 
  83. Molina-Leyva A, Cuenca-Barrales C, Vega-Castillo JJ, et al. Adherence to Mediterranean diet in Spanish patients with psoriasis: cardiovascular benefits? Dermatol Ther. 2019;32:E12810. 
  84. Barrea L, Balato N, Di Somma C, et al. Nutrition and psoriasis: is there any association between the severity of the disease and adherence to the Mediterranean diet? J Transl Med. 2015;13:1-10. 
  85. Phan C, Touvier M, Kesse-Guyot E, et al. Association between Mediterranean anti-inflammatory dietary profile and severity of psoriasis: results from the NutriNet-Santé cohort. JAMA Dermatol. 2018;154:1017-1024. 
  86. Korovesi A, Dalamaga M, Kotopouli M, et al. Adherence to the Mediterranean diet is independently associated with psoriasis risk, severity, and quality of life: a cross-sectional observational study. Int J Dermatol. 2019;58:E164-E165. 
  87. Ford AR, Siegel M, Bagel J, et al. Dietary recommendations for adults with psoriasis or psoriatic arthritis from the medical board of the National Psoriasis Foundation: a systematic review. JAMA Dermatol. 2018;154:934-950. 
  88. Skroza N, Tolino E, Semyonov L, et al. Mediterranean diet and familial dysmetabolism as factors influencing the development of acne. Scand J Public Health. 2012;40:466-474. 
  89. Barrea L, Fabbrocini G, Annunziata G, et al. Role of nutrition and adherence to the Mediterranean diet in the multidisciplinary approach of hidradenitis suppurativa: evaluation of nutritional status and its association with severity of disease. Nutrients. 2018;11:57. 
  90. Nichols JA, Katiyar SK. Skin photoprotection by natural polyphenols: anti-inflammatory, antioxidant and DNA repair mechanisms. Arch Dermatol Res. 2010;302:71-83. 
  91. Huang T-H, Wang P-W, Yang S-C, et al. Cosmetic and therapeutic applications of fish oil's fatty acids on the skin. Mar Drugs. 2018;16:256. 
  92. Rizwan M, Rodriguez-Blanco I, Harbottle A, et al. Tomato paste rich in lycopene protects against cutaneous photodamage in humans in vivo: a randomized controlled trial. Br J Dermatol. 2011;164:154-162. 
  93. Leone A, Martínez-González M, Martin-Gorgojo A, et al. Mediterranean diet, dietary approaches to stop hypertension, and pro-vegetarian dietary pattern in relation to the risk of basal cell carcinoma: a nested case-control study within the Seguimiento Universidad de Navarra (SUN) cohort. Am J Clin Nutr. 2020;112:364-372. 
  94. Solway J, McBride M, Haq F, et al. Diet and dermatology: the role of a whole-food, plant-based diet in preventing and reversing skin aging--a review. J Clin Aesthet Dermatol. 2020;13:38-43. 
  95. Greger M. A whole food plant-based diet is effective for weight loss: the evidence. Am J Lifestyle Med. 2020;14:500-510. 
  96. Wright N, Wilson L, Smith M, et al. The BROAD study: a randomised controlled trial using a whole food plant-based diet in the community for obesity, ischaemic heart disease or diabetes. Nutr Diabetes. 2017;7:E256. 
  97. Ornish D, Lin J, Chan JM, et al. Effect of comprehensive lifestyle changes on telomerase activity and telomere length in men with biopsy-proven low-risk prostate cancer: 5-year follow-up of a descriptive pilot study. Lancet Oncol. 2013;14:1112-1120. 
  98. Ornish D, Lin J, Daubenmier J, et al. Increased telomerase activity and comprehensive lifestyle changes: a pilot study. Lancet Oncol. 2008;9:1048-1057. 
  99. Zouboulis CC, Makrantonaki E. Clinical aspects and molecular diagnostics of skin aging. Clin Dermatol. 2011;29:3-14. 
  100. Gupta L, Khandelwal D, Lal PR, et al. Palaeolithic diet in diabesity and endocrinopathies--a vegan's perspective. Eur Endocrinol. 2019;15:77-82. 
  101. Chassaing B, Van de Wiele T, De Bodt J, et al. Dietary emulsifiers directly alter human microbiota composition and gene expression ex vivo potentiating intestinal inflammation. Gut. 2017;66:1414-1427. 
  102. Thorburn Alison N, Macia L, Mackay Charles R. Diet, metabolites, and "Western lifestyle" inflammatory diseases. Immunity. 2014;40:833-842. 
  103. Katta R, Schlichte M. Diet and dermatitis: food triggers. J Clin Aesthet Dermatol. 2014;7:30-36. 
  104. Dhar S, Srinivas SM. Food allergy in atopic dermatitis. Indian J Dermatol. 2016;61:645-648. 
  105. Birmingham N, Thanesvorakul S, Gangur V. Relative immunogenicity of commonly allergenic foods versus rarely allergenic and nonallergenic foods in mice. J Food Prot. 2002;65:1988-1991. 
  106. Yu W, Freeland DMH, Nadeau KC. Food allergy: immune mechanisms, diagnosis and immunotherapy. Nat Rev Immunol. 2016;16:751-765. 
  107. Kowalski LM, Bujko J. Evaluation of biological and clinical potential of paleolithic diet [in Polish]. Rocz Panstw Zakl Hig. 2012;63:9-15. 
  108. Lee JE, Titcomb TJ, Bisht B, et al. A modified MCT-based ketogenic diet increases plasma β-hydroxybutyrate but has less effect on fatigue and quality of life in people with multiple sclerosis compared to a modified paleolithic diet: a waitlist-controlled, randomized pilot study. J Am Coll Nutr. 2021;40:13-25. 
  109. Abbott RD, Sadowski A, Alt AG. Efficacy of the autoimmune protocol diet as part of a multi-disciplinary, supported lifestyle intervention for Hashimoto's thyroiditis. Cureus. 2019;11:E4556. 
  110. Lindeberg S, Jönsson T, Granfeldt Y, et al. A palaeolithic diet improves glucose tolerance more than a Mediterranean-like diet in individuals with ischaemic heart disease. Diabetologia. 2007;50:1795-1807. 
  111. Jönsson T, Granfeldt Y, Ahrén B, et al. Beneficial effects of a paleolithic diet on cardiovascular risk factors in type 2 diabetes: a randomized cross-over pilot study. Cardiovasc Diabetol. 2009;8:35. 
  112. Boers I, Muskiet FAJ, Berkelaar E, et al. Favourable effects of consuming a palaeolithic-type diet on characteristics of the metabolic syndrome: a randomized controlled pilot-study. Lipids Health Dis. 2014;13:160. 
  113. Ghaedi E, Mohammadi M, Mohammadi H, et al. Effects of a paleolithic diet on cardiovascular disease risk factors: a systematic review and meta-analysis of randomized controlled trials. Adv Nutr. 2019;10:634-646. 
  114. Mellberg C, Sandberg S, Ryberg M, et al. Long-term effects of a palaeolithic-type diet in obese postmenopausal women: a 2-year randomized trial. Eur J Clin Nutr. 2014;68:350-357. 
  115. Pastore RL, Brooks JT, Carbone JW. Paleolithic nutrition improves plasma lipid concentrations of hypercholesterolemic adults to a greater extent than traditional heart-healthy dietary recommendations. Nutr Res. 2015;35:474-479. 
  116. Otten J, Stomby A, Waling M, et al. Benefits of a paleolithic diet with and without supervised exercise on fat mass, insulin sensitivity, and glycemic control: a randomized controlled trial in individuals with type 2 diabetes. Diabetes Metab Res Rev. 2017;33:E2828. 
  117. Stefanadi EC, Dimitrakakis G, Antoniou C-K, et al. Metabolic syndrome and the skin: a more than superficial association. reviewing the association between skin diseases and metabolic syndrome and a clinical decision algorithm for high risk patients. Diabetol Metab Syndr. 2018;10:9. 
  118. Mann N. Meat in the human diet: an anthropological perspective. Nutr Dietetics. 2007;64(suppl 4):S102-S107. 
  119. Bramble DM, Lieberman DE. Endurance running and the evolution of Homo. Nature. 2004;432:345-352. 
  120. Kuhn JE. Throwing, the shoulder, and human evolution. Am J Orthop (Belle Mead NJ). 2016;45:110-114. 
  121. Kobayashi H, Kohshima S. Unique morphology of the human eye and its adaptive meaning: comparative studies on external morphology of the primate eye. J Hum Evol. 2001;40:419-435. 
  122. Cordain L, Eaton SB, Miller JB, et al. The paradoxical nature of hunter-gatherer diets: meat-based, yet non-atherogenic. Eur J Clin Nutr. 2002;56(suppl 1):S42-S52. 
  123. McClellan WS, Du Bois EF. Clinical calorimetry: XLV. prolonged meat diets with a study of kidney function and ketosis. J Biol Chem. 1930;87:651-668. 
  124. O'Hearn A. Can a carnivore diet provide all essential nutrients? Curr Opin Endocrinol Diabetes Obes. 2020;27:312-316. 
  125. O'Hearn LA. A survey of improvements experienced on a carnivore diet compared to only carbohydrate restriction. Open Science Forum website. Published February 12, 2019. Accessed May 17, 2021. doi:10.17605/OSF.IO/5FU4D 
  126. Williams P. Nutritional composition of red meat. Nutrition & Dietetics. 2007;64(suppl 4):S113-S119. 
  127. Biel W, Czerniawska-Piątkowska E, Kowalczyk A. Offal chemical composition from veal, beef, and lamb maintained in organic production systems. Animals (Basel). 2019;9:489. 
  128. Elmadfa I, Meyer AL. The role of the status of selected micronutrients in shaping the immune function. Endocr Metab Immune Disord Drug Targets. 2019;19:1100-1115. 
  129. Babizhayev M. Treatment of skin aging and photoaging with innovative oral dosage forms of nonhydrolized carnosine and carcinine. Int J Clin Derm Res. 2017;5:116-143. 
  130. Danby FW. Nutrition and aging skin: sugar and glycation. Clin Dermatol. 2010;28:409-411. 
  131. Siefken W, Carstensen S, Springmann G, et al. Role of taurine accumulation in keratinocyte hydration. J Invest Dermatol. 2003;121:354-361. 
  132. Vollmer DL, West VA, Lephart ED. Enhancing skin health: by oral administration of natural compounds and minerals with implications to the dermal microbiome. Int J Mol Sci. 2018;19:3059. 
  133. Fischer F, Achterberg V, März A, et al. Folic acid and creatineimprove the firmness of human skin in vivo. J Cosmet Dermatol. 2011;10:15-23. 
  134. Blatt T, Lenz H, Weber T. Topical application of creatine is multibeneficial for human skin. J Am Acad Dermatol. 2005;52:P32.
References
  1. Jaros J, Katta R, Shi VY. Dermatonutrigenomics: past, present, and future. Dermatology. 2019;235:164-166. 
  2. Paoli A, Grimaldi K, Toniolo L, et al. Nutrition and acne: therapeutic potential of ketogenic diets. Skin Pharmacol Physiol. 2012;25:111-117. 
  3. Melnik BC, Schmitz G. Role of insulin, insulin-like growth factor-1, hyperglycaemic food and milk consumption in the pathogenesis of acne vulgaris. Exp Dermatol. 2009;18:833-841. 
  4. Smith RN, Mann NJ, Braue A, et al. The effect of a high-protein, low glycemic-load diet versus a conventional, high glycemic-load diet on biochemical parameters associated with acne vulgaris: a randomized, investigator-masked, controlled trial. J Am Acad Dermatol. 2007;57:247-256. 
  5. Smith R, Mann N, Mäkeläinen H, et al. A pilot study to determine the short-term effects of a low glycemic load diet on hormonal markers of acne: a nonrandomized, parallel, controlled feeding trial. Mol Nutr Food Res. 2008;52:718-726. 
  6. Smith RN, Braue A, Varigos GA, et al. The effect of a low glycemic load diet on acne vulgaris and the fatty acid composition of skin surface triglycerides. J Dermatol Sci. 2008;50:41-52. 
  7. Kwon HH, Yoon JY, Hong JS, et al. Clinical and histological effect of a low glycaemic load diet in treatment of acne vulgaris in Korean patients: a randomized, controlled trial. Acta Derm Venereol. 2012;92:241-246. 
  8. Khandalavala BN, Do MV. Finasteride in hidradenitis suppurativa: a "male" therapy for a predominantly "female" disease. J Clin Aesthet Dermatol. 2016;9:44. 
  9. Nikolakis G, Karagiannidis I, Vaiopoulos AG, et al. Endocrinological mechanisms in the pathophysiology of hidradenitis suppurativa [in German]. Hautarzt. 2020;71:762-771. 
  10. Karadag AS, Ozlu E, Lavery MJ. Cutaneous manifestations of diabetes mellitus and the metabolic syndrome. Clin Dermatology. 2018;36:89-93. 
  11. Gardner CD, Kiazand A, Alhassan S, et al. Comparison of the Atkins, Zone, Ornish, and LEARN diets for change in weight and related risk factors among overweight premenopausal women: the A TO Z Weight Loss Study: a randomized trial. JAMA. 2007;297:969-977. 
  12. Anton SD, Hida A, Heekin K, et al. Effects of popular diets without specific calorie targets on weight loss outcomes: systematic review of findings from clinical trials. Nutrients. 2017;9:822. 
  13. Castellana M, Conte E, Cignarelli A, et al. Efficacy and safety of very low calorie ketogenic diet (VLCKD) in patients with overweight and obesity: a systematic review and meta-analysis. Rev Endocr Metab Disord. 2020;21:5-16. 
  14. Paoli A, Mancin L, Giacona MC, et al. Effects of a ketogenic diet in overweight women with polycystic ovary syndrome. J Transl Med. 2020;18:104. 
  15. Dashti HM, Mathew TC, Hussein T, et al. Long-term effects of a ketogenic diet in obese patients. Exp Clin Cardiol. 2004;9:200-205. 
  16. Lian N, Chen M. Metabolic syndrome and skin disease: potential connection and risk. Int J Dermatol Venereol. 2019;2:89-93. 
  17. Hu Y, Zhu Y, Lian N, et al. Metabolic syndrome and skin diseases. Front Endocrinol (Lausanne). 2019;10:788. 
  18. Castaldo G, Rastrelli L, Galdo G, et al. Aggressive weight-loss program with a ketogenic induction phase for the treatment of chronic plaque psoriasis: a proof-of-concept, single-arm, open-label clinical trial. Nutrition. 2020;74:110757. 
  19. Castaldo G, Pagano I, Grimaldi M, et al. Effect of very-low-calorie ketogenic diet on psoriasis patients: a nuclear magnetic resonance-based metabolomic study. J Proteome Res. 2021;20:1509-1521. 
  20. Milder J, Liang L-P, Patel M. Acute oxidative stress and systemic Nrf2 activation by the ketogenic diet. Neurobiol Dis. 2010;40:238-244. 
  21. Kubo E, Chhunchha B, Singh P, et al. Sulforaphane reactivates cellular antioxidant defense by inducing Nrf2/ARE/Prdx6 activity during aging and oxidative stress. Sci Rep. 2017;7:14130. 
  22. Pinto A, Bonucci A, Maggi E, et al. Anti-oxidant and anti-inflammatory activity of ketogenic diet: new perspectives for neuroprotection in Alzheimer's disease. Antioxidants (Basel). 2018;7:63. 
  23. Youm Y-H, Nguyen KY, Grant RW, et al. The ketone metabolite &#946;-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21:263-269. 
  24. Kelley N, Jeltema D, Duan Y, et al. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20:3328. 
  25. Fomin DA, McDaniel B, Crane J. The promising potential role of ketones in inflammatory dermatologic disease: a new frontier in treatment research. J Dermatol Treat. 2017;28:484-487. 
  26. Rahman M, Muhammad S, Khan MA, et al. The β-hydroxybutyrate receptor HCA 2 activates a neuroprotective subset of macrophages. Nat Commun. 2014;5:1-11. 
  27. Lu Y, Yang YY, Zhou MW, et al. Ketogenic diet attenuates oxidative stress and inflammation after spinal cord injury by activating Nrf2 and suppressing the NF-&#954;B signaling pathways. Neurosci Lett. 2018;683:13-18. 
  28.  Hamarsheh S, Zeiser R. NLRP3 inflammasome activation in cancer: a double-edged sword. Front Immunol. 2020;11:1444. 
  29. Bell S, Degitz K, Quirling M, et al. Involvement of NF-&#954;B signalling in skin physiology and disease. Cell Signal. 2003;15:1-7. 
  30. Goldminz AM, Au SC, Kim N, et al. NF-κB: an essential transcription factor in psoriasis. J Dermatol Sci. 2013;69:89-94. 
  31. Laplante M, Sabatini DM. mTOR signaling at a glance. J Cell Sci. 2009;122:3589. 
  32. McDaniel S, Rensing N, Yamada K, et al. The ketogenic diet inhibits the mammalian target of rapamycin (mTOR) pathway. Epilepsia. 2011;52:E7-E11. 
  33. Alter M, Satzger I, Schrem H, et al. Non-melanoma skin cancer is reduced after switch of immunosuppression to mTOR-inhibitors in organ transplant recipients. J Dtsch Dermatol Ges. 2014;12:480-488. 
  34. Feldmeyer L, Hofbauer GF, Böni T, et al. Mammalian target of rapamycin (mTOR) inhibitors slow skin carcinogenesis, but impair wound healing. Br J Dermatol. 2012;166:422-424. 
  35. Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016;41:211-218. 
  36. Li W. "Warburg effect" and mitochondrial metabolism in skin cancer.J Carcinogene Mutagene. 2012:S4. 
  37. Naveed S, Aslam M, Ahmad A. Starvation based differential chemotherapy: a novel approach for cancer treatment. Oman Med J. 2014;29:391-398. 
  38. Raffaghello L, Lee C, Safdie FM, et al. Starvation-dependent differential stress resistance protects normal but not cancer cells against high-dose chemotherapy. Proc Natl Acad Sci U S A. 2008;105:8215-8220. 
  39. Buono R, Longo VD. Starvation, stress resistance, and cancer. Trends Endocrinol Metab. 2018;29:271-280. 
  40. de Groot S, Pijl H, van der Hoeven JJM, et al. Effects of short-term fasting on cancer treatment. J Exp Clin Cancer Res. 2019;38:209. 
  41. Hosseini M, Kasraian Z, Rezvani HR. Energy metabolism in skin cancers: a therapeutic perspective. Biochim Biophys Acta Bioenerg. 2017;1858:712-722. 
  42. Feichtinger RG, Lang R, Geilberger R, et al. Melanoma tumors exhibit a variable but distinct metabolic signature. Exp Dermatol. 2018;27:204-207. 
  43. Alshaya MA, Turkmani MG, Alissa AM. Prurigo pigmentosa following ketogenic diet and bariatric surgery: a growing association. JAAD Case Rep. 2019;5:504-507. 
  44. Bellini M, Tonarelli S, Nagy AG, et al. Low FODMAP diet: evidence, doubts, and hopes. Nutrients. 2020;12:148. 
  45. Kwiatkowski L, Rice E, Langland J. Integrative treatment of chronic abdominal bloating and pain associated with overgrowth of small intestinal bacteria: a case report. Altern Ther Health Med. 2017;23:56-61. 
  46. Hubkova T. No more pain in the gut: lifestyle medicine approach to irritable bowel syndrome. Am J Lifestyle Med. 2017;11:223-226. 
  47. Schumann D, Klose P, Lauche R, et al. Low fermentable, oligo-, di-, mono-saccharides and polyol diet in the treatment of irritable bowel syndrome: a systematic review and meta-analysis. Nutrition. 2018;45:24-31. 
  48. Cox SR, Prince AC, Myers CE, et al. Fermentable carbohydrates [FODMAPs] exacerbate functional gastrointestinal symptoms in patients with inflammatory bowel disease: a randomised, double-blind, placebo-controlled, cross-over, re-challenge trial. J Crohns Colitis. 2017;11:1420-1429. 
  49. Damas OM, Garces L, Abreu MT. Diet as adjunctive treatment for inflammatory bowel disease: review and update of the latest literature. Curr Treat Options Gastroenterol. 2019;17:313-325. 
  50. Wang FY, Chi CC. Rosacea, germs, and bowels: a review on gastrointestinal comorbidities and gut-skin axis of rosacea. Adv Ther. 2021;38:1415-1424. 
  51. Daou H, Paradiso M, Hennessy K, et al. Rosacea and the microbiome: a systematic review. Dermatol Ther (Heidelb). 2021;11:1-12. 
  52. Weinstock LB, Steinhoff M. Rosacea and small intestinal bacterial overgrowth: prevalence and response to rifaximin. J Am Acad Dermatol. 2013;68:875-876. 
  53. Wu CY, Chang YT, Juan CK, et al. Risk of inflammatory bowel disease in patients with rosacea: results from a nationwide cohort study in Taiwan. J Am Acad Dermatol. 2017;76:911-917. 
  54. Egeberg A, Weinstock LB, Thyssen EP, et al. Rosacea and gastrointestinal disorders: a population-based cohort study. Br J Dermatol. 2017;176:100-106. 
  55. Drago F, De Col E, Agnoletti AF, et al. The role of small intestinal bacterial overgrowth in rosacea: a 3-year follow-up. J Am Acad Dermatol. 2016;75:E113-E115. 
  56. Parodi A, Paolino S, Greco A, et al. Small intestinal bacterial overgrowth in rosacea: clinical effectiveness of its eradication. Clin Gastroenterol Hepatol. 2008;6:759-764. 
  57. Ojetti V, De Simone C, Aguilar Sanchez J, et al. Malabsorption in psoriatic patients: cause or consequence? Scand J Gastroenterol. 2006;41:1267-1271. 
  58. Kim M, Choi KH, Hwang SW, et al. Inflammatory bowel disease is associated with an increased risk of inflammatory skin diseases: a population-based cross-sectional study. J Am Acad Dermatol. 2017;76:40-48. 
  59. Drago F, Ciccarese G, Indemini E, et al. Psoriasis and small intestine bacterial overgrowth. Int J Dermatol. 2018;57:112-113. 
  60. Acharya P, Mathur M. Association between psoriasis and celiac disease: a systematic review and meta-analysis. J Am Acad Dermatol. 2020;82:1376-1385. 
  61. Bhatia BK, Millsop JW, Debbaneh M, et al. Diet and psoriasis, part II: celiac disease and role of a gluten-free diet. J Am Acad Dermatol. 2014;71:350-358. 
  62. Michaëlsson G, Gerdén B, Hagforsen E, et al. Psoriasis patients with antibodies to gliadin can be improved by a gluten-free diet. Br J Dermatol. 2000;142:44-51. 
  63. Kolchak NA, Tetarnikova MK, Theodoropoulou MS, et al. Prevalence of antigliadin IgA antibodies in psoriasis vulgaris and response of seropositive patients to a gluten-free diet. J Multidiscip Healthc. 2018;11:13-19. 
  64. De Bastiani R, Gabrielli M, Lora L, et al. Association between coeliac disease and psoriasis: Italian primary care multicentre study. Dermatology. 2015;230:156-160. 
  65. Afifi L, Danesh MJ, Lee KM, et al. Dietary behaviors in psoriasis: patient-reported outcomes from a U.S. national survey. Dermatol Ther (Heidelb). 2017;7:227-242. 
  66. Loche F, Bazex J. Celiac disease associated with cutaneous sarcoidosic granuloma [in French]. Rev Med Interne. 1997;18:975-978. 
  67. Rodríguez-García C, González-Hernández S, Pérez-Robayna N, et al. Repigmentation of vitiligo lesions in a child with celiac disease after a gluten-free diet. Pediatr Dermatol. 2011;28:209-210. 
  68. Wijarnpreecha K, Panjawatanan P, Corral JE, et al. Celiac disease and risk of sarcoidosis: a systematic review and meta-analysis. J Evid Based Med. 2019;12:194-199. 
  69. Rodrigo L, Beteta-Gorriti V, Alvarez N, et al. Cutaneous and mucosal manifestations associated with celiac disease. Nutrients. 2018;10:800. 
  70. Song MS, Farber D, Bitton A, et al. Dermatomyositis associated with celiac disease: response to a gluten-free diet. Can J Gastroenterol. 2006;20:433-435. 
  71. Egan CA, Smith EP, Taylor TB, et al. Linear IgA bullous dermatosis responsive to a gluten-free diet. Am J Gastroenterol. 2001;96:1927-1929. 
  72. Comas-Basté O, Sánchez-Pérez S, Veciana-Nogués MT, et al. Histamine intolerance: the current state of the art. Biomolecules. 2020;10:1181. 
  73. Ring J. Plasma histamine concentrations in atopic eczema. Clin Allergy. 1983;13:545-552. 
  74. Maintz L, Benfadal S, Allam JP, et al. Evidence for a reduced histamine degradation capacity in a subgroup of patients with atopic eczema. J Allergy Clin Immunol. 2006;117:1106-1112. 
  75. Cornillier H, Giraudeau B, Samimi M, et al. Effect of diet in chronic spontaneous urticaria: a systematic review. Acta Derm Venereol. 2019;99:127-132. 
  76. Son JH, Chung BY, Kim HO, et al. A histamine-free diet is helpful for treatment of adult patients with chronic spontaneous urticaria. Ann Dermatol. 2018;30:164-172. 
  77. Wagner N, Dirk D, Peveling-Oberhag A, et al. A popular myth - low-histamine diet improves chronic spontaneous urticaria - fact or fiction? J Eur Acad Dermatol Venereol. 2017;31:650-655. 
  78. Esposito K, Marfella R, Ciotola M, et al. Effect of a Mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: a randomized trial. JAMA. 2004;292:1440-1446. 
  79. Steffen LM, Van Horn L, Daviglus ML, et al. A modified Mediterranean diet score is associated with a lower risk of incident metabolic syndrome over 25 years among young adults: the CARDIA (coronary artery risk development in young adults) study. Br J Nutr. 2014;112:1654-1661. 
  80. Bower A, Marquez S, de Mejia EG. The health benefits of selected culinary herbs and spices found in the traditional Mediterranean diet. Crit Rev Food Sci Nutr. 2016;56:2728-2746. 
  81. Bosch R, Philips N, Suárez-Pérez JA, et al. Mechanisms of photoaging and cutaneous photocarcinogenesis, and photoprotective strategies with phytochemicals. Antioxidants (Basel). 2015;4:248-268. 
  82. Katsimbri P, Korakas E, Kountouri A, et al. The effect of antioxidant and anti-inflammatory capacity of diet on psoriasis and psoriatic arthritis phenotype: nutrition as therapeutic tool? Antioxidants. 2021;10:157. 
  83. Molina-Leyva A, Cuenca-Barrales C, Vega-Castillo JJ, et al. Adherence to Mediterranean diet in Spanish patients with psoriasis: cardiovascular benefits? Dermatol Ther. 2019;32:E12810. 
  84. Barrea L, Balato N, Di Somma C, et al. Nutrition and psoriasis: is there any association between the severity of the disease and adherence to the Mediterranean diet? J Transl Med. 2015;13:1-10. 
  85. Phan C, Touvier M, Kesse-Guyot E, et al. Association between Mediterranean anti-inflammatory dietary profile and severity of psoriasis: results from the NutriNet-Santé cohort. JAMA Dermatol. 2018;154:1017-1024. 
  86. Korovesi A, Dalamaga M, Kotopouli M, et al. Adherence to the Mediterranean diet is independently associated with psoriasis risk, severity, and quality of life: a cross-sectional observational study. Int J Dermatol. 2019;58:E164-E165. 
  87. Ford AR, Siegel M, Bagel J, et al. Dietary recommendations for adults with psoriasis or psoriatic arthritis from the medical board of the National Psoriasis Foundation: a systematic review. JAMA Dermatol. 2018;154:934-950. 
  88. Skroza N, Tolino E, Semyonov L, et al. Mediterranean diet and familial dysmetabolism as factors influencing the development of acne. Scand J Public Health. 2012;40:466-474. 
  89. Barrea L, Fabbrocini G, Annunziata G, et al. Role of nutrition and adherence to the Mediterranean diet in the multidisciplinary approach of hidradenitis suppurativa: evaluation of nutritional status and its association with severity of disease. Nutrients. 2018;11:57. 
  90. Nichols JA, Katiyar SK. Skin photoprotection by natural polyphenols: anti-inflammatory, antioxidant and DNA repair mechanisms. Arch Dermatol Res. 2010;302:71-83. 
  91. Huang T-H, Wang P-W, Yang S-C, et al. Cosmetic and therapeutic applications of fish oil's fatty acids on the skin. Mar Drugs. 2018;16:256. 
  92. Rizwan M, Rodriguez-Blanco I, Harbottle A, et al. Tomato paste rich in lycopene protects against cutaneous photodamage in humans in vivo: a randomized controlled trial. Br J Dermatol. 2011;164:154-162. 
  93. Leone A, Martínez-González M, Martin-Gorgojo A, et al. Mediterranean diet, dietary approaches to stop hypertension, and pro-vegetarian dietary pattern in relation to the risk of basal cell carcinoma: a nested case-control study within the Seguimiento Universidad de Navarra (SUN) cohort. Am J Clin Nutr. 2020;112:364-372. 
  94. Solway J, McBride M, Haq F, et al. Diet and dermatology: the role of a whole-food, plant-based diet in preventing and reversing skin aging--a review. J Clin Aesthet Dermatol. 2020;13:38-43. 
  95. Greger M. A whole food plant-based diet is effective for weight loss: the evidence. Am J Lifestyle Med. 2020;14:500-510. 
  96. Wright N, Wilson L, Smith M, et al. The BROAD study: a randomised controlled trial using a whole food plant-based diet in the community for obesity, ischaemic heart disease or diabetes. Nutr Diabetes. 2017;7:E256. 
  97. Ornish D, Lin J, Chan JM, et al. Effect of comprehensive lifestyle changes on telomerase activity and telomere length in men with biopsy-proven low-risk prostate cancer: 5-year follow-up of a descriptive pilot study. Lancet Oncol. 2013;14:1112-1120. 
  98. Ornish D, Lin J, Daubenmier J, et al. Increased telomerase activity and comprehensive lifestyle changes: a pilot study. Lancet Oncol. 2008;9:1048-1057. 
  99. Zouboulis CC, Makrantonaki E. Clinical aspects and molecular diagnostics of skin aging. Clin Dermatol. 2011;29:3-14. 
  100. Gupta L, Khandelwal D, Lal PR, et al. Palaeolithic diet in diabesity and endocrinopathies--a vegan's perspective. Eur Endocrinol. 2019;15:77-82. 
  101. Chassaing B, Van de Wiele T, De Bodt J, et al. Dietary emulsifiers directly alter human microbiota composition and gene expression ex vivo potentiating intestinal inflammation. Gut. 2017;66:1414-1427. 
  102. Thorburn Alison N, Macia L, Mackay Charles R. Diet, metabolites, and "Western lifestyle" inflammatory diseases. Immunity. 2014;40:833-842. 
  103. Katta R, Schlichte M. Diet and dermatitis: food triggers. J Clin Aesthet Dermatol. 2014;7:30-36. 
  104. Dhar S, Srinivas SM. Food allergy in atopic dermatitis. Indian J Dermatol. 2016;61:645-648. 
  105. Birmingham N, Thanesvorakul S, Gangur V. Relative immunogenicity of commonly allergenic foods versus rarely allergenic and nonallergenic foods in mice. J Food Prot. 2002;65:1988-1991. 
  106. Yu W, Freeland DMH, Nadeau KC. Food allergy: immune mechanisms, diagnosis and immunotherapy. Nat Rev Immunol. 2016;16:751-765. 
  107. Kowalski LM, Bujko J. Evaluation of biological and clinical potential of paleolithic diet [in Polish]. Rocz Panstw Zakl Hig. 2012;63:9-15. 
  108. Lee JE, Titcomb TJ, Bisht B, et al. A modified MCT-based ketogenic diet increases plasma β-hydroxybutyrate but has less effect on fatigue and quality of life in people with multiple sclerosis compared to a modified paleolithic diet: a waitlist-controlled, randomized pilot study. J Am Coll Nutr. 2021;40:13-25. 
  109. Abbott RD, Sadowski A, Alt AG. Efficacy of the autoimmune protocol diet as part of a multi-disciplinary, supported lifestyle intervention for Hashimoto's thyroiditis. Cureus. 2019;11:E4556. 
  110. Lindeberg S, Jönsson T, Granfeldt Y, et al. A palaeolithic diet improves glucose tolerance more than a Mediterranean-like diet in individuals with ischaemic heart disease. Diabetologia. 2007;50:1795-1807. 
  111. Jönsson T, Granfeldt Y, Ahrén B, et al. Beneficial effects of a paleolithic diet on cardiovascular risk factors in type 2 diabetes: a randomized cross-over pilot study. Cardiovasc Diabetol. 2009;8:35. 
  112. Boers I, Muskiet FAJ, Berkelaar E, et al. Favourable effects of consuming a palaeolithic-type diet on characteristics of the metabolic syndrome: a randomized controlled pilot-study. Lipids Health Dis. 2014;13:160. 
  113. Ghaedi E, Mohammadi M, Mohammadi H, et al. Effects of a paleolithic diet on cardiovascular disease risk factors: a systematic review and meta-analysis of randomized controlled trials. Adv Nutr. 2019;10:634-646. 
  114. Mellberg C, Sandberg S, Ryberg M, et al. Long-term effects of a palaeolithic-type diet in obese postmenopausal women: a 2-year randomized trial. Eur J Clin Nutr. 2014;68:350-357. 
  115. Pastore RL, Brooks JT, Carbone JW. Paleolithic nutrition improves plasma lipid concentrations of hypercholesterolemic adults to a greater extent than traditional heart-healthy dietary recommendations. Nutr Res. 2015;35:474-479. 
  116. Otten J, Stomby A, Waling M, et al. Benefits of a paleolithic diet with and without supervised exercise on fat mass, insulin sensitivity, and glycemic control: a randomized controlled trial in individuals with type 2 diabetes. Diabetes Metab Res Rev. 2017;33:E2828. 
  117. Stefanadi EC, Dimitrakakis G, Antoniou C-K, et al. Metabolic syndrome and the skin: a more than superficial association. reviewing the association between skin diseases and metabolic syndrome and a clinical decision algorithm for high risk patients. Diabetol Metab Syndr. 2018;10:9. 
  118. Mann N. Meat in the human diet: an anthropological perspective. Nutr Dietetics. 2007;64(suppl 4):S102-S107. 
  119. Bramble DM, Lieberman DE. Endurance running and the evolution of Homo. Nature. 2004;432:345-352. 
  120. Kuhn JE. Throwing, the shoulder, and human evolution. Am J Orthop (Belle Mead NJ). 2016;45:110-114. 
  121. Kobayashi H, Kohshima S. Unique morphology of the human eye and its adaptive meaning: comparative studies on external morphology of the primate eye. J Hum Evol. 2001;40:419-435. 
  122. Cordain L, Eaton SB, Miller JB, et al. The paradoxical nature of hunter-gatherer diets: meat-based, yet non-atherogenic. Eur J Clin Nutr. 2002;56(suppl 1):S42-S52. 
  123. McClellan WS, Du Bois EF. Clinical calorimetry: XLV. prolonged meat diets with a study of kidney function and ketosis. J Biol Chem. 1930;87:651-668. 
  124. O'Hearn A. Can a carnivore diet provide all essential nutrients? Curr Opin Endocrinol Diabetes Obes. 2020;27:312-316. 
  125. O'Hearn LA. A survey of improvements experienced on a carnivore diet compared to only carbohydrate restriction. Open Science Forum website. Published February 12, 2019. Accessed May 17, 2021. doi:10.17605/OSF.IO/5FU4D 
  126. Williams P. Nutritional composition of red meat. Nutrition & Dietetics. 2007;64(suppl 4):S113-S119. 
  127. Biel W, Czerniawska-Piątkowska E, Kowalczyk A. Offal chemical composition from veal, beef, and lamb maintained in organic production systems. Animals (Basel). 2019;9:489. 
  128. Elmadfa I, Meyer AL. The role of the status of selected micronutrients in shaping the immune function. Endocr Metab Immune Disord Drug Targets. 2019;19:1100-1115. 
  129. Babizhayev M. Treatment of skin aging and photoaging with innovative oral dosage forms of nonhydrolized carnosine and carcinine. Int J Clin Derm Res. 2017;5:116-143. 
  130. Danby FW. Nutrition and aging skin: sugar and glycation. Clin Dermatol. 2010;28:409-411. 
  131. Siefken W, Carstensen S, Springmann G, et al. Role of taurine accumulation in keratinocyte hydration. J Invest Dermatol. 2003;121:354-361. 
  132. Vollmer DL, West VA, Lephart ED. Enhancing skin health: by oral administration of natural compounds and minerals with implications to the dermal microbiome. Int J Mol Sci. 2018;19:3059. 
  133. Fischer F, Achterberg V, März A, et al. Folic acid and creatineimprove the firmness of human skin in vivo. J Cosmet Dermatol. 2011;10:15-23. 
  134. Blatt T, Lenz H, Weber T. Topical application of creatine is multibeneficial for human skin. J Am Acad Dermatol. 2005;52:P32.
Issue
cutis - 107(6)
Issue
cutis - 107(6)
Page Number
308-314
Page Number
308-314
Publications
Publications
Topics
Article Type
Sections
Inside the Article

Practice Points

  • Patients are increasingly interested in dietary modifications that may influence skin appearance and aid in the treatment of cutaneous disease.
  • Although evidence-based dietary recommendations currently are limited, it is important for dermatologists to be aware of the varied and nuanced dietary interventions employed by patients.
  • There remains a lack of randomized controlled trials assessing the efficacy of various dietary interventions in the dermatologic setting.
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
Article PDF Media

Review finds diverse outcomes in clinical trials of rosacea

Article Type
Changed
Thu, 05/27/2021 - 15:58

There is an unmet need to standardize outcomes that are reported in clinical trials of rosacea, according to authors of a new systematic review of rosacea treatment studies.

Sarah A. Ibrahim predoctoral research fellow in Northwestern University's Department of Dermatology
Sarah A. Ibrahim

“Rosacea is a chronic dermatologic condition that affects 16 million Americans,” one of the study authors, Sarah A. Ibrahim, told this news organization after the annual conference of the American Society for Laser Medicine and Surgery. “The features of rosacea, such as inflammatory lesions, redness, burning sensations, and swelling, can have a negative impact on the quality of life for many patients. Additionally, patients with rosacea are at an increased risk for other conditions such as autoimmune diseases, like inflammatory bowel disease.”

In an effort led by principal investigator Murad Alam, MD, vice chair of the department of dermatology at Northwestern University, Chicago, Ms. Ibrahim conducted a systematic review to identify all outcomes that have previously been reported in clinical trials of rosacea, as part of the development of the core outcome set established by the Measurement of Priority Outcome Variables in Dermatologic Surgery (IMPROVED) group. “This has not been done before and is an important first step in understanding what outcomes should be measured in every future clinical study of rosacea,” said Ms. Ibrahim, a medical student at Northwestern University, and predoctoral research fellow in Northwestern’s department of dermatology.



The researchers limited their analysis to randomized, controlled trials of rosacea interventions published between 2010 and 2020 and categorized outcomes into domains based on similar themes.

A total of 58 studies were included in the systematic review, of which 7 (12%) evaluated laser-based interventions. The researchers identified 55 unique outcomes that encompassed eight domains: Quality of life, treatment effects, patient perception of health, clinical assessment, acceptance of care, laboratory assessment, physiological skin assessment, and patient satisfaction. Of the eight domains, clinical assessment-related outcomes were measured in all studies. Nontransient erythema was the most commonly reported outcome (43 studies, 78%), followed by inflammatory lesions (36 studies, 65%) and telangiectasia (22 studies, 40%).

Outcomes pertaining to treatment effects such as adverse events were measured in 49 of the 55 studies (89%), while patient-reported outcomes were measured in 21 (38%). Quality of life and patient satisfaction were reported in 18 (33%) and 13 (24%) studies, respectively.

sruilk/shutterstock

“There were two main take-home messages of our study,” said Ms. Ibrahim, who presented the results at the meeting. “The first is that there is a wide range of outcomes that are reported in clinical trials of rosacea therapies. Second, that there is a need to standardize the outcomes that are reported in clinical trials of rosacea, in order to be able to combine the results from different studies to better understand which interventions for rosacea are most effective.”

She acknowledged certain limitations of the review, including that other trials related to the topic were not included. “Because of the date range and types of studies that we used to narrow down our search, it is possible that additional outcomes were reported in studies that were not included here,” she said.

“This is a very important study because rosacea is a very common condition and one that I have seen more frequently in clinic since the pandemic started,” said Omar Ibrahimi, PhD, MD, a dermatologist with the Connecticut Skin Institute in Stamford, who was asked to comment on the work. “One of the limitations with rosacea studies is that the studies done are often fairly small and the outcome measures are heterogenous. The current study by Ibrahim and coworkers does a wonderful job of highlighting the various outcomes measures used to measure the success of rosacea treatments with energy-based devices.”

This information, he added, “will be very useful for further research studies because it forms the basis for formulating a set of core outcome measures to judge treatment interventions with consensus input from a variety of key opinion leaders. This will prove to be valuable because if we can have a uniform set of outcome measures to judge rosacea treatments with then we will be able to compare the results from different studies better.”

Ms. Ibrahim and colleagues reported having no relevant financial disclosures. Dr. Ibrahimi disclosed that he has been a speaker for both Candela and Cutera and he is currently on the medical advisory board for Cutera.
 

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

There is an unmet need to standardize outcomes that are reported in clinical trials of rosacea, according to authors of a new systematic review of rosacea treatment studies.

Sarah A. Ibrahim predoctoral research fellow in Northwestern University's Department of Dermatology
Sarah A. Ibrahim

“Rosacea is a chronic dermatologic condition that affects 16 million Americans,” one of the study authors, Sarah A. Ibrahim, told this news organization after the annual conference of the American Society for Laser Medicine and Surgery. “The features of rosacea, such as inflammatory lesions, redness, burning sensations, and swelling, can have a negative impact on the quality of life for many patients. Additionally, patients with rosacea are at an increased risk for other conditions such as autoimmune diseases, like inflammatory bowel disease.”

In an effort led by principal investigator Murad Alam, MD, vice chair of the department of dermatology at Northwestern University, Chicago, Ms. Ibrahim conducted a systematic review to identify all outcomes that have previously been reported in clinical trials of rosacea, as part of the development of the core outcome set established by the Measurement of Priority Outcome Variables in Dermatologic Surgery (IMPROVED) group. “This has not been done before and is an important first step in understanding what outcomes should be measured in every future clinical study of rosacea,” said Ms. Ibrahim, a medical student at Northwestern University, and predoctoral research fellow in Northwestern’s department of dermatology.



The researchers limited their analysis to randomized, controlled trials of rosacea interventions published between 2010 and 2020 and categorized outcomes into domains based on similar themes.

A total of 58 studies were included in the systematic review, of which 7 (12%) evaluated laser-based interventions. The researchers identified 55 unique outcomes that encompassed eight domains: Quality of life, treatment effects, patient perception of health, clinical assessment, acceptance of care, laboratory assessment, physiological skin assessment, and patient satisfaction. Of the eight domains, clinical assessment-related outcomes were measured in all studies. Nontransient erythema was the most commonly reported outcome (43 studies, 78%), followed by inflammatory lesions (36 studies, 65%) and telangiectasia (22 studies, 40%).

Outcomes pertaining to treatment effects such as adverse events were measured in 49 of the 55 studies (89%), while patient-reported outcomes were measured in 21 (38%). Quality of life and patient satisfaction were reported in 18 (33%) and 13 (24%) studies, respectively.

sruilk/shutterstock

“There were two main take-home messages of our study,” said Ms. Ibrahim, who presented the results at the meeting. “The first is that there is a wide range of outcomes that are reported in clinical trials of rosacea therapies. Second, that there is a need to standardize the outcomes that are reported in clinical trials of rosacea, in order to be able to combine the results from different studies to better understand which interventions for rosacea are most effective.”

She acknowledged certain limitations of the review, including that other trials related to the topic were not included. “Because of the date range and types of studies that we used to narrow down our search, it is possible that additional outcomes were reported in studies that were not included here,” she said.

“This is a very important study because rosacea is a very common condition and one that I have seen more frequently in clinic since the pandemic started,” said Omar Ibrahimi, PhD, MD, a dermatologist with the Connecticut Skin Institute in Stamford, who was asked to comment on the work. “One of the limitations with rosacea studies is that the studies done are often fairly small and the outcome measures are heterogenous. The current study by Ibrahim and coworkers does a wonderful job of highlighting the various outcomes measures used to measure the success of rosacea treatments with energy-based devices.”

This information, he added, “will be very useful for further research studies because it forms the basis for formulating a set of core outcome measures to judge treatment interventions with consensus input from a variety of key opinion leaders. This will prove to be valuable because if we can have a uniform set of outcome measures to judge rosacea treatments with then we will be able to compare the results from different studies better.”

Ms. Ibrahim and colleagues reported having no relevant financial disclosures. Dr. Ibrahimi disclosed that he has been a speaker for both Candela and Cutera and he is currently on the medical advisory board for Cutera.
 

There is an unmet need to standardize outcomes that are reported in clinical trials of rosacea, according to authors of a new systematic review of rosacea treatment studies.

Sarah A. Ibrahim predoctoral research fellow in Northwestern University's Department of Dermatology
Sarah A. Ibrahim

“Rosacea is a chronic dermatologic condition that affects 16 million Americans,” one of the study authors, Sarah A. Ibrahim, told this news organization after the annual conference of the American Society for Laser Medicine and Surgery. “The features of rosacea, such as inflammatory lesions, redness, burning sensations, and swelling, can have a negative impact on the quality of life for many patients. Additionally, patients with rosacea are at an increased risk for other conditions such as autoimmune diseases, like inflammatory bowel disease.”

In an effort led by principal investigator Murad Alam, MD, vice chair of the department of dermatology at Northwestern University, Chicago, Ms. Ibrahim conducted a systematic review to identify all outcomes that have previously been reported in clinical trials of rosacea, as part of the development of the core outcome set established by the Measurement of Priority Outcome Variables in Dermatologic Surgery (IMPROVED) group. “This has not been done before and is an important first step in understanding what outcomes should be measured in every future clinical study of rosacea,” said Ms. Ibrahim, a medical student at Northwestern University, and predoctoral research fellow in Northwestern’s department of dermatology.



The researchers limited their analysis to randomized, controlled trials of rosacea interventions published between 2010 and 2020 and categorized outcomes into domains based on similar themes.

A total of 58 studies were included in the systematic review, of which 7 (12%) evaluated laser-based interventions. The researchers identified 55 unique outcomes that encompassed eight domains: Quality of life, treatment effects, patient perception of health, clinical assessment, acceptance of care, laboratory assessment, physiological skin assessment, and patient satisfaction. Of the eight domains, clinical assessment-related outcomes were measured in all studies. Nontransient erythema was the most commonly reported outcome (43 studies, 78%), followed by inflammatory lesions (36 studies, 65%) and telangiectasia (22 studies, 40%).

Outcomes pertaining to treatment effects such as adverse events were measured in 49 of the 55 studies (89%), while patient-reported outcomes were measured in 21 (38%). Quality of life and patient satisfaction were reported in 18 (33%) and 13 (24%) studies, respectively.

sruilk/shutterstock

“There were two main take-home messages of our study,” said Ms. Ibrahim, who presented the results at the meeting. “The first is that there is a wide range of outcomes that are reported in clinical trials of rosacea therapies. Second, that there is a need to standardize the outcomes that are reported in clinical trials of rosacea, in order to be able to combine the results from different studies to better understand which interventions for rosacea are most effective.”

She acknowledged certain limitations of the review, including that other trials related to the topic were not included. “Because of the date range and types of studies that we used to narrow down our search, it is possible that additional outcomes were reported in studies that were not included here,” she said.

“This is a very important study because rosacea is a very common condition and one that I have seen more frequently in clinic since the pandemic started,” said Omar Ibrahimi, PhD, MD, a dermatologist with the Connecticut Skin Institute in Stamford, who was asked to comment on the work. “One of the limitations with rosacea studies is that the studies done are often fairly small and the outcome measures are heterogenous. The current study by Ibrahim and coworkers does a wonderful job of highlighting the various outcomes measures used to measure the success of rosacea treatments with energy-based devices.”

This information, he added, “will be very useful for further research studies because it forms the basis for formulating a set of core outcome measures to judge treatment interventions with consensus input from a variety of key opinion leaders. This will prove to be valuable because if we can have a uniform set of outcome measures to judge rosacea treatments with then we will be able to compare the results from different studies better.”

Ms. Ibrahim and colleagues reported having no relevant financial disclosures. Dr. Ibrahimi disclosed that he has been a speaker for both Candela and Cutera and he is currently on the medical advisory board for Cutera.
 

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM ASLMS 2021

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

Oral sarecycline promising for papulopustular rosacea

Article Type
Changed
Wed, 03/31/2021 - 09:25

Oral sarecycline proved rapidly effective for the treatment of moderate to severe papulopustular rosacea in a proof-of-concept pilot study, Linda Stein Gold, MD, said at Innovations in Dermatology: Virtual Spring Conference 2021.

Patrick McNamara Photography
Dr. Linda Stein Gold

The oral broad-spectrum second-generation tetracyclines doxycycline and minocycline have long been considered first-line therapy for papulopustular rosacea that isn’t cleared using topical agents. But the widespread use of these oral tetracyclines has encouraged the development of antimicrobial resistance. In contrast, sarecycline (Seysara) is a third-generation, narrow-spectrum tetracycline designed to minimize antibiotic resistance. The Food and Drug Administration approved the drug for treatment of moderate to severe acne vulgaris in 2018.

At the meeting, Dr. Stein Gold, director of dermatology clinical research at the Henry Ford Health System in Detroit, highlighted a recent pilot study of oral sarecycline for papulopustular rosacea carried out by James Q. Del Rosso, DO, of Las Vegas and coinvestigators. Although she wasn’t involved in the study, she is a veteran clinical trialist with vast experience leading studies of new therapies for rosacea, acne, and other major dermatologic disorders.

The 12-week, prospective, investigator-blinded study included 97 adults with moderate to severe papulopustular rosacea; 72 were randomized to weight-based dosing of once-daily sarecycline, while the 25 controls took a daily oral vitamin.



One coprimary endpoint was achievement of an Investigator Global Assessment score of 0 or 1, meaning clear or almost clear skin, at week 12. The rates were 75% in the sarecycline group and 16% in controls. The other coprimary endpoint was the percent reduction from baseline to week 12 in inflammatory lesion count. Here again, there was a statistically significant difference in favor of the third-generation tetracycline derivative, which achieved an 80% reduction, compared with 50% in the control group.

Of note, the difference was already significant at the first evaluation at week 4, with a 58% reduction in inflammatory lesions in the sarecycline group versus 31% decrease in controls, Dr. Stein Gold observed at the conference, sponsored by MedscapeLIVE! and the producers of the Hawaii Dermatology Seminar and Caribbean Dermatology Symposium.

Also at week 12, 96% of patients on sarecycline reported having no or only trace symptoms of facial burning, 63% had no or only trace facial erythema, and 94% had no or trace facial itch, compared with 76%, 12%, and 76% of controls, respectively. The sarecycline group was also significantly more likely to report no or trace skin dryness and oiliness.

The side-effect profile was favorable and the same as encountered with the use of sarecycline for acne: no major photosensitivity issues, no serious adverse events, and only 2 of the original 75 patients in the active-treatment arm discontinued sarecycline for treatment-emergent headache or gastroenteritis considered “probably” related to the study drug. The investigators deemed further studies of sarecycline for rosacea to be warranted as a potential expanded indication.

Aiming for clear skin rather than ‘almost clear’

Dr. Stein Gold shared her mantra for rosacea therapy: “Always aim for clear skin.”

She cited a study led by Guy Webster, MD, professor of dermatology, Thomas Jefferson University, Philadelphia, in which he and his coinvestigators looked at the durability of treatment response in a pooled analysis of 1,366 rosacea patients in four clinical trials. If patients improved to “almost clear” after treatment, their median time to relapse was 3 months; if they reached “clear,” it was more than 8 months. Also, more clear patients rated their outcomes as excellent and reported that their skin disease no longer had any effect on their quality of life.

“That’s more than a 5-month difference,” Dr. Stein Gold noted. “It shows the importance of really striving to get that skin completely clear.”

The sarecycline study was funded by Almirall, which markets the antibiotic. Dr. Stein Gold, who has no financial relationship with Almirall, has received research funding from and/or served as a consultant to roughly a dozen other pharmaceutical companies. MedscapeLIVE! and this news organization are owned by the same parent company.
 

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

Oral sarecycline proved rapidly effective for the treatment of moderate to severe papulopustular rosacea in a proof-of-concept pilot study, Linda Stein Gold, MD, said at Innovations in Dermatology: Virtual Spring Conference 2021.

Patrick McNamara Photography
Dr. Linda Stein Gold

The oral broad-spectrum second-generation tetracyclines doxycycline and minocycline have long been considered first-line therapy for papulopustular rosacea that isn’t cleared using topical agents. But the widespread use of these oral tetracyclines has encouraged the development of antimicrobial resistance. In contrast, sarecycline (Seysara) is a third-generation, narrow-spectrum tetracycline designed to minimize antibiotic resistance. The Food and Drug Administration approved the drug for treatment of moderate to severe acne vulgaris in 2018.

At the meeting, Dr. Stein Gold, director of dermatology clinical research at the Henry Ford Health System in Detroit, highlighted a recent pilot study of oral sarecycline for papulopustular rosacea carried out by James Q. Del Rosso, DO, of Las Vegas and coinvestigators. Although she wasn’t involved in the study, she is a veteran clinical trialist with vast experience leading studies of new therapies for rosacea, acne, and other major dermatologic disorders.

The 12-week, prospective, investigator-blinded study included 97 adults with moderate to severe papulopustular rosacea; 72 were randomized to weight-based dosing of once-daily sarecycline, while the 25 controls took a daily oral vitamin.



One coprimary endpoint was achievement of an Investigator Global Assessment score of 0 or 1, meaning clear or almost clear skin, at week 12. The rates were 75% in the sarecycline group and 16% in controls. The other coprimary endpoint was the percent reduction from baseline to week 12 in inflammatory lesion count. Here again, there was a statistically significant difference in favor of the third-generation tetracycline derivative, which achieved an 80% reduction, compared with 50% in the control group.

Of note, the difference was already significant at the first evaluation at week 4, with a 58% reduction in inflammatory lesions in the sarecycline group versus 31% decrease in controls, Dr. Stein Gold observed at the conference, sponsored by MedscapeLIVE! and the producers of the Hawaii Dermatology Seminar and Caribbean Dermatology Symposium.

Also at week 12, 96% of patients on sarecycline reported having no or only trace symptoms of facial burning, 63% had no or only trace facial erythema, and 94% had no or trace facial itch, compared with 76%, 12%, and 76% of controls, respectively. The sarecycline group was also significantly more likely to report no or trace skin dryness and oiliness.

The side-effect profile was favorable and the same as encountered with the use of sarecycline for acne: no major photosensitivity issues, no serious adverse events, and only 2 of the original 75 patients in the active-treatment arm discontinued sarecycline for treatment-emergent headache or gastroenteritis considered “probably” related to the study drug. The investigators deemed further studies of sarecycline for rosacea to be warranted as a potential expanded indication.

Aiming for clear skin rather than ‘almost clear’

Dr. Stein Gold shared her mantra for rosacea therapy: “Always aim for clear skin.”

She cited a study led by Guy Webster, MD, professor of dermatology, Thomas Jefferson University, Philadelphia, in which he and his coinvestigators looked at the durability of treatment response in a pooled analysis of 1,366 rosacea patients in four clinical trials. If patients improved to “almost clear” after treatment, their median time to relapse was 3 months; if they reached “clear,” it was more than 8 months. Also, more clear patients rated their outcomes as excellent and reported that their skin disease no longer had any effect on their quality of life.

“That’s more than a 5-month difference,” Dr. Stein Gold noted. “It shows the importance of really striving to get that skin completely clear.”

The sarecycline study was funded by Almirall, which markets the antibiotic. Dr. Stein Gold, who has no financial relationship with Almirall, has received research funding from and/or served as a consultant to roughly a dozen other pharmaceutical companies. MedscapeLIVE! and this news organization are owned by the same parent company.
 

Oral sarecycline proved rapidly effective for the treatment of moderate to severe papulopustular rosacea in a proof-of-concept pilot study, Linda Stein Gold, MD, said at Innovations in Dermatology: Virtual Spring Conference 2021.

Patrick McNamara Photography
Dr. Linda Stein Gold

The oral broad-spectrum second-generation tetracyclines doxycycline and minocycline have long been considered first-line therapy for papulopustular rosacea that isn’t cleared using topical agents. But the widespread use of these oral tetracyclines has encouraged the development of antimicrobial resistance. In contrast, sarecycline (Seysara) is a third-generation, narrow-spectrum tetracycline designed to minimize antibiotic resistance. The Food and Drug Administration approved the drug for treatment of moderate to severe acne vulgaris in 2018.

At the meeting, Dr. Stein Gold, director of dermatology clinical research at the Henry Ford Health System in Detroit, highlighted a recent pilot study of oral sarecycline for papulopustular rosacea carried out by James Q. Del Rosso, DO, of Las Vegas and coinvestigators. Although she wasn’t involved in the study, she is a veteran clinical trialist with vast experience leading studies of new therapies for rosacea, acne, and other major dermatologic disorders.

The 12-week, prospective, investigator-blinded study included 97 adults with moderate to severe papulopustular rosacea; 72 were randomized to weight-based dosing of once-daily sarecycline, while the 25 controls took a daily oral vitamin.



One coprimary endpoint was achievement of an Investigator Global Assessment score of 0 or 1, meaning clear or almost clear skin, at week 12. The rates were 75% in the sarecycline group and 16% in controls. The other coprimary endpoint was the percent reduction from baseline to week 12 in inflammatory lesion count. Here again, there was a statistically significant difference in favor of the third-generation tetracycline derivative, which achieved an 80% reduction, compared with 50% in the control group.

Of note, the difference was already significant at the first evaluation at week 4, with a 58% reduction in inflammatory lesions in the sarecycline group versus 31% decrease in controls, Dr. Stein Gold observed at the conference, sponsored by MedscapeLIVE! and the producers of the Hawaii Dermatology Seminar and Caribbean Dermatology Symposium.

Also at week 12, 96% of patients on sarecycline reported having no or only trace symptoms of facial burning, 63% had no or only trace facial erythema, and 94% had no or trace facial itch, compared with 76%, 12%, and 76% of controls, respectively. The sarecycline group was also significantly more likely to report no or trace skin dryness and oiliness.

The side-effect profile was favorable and the same as encountered with the use of sarecycline for acne: no major photosensitivity issues, no serious adverse events, and only 2 of the original 75 patients in the active-treatment arm discontinued sarecycline for treatment-emergent headache or gastroenteritis considered “probably” related to the study drug. The investigators deemed further studies of sarecycline for rosacea to be warranted as a potential expanded indication.

Aiming for clear skin rather than ‘almost clear’

Dr. Stein Gold shared her mantra for rosacea therapy: “Always aim for clear skin.”

She cited a study led by Guy Webster, MD, professor of dermatology, Thomas Jefferson University, Philadelphia, in which he and his coinvestigators looked at the durability of treatment response in a pooled analysis of 1,366 rosacea patients in four clinical trials. If patients improved to “almost clear” after treatment, their median time to relapse was 3 months; if they reached “clear,” it was more than 8 months. Also, more clear patients rated their outcomes as excellent and reported that their skin disease no longer had any effect on their quality of life.

“That’s more than a 5-month difference,” Dr. Stein Gold noted. “It shows the importance of really striving to get that skin completely clear.”

The sarecycline study was funded by Almirall, which markets the antibiotic. Dr. Stein Gold, who has no financial relationship with Almirall, has received research funding from and/or served as a consultant to roughly a dozen other pharmaceutical companies. MedscapeLIVE! and this news organization are owned by the same parent company.
 

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM INNOVATIONS IN DERMATOLOGY

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