User login
Cutis is a peer-reviewed clinical journal for the dermatologist, allergist, and general practitioner published monthly since 1965. Concise clinical articles present the practical side of dermatology, helping physicians to improve patient care. Cutis is referenced in Index Medicus/MEDLINE and is written and edited by industry leaders.
ass lick
assault rifle
balls
ballsac
black jack
bleach
Boko Haram
bondage
causas
cheap
child abuse
cocaine
compulsive behaviors
cost of miracles
cunt
Daech
display network stats
drug paraphernalia
explosion
fart
fda and death
fda AND warn
fda AND warning
fda AND warns
feom
fuck
gambling
gfc
gun
human trafficking
humira AND expensive
illegal
ISIL
ISIS
Islamic caliphate
Islamic state
madvocate
masturbation
mixed martial arts
MMA
molestation
national rifle association
NRA
nsfw
nuccitelli
pedophile
pedophilia
poker
porn
porn
pornography
psychedelic drug
recreational drug
sex slave rings
shit
slot machine
snort
substance abuse
terrorism
terrorist
texarkana
Texas hold 'em
UFC
section[contains(@class, 'nav-hidden')]
section[contains(@class, 'nav-hidden active')
A peer-reviewed, indexed journal for dermatologists with original research, image quizzes, cases and reviews, and columns.
Atopic Dermatitis Topical Therapies: Study of YouTube Videos as a Source of Patient Information
To the Editor:
Atopic dermatitis (eczema) affects approximately 20% of children worldwide.1 In atopic dermatitis management, patient education is crucial for optimal outcomes.2 The COVID-19 pandemic has impacted patient-physician interactions. To ensure safety of patients and physicians, visits may have been canceled, postponed, or conducted virtually, leaving less time for discussion and questions.3 As a consequence, patients may seek information about atopic dermatitis from alternative sources, including YouTube videos. We performed a cross-sectional study to analyze YouTube videos about topical treatments for atopic dermatitis.
During the week of July 16, 2020, we performed 4 private browser YouTube searches with default filters using the following terms: eczema topicals, eczema topical treatments, atopic dermatitis topicals, and atopic dermatitis topical treatments. For video selection, we defined topical treatments as topical corticosteroids, topical calcineurin inhibitors, crisaborole, emollients, wet wraps, and any prospective treatment topically administered. For each of the 4 searches, 2 researchers (A.M. and A.T.) independently examined the top 75 videos, yielding a total of 300 videos. Of them, 98 videos were duplicates, 19 videos were not about atopic dermatitis, and 91 videos were not about topical treatments, leaving a total of 92 videos for analysis (Figure 1).
For the 92 included videos, the length; upload year; number of views, likes, dislikes, and comments; interaction ratio (IR)(the sum of likes, dislikes, and comments divided by the number of views); and video content were determined. The videos were placed into mutually exclusive categories as follows: (1) patient experience, defined as a video about patient perspective; (2) professional source, defined as a video featuring a physician, physician extender, pharmacist, or scientist, or produced by a formal organization; or (3) other. The DISCERN Instrument was used for grading the reliability and quality of the 92 included videos. This instrument consists of 16 questions with the responses rated on a scale of 1 to 5.4 For analysis of DISCERN scores, patient experience and other videos were grouped together as nonprofessional source videos. A 2-sample t-test was used to compare DISCERN scores between professional source and nonprofessional source videos.
Most videos were uploaded in 2017 (n=19), 2018 (n=23), and 2019 (n=25), but 20 were uploaded in 2012-2016 and 5 were uploaded in 2020. The 92 videos had a mean length of 8 minutes and 35 seconds (range, 30 seconds to 62 minutes and 23 seconds).
Patient experience videos accounted for 23.9% (n=22) of videos. These videos discussed topical steroid withdrawal (TSW)(n=16), instructions for making emollients (n=2), and treatment successes (n=4). Professional source videos represented 67.4% (n=62) of videos. Of them, 40.3% (n=25) were physician oriented, defined as having extensive medical terminology or qualifying for continuing medical education credit. Three (4.8%) of the professional source videos were sponsored by a drug company. Other constituted the remaining 8.7% (n=8) of videos. Patient experience videos had more views (median views [interquartile range], 6865 [10,307]) and higher engagement (median IR [interquartile range], 0.038 [0.022]) than professional source videos (views: median views [interquartile range], 1052.5 [10,610.5]; engagement: median IR [interquartile range], 0.006 [0.008]).
Although less popular, professional source videos had a significantly higher DISCERN overall quality rating score (question 16) compared to those categorized as nonprofessional source (3.92 vs 1.53; P<.001). In contrast, nonprofessional source videos scored significantly higher on the quality-of-life question (question 13) compared to professional source videos (3.90 vs 2.56; P<.001)(eTable). (Three professional source videos were removed from YouTube before DISCERN scores could be assigned.)
Notably, 20.7% (n=19) of the 92 videos discussed TSW, and most of them were patient experiences (n=16). Other categories included topical steroids excluding TSW (n=11), steroid phobia (n=2), topical calcineurin inhibitors (n=2), crisaborole (n=6), news broadcast (n=7), wet wraps (n=5), product advertisement (n=7), and research (n=11)(Figure 2). Interestingly, there were no videos focusing on the calcineurin inhibitor black box warning.
Similar to prior studies, our results indicate preference for patient-generated videos over videos produced by or including a professional source.5 Additionally, only 3 of 19 videos about TSW were from a professional source, increasing the potential for patient misconceptions about topical corticosteroids. Future studies should examine the educational impact of patient-generated videos as well as features that make the patient experience videos more desirable for viewing.
- Mueller SM, Hongler VNS, Jungo P, et al. Fiction, falsehoods, and few facts: cross-sectional study on the content-related quality of atopic eczema-related videos on YouTube. J Med Internet Res. 2020;22:e15599. doi:10.2196/15599
- Torres T, Ferreira EO, Gonçalo M, et al. Update on atopic dermatitis. Acta Med Port. 2019;32:606-613. doi:10.20344/amp.11963
- Vogler SA, Lightner AL. Rethinking how we care for our patients in a time of social distancing during the COVID-19 pandemic. Br J Surg. 2020;107:937-939. doi:10.1002/bjs.11636
- The DISCERN Instrument. discern online. Accessed January 22, 2021. http://www.discern.org.uk/discern_instrument.php
- Pithadia DJ, Reynolds KA, Lee EB, et al. Dupilumab for atopic dermatitis: what are patients learning on YouTube? [published online April 16, 2020]. J Dermatolog Treat. doi:10.1080/09546634.2020.1755418
To the Editor:
Atopic dermatitis (eczema) affects approximately 20% of children worldwide.1 In atopic dermatitis management, patient education is crucial for optimal outcomes.2 The COVID-19 pandemic has impacted patient-physician interactions. To ensure safety of patients and physicians, visits may have been canceled, postponed, or conducted virtually, leaving less time for discussion and questions.3 As a consequence, patients may seek information about atopic dermatitis from alternative sources, including YouTube videos. We performed a cross-sectional study to analyze YouTube videos about topical treatments for atopic dermatitis.
During the week of July 16, 2020, we performed 4 private browser YouTube searches with default filters using the following terms: eczema topicals, eczema topical treatments, atopic dermatitis topicals, and atopic dermatitis topical treatments. For video selection, we defined topical treatments as topical corticosteroids, topical calcineurin inhibitors, crisaborole, emollients, wet wraps, and any prospective treatment topically administered. For each of the 4 searches, 2 researchers (A.M. and A.T.) independently examined the top 75 videos, yielding a total of 300 videos. Of them, 98 videos were duplicates, 19 videos were not about atopic dermatitis, and 91 videos were not about topical treatments, leaving a total of 92 videos for analysis (Figure 1).
For the 92 included videos, the length; upload year; number of views, likes, dislikes, and comments; interaction ratio (IR)(the sum of likes, dislikes, and comments divided by the number of views); and video content were determined. The videos were placed into mutually exclusive categories as follows: (1) patient experience, defined as a video about patient perspective; (2) professional source, defined as a video featuring a physician, physician extender, pharmacist, or scientist, or produced by a formal organization; or (3) other. The DISCERN Instrument was used for grading the reliability and quality of the 92 included videos. This instrument consists of 16 questions with the responses rated on a scale of 1 to 5.4 For analysis of DISCERN scores, patient experience and other videos were grouped together as nonprofessional source videos. A 2-sample t-test was used to compare DISCERN scores between professional source and nonprofessional source videos.
Most videos were uploaded in 2017 (n=19), 2018 (n=23), and 2019 (n=25), but 20 were uploaded in 2012-2016 and 5 were uploaded in 2020. The 92 videos had a mean length of 8 minutes and 35 seconds (range, 30 seconds to 62 minutes and 23 seconds).
Patient experience videos accounted for 23.9% (n=22) of videos. These videos discussed topical steroid withdrawal (TSW)(n=16), instructions for making emollients (n=2), and treatment successes (n=4). Professional source videos represented 67.4% (n=62) of videos. Of them, 40.3% (n=25) were physician oriented, defined as having extensive medical terminology or qualifying for continuing medical education credit. Three (4.8%) of the professional source videos were sponsored by a drug company. Other constituted the remaining 8.7% (n=8) of videos. Patient experience videos had more views (median views [interquartile range], 6865 [10,307]) and higher engagement (median IR [interquartile range], 0.038 [0.022]) than professional source videos (views: median views [interquartile range], 1052.5 [10,610.5]; engagement: median IR [interquartile range], 0.006 [0.008]).
Although less popular, professional source videos had a significantly higher DISCERN overall quality rating score (question 16) compared to those categorized as nonprofessional source (3.92 vs 1.53; P<.001). In contrast, nonprofessional source videos scored significantly higher on the quality-of-life question (question 13) compared to professional source videos (3.90 vs 2.56; P<.001)(eTable). (Three professional source videos were removed from YouTube before DISCERN scores could be assigned.)
Notably, 20.7% (n=19) of the 92 videos discussed TSW, and most of them were patient experiences (n=16). Other categories included topical steroids excluding TSW (n=11), steroid phobia (n=2), topical calcineurin inhibitors (n=2), crisaborole (n=6), news broadcast (n=7), wet wraps (n=5), product advertisement (n=7), and research (n=11)(Figure 2). Interestingly, there were no videos focusing on the calcineurin inhibitor black box warning.
Similar to prior studies, our results indicate preference for patient-generated videos over videos produced by or including a professional source.5 Additionally, only 3 of 19 videos about TSW were from a professional source, increasing the potential for patient misconceptions about topical corticosteroids. Future studies should examine the educational impact of patient-generated videos as well as features that make the patient experience videos more desirable for viewing.
To the Editor:
Atopic dermatitis (eczema) affects approximately 20% of children worldwide.1 In atopic dermatitis management, patient education is crucial for optimal outcomes.2 The COVID-19 pandemic has impacted patient-physician interactions. To ensure safety of patients and physicians, visits may have been canceled, postponed, or conducted virtually, leaving less time for discussion and questions.3 As a consequence, patients may seek information about atopic dermatitis from alternative sources, including YouTube videos. We performed a cross-sectional study to analyze YouTube videos about topical treatments for atopic dermatitis.
During the week of July 16, 2020, we performed 4 private browser YouTube searches with default filters using the following terms: eczema topicals, eczema topical treatments, atopic dermatitis topicals, and atopic dermatitis topical treatments. For video selection, we defined topical treatments as topical corticosteroids, topical calcineurin inhibitors, crisaborole, emollients, wet wraps, and any prospective treatment topically administered. For each of the 4 searches, 2 researchers (A.M. and A.T.) independently examined the top 75 videos, yielding a total of 300 videos. Of them, 98 videos were duplicates, 19 videos were not about atopic dermatitis, and 91 videos were not about topical treatments, leaving a total of 92 videos for analysis (Figure 1).
For the 92 included videos, the length; upload year; number of views, likes, dislikes, and comments; interaction ratio (IR)(the sum of likes, dislikes, and comments divided by the number of views); and video content were determined. The videos were placed into mutually exclusive categories as follows: (1) patient experience, defined as a video about patient perspective; (2) professional source, defined as a video featuring a physician, physician extender, pharmacist, or scientist, or produced by a formal organization; or (3) other. The DISCERN Instrument was used for grading the reliability and quality of the 92 included videos. This instrument consists of 16 questions with the responses rated on a scale of 1 to 5.4 For analysis of DISCERN scores, patient experience and other videos were grouped together as nonprofessional source videos. A 2-sample t-test was used to compare DISCERN scores between professional source and nonprofessional source videos.
Most videos were uploaded in 2017 (n=19), 2018 (n=23), and 2019 (n=25), but 20 were uploaded in 2012-2016 and 5 were uploaded in 2020. The 92 videos had a mean length of 8 minutes and 35 seconds (range, 30 seconds to 62 minutes and 23 seconds).
Patient experience videos accounted for 23.9% (n=22) of videos. These videos discussed topical steroid withdrawal (TSW)(n=16), instructions for making emollients (n=2), and treatment successes (n=4). Professional source videos represented 67.4% (n=62) of videos. Of them, 40.3% (n=25) were physician oriented, defined as having extensive medical terminology or qualifying for continuing medical education credit. Three (4.8%) of the professional source videos were sponsored by a drug company. Other constituted the remaining 8.7% (n=8) of videos. Patient experience videos had more views (median views [interquartile range], 6865 [10,307]) and higher engagement (median IR [interquartile range], 0.038 [0.022]) than professional source videos (views: median views [interquartile range], 1052.5 [10,610.5]; engagement: median IR [interquartile range], 0.006 [0.008]).
Although less popular, professional source videos had a significantly higher DISCERN overall quality rating score (question 16) compared to those categorized as nonprofessional source (3.92 vs 1.53; P<.001). In contrast, nonprofessional source videos scored significantly higher on the quality-of-life question (question 13) compared to professional source videos (3.90 vs 2.56; P<.001)(eTable). (Three professional source videos were removed from YouTube before DISCERN scores could be assigned.)
Notably, 20.7% (n=19) of the 92 videos discussed TSW, and most of them were patient experiences (n=16). Other categories included topical steroids excluding TSW (n=11), steroid phobia (n=2), topical calcineurin inhibitors (n=2), crisaborole (n=6), news broadcast (n=7), wet wraps (n=5), product advertisement (n=7), and research (n=11)(Figure 2). Interestingly, there were no videos focusing on the calcineurin inhibitor black box warning.
Similar to prior studies, our results indicate preference for patient-generated videos over videos produced by or including a professional source.5 Additionally, only 3 of 19 videos about TSW were from a professional source, increasing the potential for patient misconceptions about topical corticosteroids. Future studies should examine the educational impact of patient-generated videos as well as features that make the patient experience videos more desirable for viewing.
- Mueller SM, Hongler VNS, Jungo P, et al. Fiction, falsehoods, and few facts: cross-sectional study on the content-related quality of atopic eczema-related videos on YouTube. J Med Internet Res. 2020;22:e15599. doi:10.2196/15599
- Torres T, Ferreira EO, Gonçalo M, et al. Update on atopic dermatitis. Acta Med Port. 2019;32:606-613. doi:10.20344/amp.11963
- Vogler SA, Lightner AL. Rethinking how we care for our patients in a time of social distancing during the COVID-19 pandemic. Br J Surg. 2020;107:937-939. doi:10.1002/bjs.11636
- The DISCERN Instrument. discern online. Accessed January 22, 2021. http://www.discern.org.uk/discern_instrument.php
- Pithadia DJ, Reynolds KA, Lee EB, et al. Dupilumab for atopic dermatitis: what are patients learning on YouTube? [published online April 16, 2020]. J Dermatolog Treat. doi:10.1080/09546634.2020.1755418
- Mueller SM, Hongler VNS, Jungo P, et al. Fiction, falsehoods, and few facts: cross-sectional study on the content-related quality of atopic eczema-related videos on YouTube. J Med Internet Res. 2020;22:e15599. doi:10.2196/15599
- Torres T, Ferreira EO, Gonçalo M, et al. Update on atopic dermatitis. Acta Med Port. 2019;32:606-613. doi:10.20344/amp.11963
- Vogler SA, Lightner AL. Rethinking how we care for our patients in a time of social distancing during the COVID-19 pandemic. Br J Surg. 2020;107:937-939. doi:10.1002/bjs.11636
- The DISCERN Instrument. discern online. Accessed January 22, 2021. http://www.discern.org.uk/discern_instrument.php
- Pithadia DJ, Reynolds KA, Lee EB, et al. Dupilumab for atopic dermatitis: what are patients learning on YouTube? [published online April 16, 2020]. J Dermatolog Treat. doi:10.1080/09546634.2020.1755418
Practice Points
- YouTube is a readily accessible resource for educating patients about topical treatments for atopic dermatitis.
- Although professional source videos comprised a larger percentage of the videos included within our study, patient experience videos had a higher number of views and engagement.
- Twenty-one percent (19/92) of the videos examined in our study discussed topical steroid withdrawal, and the majority of them were patient experience videos.
Plant Dermatitis: More Than Just Poison Ivy
Plants can contribute to a variety of dermatoses. The Toxicodendron genus, which includes poison ivy, poison oak, and poison sumac, is a well-known and common cause of allergic contact dermatitis (ACD), but many other plants can cause direct or airborne contact dermatitis, especially in gardeners, florists, and farmers. This article provides an overview of different plant-related dermatoses and culprit plants as well as how these dermatoses should be diagnosed and treated.
Epidemiology
Plant dermatoses affect more than 50 million individuals each year.1,2 In the United States, the Toxicodendron genus causes ACD in more than 70% of exposed individuals, leading to medical visits.3 An urgent care visit for a plant-related dermatitis is estimated to cost $168, while an emergency department visit can cost 3 times as much.4 Although less common, Compositae plants are another important culprit of plant dermatitis, particularly in gardeners, florists, and farmers. Data from the 2017-2018 North American Contact Dermatitis Group screening series (N=4947) showed sesquiterpene lactones and Compositae to be positive in 0.5% of patch-tested patients.5
Plant Dermatitis Classifications
Plant dermatitis can be classified into 5 main categories: ACD, mechanical irritant contact dermatitis, chemical irritant contact dermatitis, light-mediated dermatitis, and pseudophytodermatitis.6
Allergic contact dermatitis is an immune-mediated type IV delayed hypersensitivity reaction. The common molecular allergens in plants include phenols, α-methylene-γ-butyrolactones, quinones, terpenes, disulfides, isothiocyanates, and polyacetylenic derivatives.6
Plant contact dermatitis due to mechanical and chemical irritants is precipitated by multiple mechanisms, including disruption of the epidermal barrier and subsequent cytokine release from keratinocytes.7 Nonimmunologic contact urticaria from plants is thought to be a type of irritant reaction precipitated by mechanical or chemical trauma.8
Light-mediated dermatitis includes phytophotodermatitis and photoallergic contact dermatitis. Phytophotodermatitis is a phototoxic reaction triggered by exposure to both plant-derived furanocoumarin and UVA light.9 By contrast, photoallergic contact dermatitis is a delayed hypersensitivity reaction from prior sensitization to a light-activated antigen.10
Pseudophytodermatitis, as its name implies, is not truly mediated by an allergen or irritant intrinsic to the plant but rather by dyes, waxes, insecticides, or arthropods that inhabit the plant or are secondarily applied.6
Common Plant Allergens
Anacardiaceae Family
Most of the allergenic plants within the Anacardiaceae family belong to the Toxicodendron genus, which encompasses poison ivy (Toxicodendron radicans), poison oak (Toxicodendron pubescens,Toxicodendron quercifolium, Toxicodendron diversiloum), and poison sumac (Toxicodendron vernix). Poison ivy is the celebrity of the Anacardiaceae family and contributes to most cases of plant-related ACD. It is found in every state in the continental United States. Poison oak is another common culprit found in the western and southeastern United States.11 Plants within the Anacardiaceae family contain an oleoresin called urushiol, which is the primary sensitizing substance. Although poison ivy and poison oak grow well in full sun to partial shade, poison sumac typically is found in damp swampy areas east of the Rocky Mountains. Most cases of ACD related to Anacardiaceae species are due to direct contact with urushiol from a Toxicodendron plant, but burning of brush containing Toxicodendron can cause airborne exposure when urushiol oil is carried by smoke particles.12 Sensitization to Toxicodendron can cause ACD to other Anacardiaceae species such as the Japanese lacquer tree (Toxicodendron vernicifluum), mango tree (Mangifera indica), cashew tree (Anacardium occidentale), and Indian marking nut tree (Semecarpus anacardium).6 Cross-reactions to components of the ginkgo tree (Ginkgo biloba) also are possible.
Toxicodendron plants can be more easily identified and avoided with knowledge of their characteristic leaf patterns. The most dependable way to identify poison ivy and poison oak species is to look for plants with 3 leaves, giving rise to the common saying, “Leaves of three, leave them be.” Poison sumac plants have groups of 7 to 13 leaves arranged as pairs along a central rib. Another helpful finding is a black deposit that Toxicodendron species leave behind following trauma to the leaves. Urushiol oxidizes when exposed to air and turns into a black deposit that can be seen on damaged leaves themselves or can be demonstrated in a black spot test to verify if a plant is a Toxicodendron species. The test is performed by gathering (carefully, without direct contact) a few leaves in a paper towel and crushing them to release sap. Within minutes, the sap will turn black if the plant is indeed a Toxicodendron species.13Pruritic, edematous, erythematous papules, plaques, and eventual vesicles in a linear distribution are suspicious for Toxicodendron exposure. Although your pet will not develop Toxicodendron ACD, oleoresin-contaminated pets can transfer the oils to their owners after coming into contact with these plants. Toxicodendron dermatitis also can be acquired from oleoresin-contaminated fomites such as clothing and shoes worn in the garden or when hiking. Toxicodendron dermatitis can appear at different sites on the body at different times depending on the amount of oleoresin exposure as well as epidermal thickness. For example, the oleoresin can be transferred from the hands to body areas with a thinner stratum corneum (eg, genitalia) and cause subsequent dermatitis.1
Compositae Family
The Compositae family (also known as Asteraceae) is a large plant family with more than 20,000 species, including numerous weeds, wildflowers, and vegetables. The flowers, leaves, stems, and pollens of the Compositae family are coated by cyclic esters called sesquiterpene lactones. Mitchell and Dupuis14 showed that sesquiterpene lactones are the allergens responsible for ACD to various Compositae plants, including ragweed (Ambrosia), sneezeweed (Helenium), and chrysanthemums (Chrysanthemum). Common Compositae vegetables such as lettuce (Lactuca sativa) have been reported to cause ACD in chefs, grocery store produce handlers, gardeners, and even owners of lettuce-eating pet guinea pigs and turtles.15 Similarly, artichokes (Cynara scolymus) can cause ACD in gardeners.16 Exposure to Compositae species also has been implicated in photoallergic reactions, and studies have demonstrated that some patients with chronic actinic dermatitis also have positive patch test reactions to Compositae species and/or sesquiterpene lactones.17,18
In addition to direct contact with Compositae plants, airborne exposure to sesquiterpene lactones can cause ACD.14 The pattern of airborne contact dermatitis typically involves exposed areas such as the eyelids, central face, and/or neck. The beak sign also can be a clue to airborne contact dermatitis, which involves dermatitis of the face that spares the nasal tip and/or nasal ridge. It is thought that the beak sign may result from increased sebaceous gland concentration on the nose, which prevents penetration of allergens and irritants.19 Unlike photoallergic contact dermatitis, which also can involve the face, airborne ACD frequently involves photoprotected areas such as the submandibular chin and the upper lip. Davies and Kersey20 reported the case of a groundsman who was cutting grass with dandelions (Taraxacum officinale) and was found to have associated airborne ACD of the face, neck, and forearms due to Compositae allergy. In a different setting, the aromas of chamomile (Matricaria chamomilla) have been reported to cause airborne ACD in a tea drinker.21 Paulsen22 found that ingestion of chamomile tea can induce systemic ACD in sensitized individuals.
Alstroemeriaceae, Liliaceae, and Primulaceae
Florists are exposed to many plant species and have a high prevalence of ACD. Thiboutot et al23 found that 15 of 57 (26%) floral workers experienced hand dermatitis that cleared with time away from work. The Peruvian lily (Alstroemeria, Alstroemeriaceae family), which contains tuliposide A, was found to be the leading cause of sensitization.23 Tulips (Tulipa, Liliaceae family), as the flower name suggests, also contain tuliposide A, which along with mechanical irritation from the course tecta fibers on the bulbs lead to a dermatitis known as tulip fingers.24,25 Poison primrose (Primula obconica, Primulaceae family), cultivated for its highly colorful flowers, contains the contact allergen primin.6 A common clinical presentation of ACD for any of these culprit flowers is localized dermatitis of the thumb and index finger in a florist or gardener.
Plants That Cause Irritant Reactions
Cactuses
Although the long spines of the Cactaceae family of cactuses is a warning for passersby, it is the small and nearly invisible barbed hairs (glochids) that inflict a more dramatic cutaneous reaction. The prickly pear cactus (Opuntia species) is a good example of such a plant, as its glochids cause mechanical irritation but also can become embedded in the skin and result in subcutaneous granulomas known as sabra dermatitis.26
Stinging Nettle
The dermatologic term urticaria owes its namesake to the stinging nettle plant, which comes from the family Urticaceae. The stinging nettle has small hairs on its leaves, referred to as stinging trichomes, which have needlelike tips that pierce the skin and inject a mix of histamine, formic acid, and acetylcholine, causing a pruritic dermatitis that may last up to 12 hours.27 The plant is found worldwide and is a common weed in North America.
Phytophotodermatitis
Lemons and limes (Rutaceae family) are common culprits of phytophotodermatitis, often causing what is known as a margarita burn after outdoor consumption or preparation of this tasty citrus beverage.28 An accidental spray of lime juice on the skin while adding it to a beer, guacamole, salsa, or any other food or beverage also can cause phytophotodermatitis.29-31 Although the juice of lemons and limes contains psoralens, the rind can contain a 6- to 186-fold increased concentration.32 Psoralen is the photoactive agent in Rutaceae plants that intercalates in double-stranded DNA and promotes intrastrand cross-links when exposed to UVA light, which ultimately leads to dermatitis.9 Phytophotodermatitis commonly causes erythema, edema, and painful bullae on sun-exposed areas and classically heals with hyperpigmentation.
Pseudophytodermatitis can occur in grain farmers and harvesters who handle wheat and/or barley and incidentally come in contact with insects and chemicals on the plant material. Pseudophytodermatitis from mites in the wheat and/or barley plant can occur at harvest time when contact with the plant material is high. Insects such as the North American itch mite (Pediculoides ventricosus) can cause petechiae, wheals, and pustules. In addition, insecticides such as malathion and arsenical sprays that are applied to plant leaves can cause pseudophytodermatitis, which may be initially diagnosed as dermatitis to the plant itself.6
Patch Testing to Plants
When a patient presents with recurrent or persistent dermatitis and a plant contact allergen is suspected, patch testing is indicated. Most comprehensive patch test series contain various plant allergens, such as sesquiterpene lactones, Compositae mix, and limonene hydroperoxides, and patch testing to a specialized plant series may be necessary. Poison ivy/oak/sumac allergens typically are not included in patch test series because of the high prevalence of allergic reactions to these chemicals and the likelihood of sensitization when patch testing with urushiol. Compositae contact sensitization can be difficult to diagnose because neither sesquiterpene lactone mix 0.1% nor parthenolide 0.1% are sensitive enough to pick up all Compositae allergies.33,34 Paulsen and Andersen34 proposed that if Compositae sensitization is suspected, testing should include sesquiterpene lactone, parthenolide, and Compositae mix II 2.5%, as well as other potential Compositae allergens based on the patient’s history.34
Because plants can have geographic variability and contain potentially unknown allergens,35 testing to plant components may increase the diagnostic yield of patch testing. Dividing the plant into component parts (ie, stem, bulb, leaf, flower) is helpful, as different components have different allergen concentrations. It is important to consult expert resources before proceeding with plant component patch testing because irritant reactions are frequent and may confound the testing.36
Prevention and Treatment
For all plant dermatoses, the mainstay of prevention is to avoid contact with the offending plant material. Gloves can be an important protective tool for plant dermatitis prevention; the correct material depends on the plant species being handled. Rubber gloves should not be worn to protect against Toxicodendron plants since the catechols in urushiol are soluble in rubber; vinyl gloves should be worn instead.6 Marks37 found that tuliposide A, the allergen in the Peruvian lily (Alstroemeria), penetrates both vinyl and latex gloves; it does not penetrate nitrile gloves. If exposed, the risk of dermatitis can be decreased if the allergen is washed away with soap and water as soon as possible. Some allergens such as Toxicodendron are absorbed quickly and need to be washed off within 10 minutes of exposure.6 Importantly, exposed gardening gloves may continue to perpetuate ACD if the allergen is not also washed off the gloves themselves.
For light-mediated dermatoses, sun avoidance or use of an effective sunscreen can reduce symptoms in an individual who has already been exposed.10 UVA light activates psoralen-mediated dermatitis but not until 30 to 120 minutes after absorption into the skin.38
Barrier creams are thought to be protective against plant ACD through a variety of mechanisms. The cream itself is meant to reduce skin contact to an allergen or irritant. Additionally, barrier creams contain active ingredients such as silicone, hydrocarbons, and aluminum chlorohydrate, which are thought to trap or transform offending agents before contacting the skin. When contact with a Toxicodendron species is anticipated, Marks et al39 found that dermatitis was absent or significantly reduced when 144 patients were pretreated with quaternium-18 bentonite lotion 5% (P<.0001).
Although allergen avoidance and use of gloves and barrier creams are the mainstays of preventing plant dermatoses, treatment often is required to control postexposure symptoms. For all plant dermatoses, topical corticosteroids can be used to reduce inflammation and pruritus. In some cases, systemic steroids may be necessary. To prevent rebound of dermatitis, patients often require a 3-week or longer course of oral steroids to quell the reaction, particularly if the dermatitis is vigorous or an id reaction is present.40 Antihistamines and cold compresses also can provide symptomatic relief.
Final Interpretation
Plants can cause a variety of dermatoses. Although Toxicodendron plants are the most frequent cause of ACD, it is important to keep in mind that florists, gardeners, and farmers are exposed to a large variety of allergens, irritants, and phototoxic agents that cause dermatoses as well. Confirmation of plant-induced ACD involves patch testing against suspected species. Prevention involves use of appropriate barriers and avoidance of implicated plants. Treatment includes topical steroids, antihistamines, and prednisone.
- Gladman AC. Toxicodendron dermatitis: poison ivy, oak, and sumac. Wilderness Environ Med. 2006;17:120-128.
- Pariser D, Ceilley R, Lefkovits A, et al. Poison ivy, oak and sumac. Derm Insights. 2003;4:26-28.
- Wolff K, Johnson R. Fitzpatrick’s Color Atlas and Synopsis of Clinical Dermatology. 6th ed. McGraw Hill Education; 2009.
- Zomorodi N, Butt M, Maczuga S, et al. Cost and diagnostic characteristics of Toxicodendron dermatitis in the USA: a retrospective cross-sectional analysis. Br J Dermatol. 2020;183:772-773.
- DeKoven JG, Silverberg JI, Warshaw EM, et al. North American Contact Dermatitis Group patch test results: 2017-2018. Dermatitis. 2021;32:111-123.
- Fowler JF, Zirwas MJ. Fisher’s Contact Dermatitis. 7th ed. Contact Dermatitis Institute; 2019.
- Smith HR, Basketter DA, McFadden JP. Irritant dermatitis, irritancy and its role in allergic contact dermatitis. Clin Exp Dermatol. 2002;27:138-146.
- Wakelin SH. Contact urticaria. Clin Exp Dermatol. 2001;26:132-136.
- Ellis CR, Elston DM. Psoralen-induced phytophotodermatitis. Dermatitis. 2021;32:140-143.
- Deleo VA. Photocontact dermatitis. Dermatol Ther. 2004;17:279-288.
- National Institute for Occupational Safety and Health. Poisonous plants. Centers for Disease Control and Prevention website. Updated June 1, 2018. Accessed August 10, 2021. https://www.cdc.gov/niosh/topics/plants/geographic.html
- Schloemer JA, Zirwas MJ, Burkhart CG. Airborne contact dermatitis: common causes in the USA. Int J Dermatol. 2015;54:271-274.
- Guin JD. The black spot test for recognizing poison ivy and related species. J Am Acad Dermatol. 1980;2:332-333.
- Mitchell J, Dupuis G. Allergic contact dermatitis from sesquiterpenoids of the Compositae family of plants. Br J Dermatol. 1971;84:139-150.
- Paulsen E, Andersen KE. Lettuce contact allergy. Contact Dermatitis. 2016;74:67-75.
- Samaran Q, Clark E, Dereure O, et al. Airborne allergic contact dermatitis caused by artichoke. Contact Dermatitis. 2020;82:395-397.
- Du H, Ross JS, Norris PG, et al. Contact and photocontact sensitization in chronic actinic dermatitis: sesquiterpene lactone mix is an important allergen. Br J Dermatol. 1995;132:543-547.
- Wrangsjo K, Marie Ros A, Walhberg JE. Contact allergy to Compositae plants in patients with summer-exacerbated dermatitis. Contact Dermatitis. 1990;22:148-154.
- Staser K, Ezra N, Sheehan MP, et al. The beak sign: a clinical clue to airborne contact dermatitis. Dermatitis. 2014;25:97-98.
- Davies M, Kersey J. Contact allergy to yarrow and dandelion. Contact Dermatitis. 1986;14:256-257.
- Anzai A, Vázquez Herrera NE, Tosti A. Airborne allergic contact dermatitis caused by chamomile tea. Contact Dermatitis. 2015;72:254-255.
- Paulsen E. Systemic allergic dermatitis caused by sesquiterpene lactones. Contact Dermatitis. 2017;76:1-10.
- Thiboutot DM, Hamory BH, Marks JG. Dermatoses among floral shop workers. J Am Acad Dermatol. 1990;22:54-58.
- Hjorth N, Wilkinson DS. Contact dermatitis IV. tulip fingers, hyacinth itch and lily rash. Br J Dermatol. 1968;80:696-698.
- Guin JD, Franks H. Fingertip dermatitis in a retail florist. Cutis. 2001;67:328-330.
- Magro C, Lipner S. Sabra dermatitis: combined features of delayed hypersensitivity and foreign body reaction to implanted glochidia. Dermatol Online J. 2020;26:13030/qt2157f9g0.
- Cummings AJ, Olsen M. Mechanism of action of stinging nettles. Wilderness Environ Med. 2011;22:136-139.
- Maniam G, Light KML, Wilson J. Margarita burn: recognition and treatment of phytophotodermatitis. J Am Board Fam Med. 2021;34:398-401.
- Flugman SL. Mexican beer dermatitis: a unique variant of lime phytophotodermatitis attributable to contemporary beer-drinking practices. Arch Dermatol. 2010;146:1194-1195.
- Kung AC, Stephens MB, Darling T. Phytophotodermatitis: bulla formation and hyperpigmentation during spring break. Mil Med. 2009;174:657-661.
- Smith LG. Phytophotodermatitis. Images Emerg Med. 2017;1:146-147.
- Wagner AM, Wu JJ, Hansen RC, et al. Bullous phytophotodermatitis associated with high natural concentrations of furanocoumarins in limes. Am J Contact Dermat. 2002;13:10-14.
- Green C, Ferguson J. Sesquiterpene lactone mix is not an adequate screen for Compositae allergy. Contact Dermatitis. 1994;31:151-153.
- Paulsen E, Andersen KE. Screening for Compositae contact sensitization with sesquiterpene lactones and Compositae mix 2.5% pet. Contact Dermatitis. 2019;81:368-373.
- Paulsen E, Andersen KE. Patch testing with constituents of Compositae mixes. Contact Dermatitis. 2012;66:241-246.
- Frosch PJ, Geier J, Uter W, et al. Patch testing with the patients’ own products. Contact Dermatitis. 2011:929-941.
- Marks JG. Allergic contact dermatitis to Alstroemeria. Arch Dermatol. 1988;124:914-916.
- Moreau JF, English JC, Gehris RP. Phytophotodermatitis. J Pediatr Adolesc Gynecol. 2014;27:93-94.
- Marks JG, Fowler JF, Sherertz EF, et al. Prevention of poison ivy and poison oak allergic contact dermatitis by quaternium-18 bentonite. J Am Acad Dermatol. 1995;33:212-216.
- Craig K, Meadows SE. What is the best duration of steroid therapy for contact dermatitis (rhus)? J Fam Pract. 2006;55:166-167.
Plants can contribute to a variety of dermatoses. The Toxicodendron genus, which includes poison ivy, poison oak, and poison sumac, is a well-known and common cause of allergic contact dermatitis (ACD), but many other plants can cause direct or airborne contact dermatitis, especially in gardeners, florists, and farmers. This article provides an overview of different plant-related dermatoses and culprit plants as well as how these dermatoses should be diagnosed and treated.
Epidemiology
Plant dermatoses affect more than 50 million individuals each year.1,2 In the United States, the Toxicodendron genus causes ACD in more than 70% of exposed individuals, leading to medical visits.3 An urgent care visit for a plant-related dermatitis is estimated to cost $168, while an emergency department visit can cost 3 times as much.4 Although less common, Compositae plants are another important culprit of plant dermatitis, particularly in gardeners, florists, and farmers. Data from the 2017-2018 North American Contact Dermatitis Group screening series (N=4947) showed sesquiterpene lactones and Compositae to be positive in 0.5% of patch-tested patients.5
Plant Dermatitis Classifications
Plant dermatitis can be classified into 5 main categories: ACD, mechanical irritant contact dermatitis, chemical irritant contact dermatitis, light-mediated dermatitis, and pseudophytodermatitis.6
Allergic contact dermatitis is an immune-mediated type IV delayed hypersensitivity reaction. The common molecular allergens in plants include phenols, α-methylene-γ-butyrolactones, quinones, terpenes, disulfides, isothiocyanates, and polyacetylenic derivatives.6
Plant contact dermatitis due to mechanical and chemical irritants is precipitated by multiple mechanisms, including disruption of the epidermal barrier and subsequent cytokine release from keratinocytes.7 Nonimmunologic contact urticaria from plants is thought to be a type of irritant reaction precipitated by mechanical or chemical trauma.8
Light-mediated dermatitis includes phytophotodermatitis and photoallergic contact dermatitis. Phytophotodermatitis is a phototoxic reaction triggered by exposure to both plant-derived furanocoumarin and UVA light.9 By contrast, photoallergic contact dermatitis is a delayed hypersensitivity reaction from prior sensitization to a light-activated antigen.10
Pseudophytodermatitis, as its name implies, is not truly mediated by an allergen or irritant intrinsic to the plant but rather by dyes, waxes, insecticides, or arthropods that inhabit the plant or are secondarily applied.6
Common Plant Allergens
Anacardiaceae Family
Most of the allergenic plants within the Anacardiaceae family belong to the Toxicodendron genus, which encompasses poison ivy (Toxicodendron radicans), poison oak (Toxicodendron pubescens,Toxicodendron quercifolium, Toxicodendron diversiloum), and poison sumac (Toxicodendron vernix). Poison ivy is the celebrity of the Anacardiaceae family and contributes to most cases of plant-related ACD. It is found in every state in the continental United States. Poison oak is another common culprit found in the western and southeastern United States.11 Plants within the Anacardiaceae family contain an oleoresin called urushiol, which is the primary sensitizing substance. Although poison ivy and poison oak grow well in full sun to partial shade, poison sumac typically is found in damp swampy areas east of the Rocky Mountains. Most cases of ACD related to Anacardiaceae species are due to direct contact with urushiol from a Toxicodendron plant, but burning of brush containing Toxicodendron can cause airborne exposure when urushiol oil is carried by smoke particles.12 Sensitization to Toxicodendron can cause ACD to other Anacardiaceae species such as the Japanese lacquer tree (Toxicodendron vernicifluum), mango tree (Mangifera indica), cashew tree (Anacardium occidentale), and Indian marking nut tree (Semecarpus anacardium).6 Cross-reactions to components of the ginkgo tree (Ginkgo biloba) also are possible.
Toxicodendron plants can be more easily identified and avoided with knowledge of their characteristic leaf patterns. The most dependable way to identify poison ivy and poison oak species is to look for plants with 3 leaves, giving rise to the common saying, “Leaves of three, leave them be.” Poison sumac plants have groups of 7 to 13 leaves arranged as pairs along a central rib. Another helpful finding is a black deposit that Toxicodendron species leave behind following trauma to the leaves. Urushiol oxidizes when exposed to air and turns into a black deposit that can be seen on damaged leaves themselves or can be demonstrated in a black spot test to verify if a plant is a Toxicodendron species. The test is performed by gathering (carefully, without direct contact) a few leaves in a paper towel and crushing them to release sap. Within minutes, the sap will turn black if the plant is indeed a Toxicodendron species.13Pruritic, edematous, erythematous papules, plaques, and eventual vesicles in a linear distribution are suspicious for Toxicodendron exposure. Although your pet will not develop Toxicodendron ACD, oleoresin-contaminated pets can transfer the oils to their owners after coming into contact with these plants. Toxicodendron dermatitis also can be acquired from oleoresin-contaminated fomites such as clothing and shoes worn in the garden or when hiking. Toxicodendron dermatitis can appear at different sites on the body at different times depending on the amount of oleoresin exposure as well as epidermal thickness. For example, the oleoresin can be transferred from the hands to body areas with a thinner stratum corneum (eg, genitalia) and cause subsequent dermatitis.1
Compositae Family
The Compositae family (also known as Asteraceae) is a large plant family with more than 20,000 species, including numerous weeds, wildflowers, and vegetables. The flowers, leaves, stems, and pollens of the Compositae family are coated by cyclic esters called sesquiterpene lactones. Mitchell and Dupuis14 showed that sesquiterpene lactones are the allergens responsible for ACD to various Compositae plants, including ragweed (Ambrosia), sneezeweed (Helenium), and chrysanthemums (Chrysanthemum). Common Compositae vegetables such as lettuce (Lactuca sativa) have been reported to cause ACD in chefs, grocery store produce handlers, gardeners, and even owners of lettuce-eating pet guinea pigs and turtles.15 Similarly, artichokes (Cynara scolymus) can cause ACD in gardeners.16 Exposure to Compositae species also has been implicated in photoallergic reactions, and studies have demonstrated that some patients with chronic actinic dermatitis also have positive patch test reactions to Compositae species and/or sesquiterpene lactones.17,18
In addition to direct contact with Compositae plants, airborne exposure to sesquiterpene lactones can cause ACD.14 The pattern of airborne contact dermatitis typically involves exposed areas such as the eyelids, central face, and/or neck. The beak sign also can be a clue to airborne contact dermatitis, which involves dermatitis of the face that spares the nasal tip and/or nasal ridge. It is thought that the beak sign may result from increased sebaceous gland concentration on the nose, which prevents penetration of allergens and irritants.19 Unlike photoallergic contact dermatitis, which also can involve the face, airborne ACD frequently involves photoprotected areas such as the submandibular chin and the upper lip. Davies and Kersey20 reported the case of a groundsman who was cutting grass with dandelions (Taraxacum officinale) and was found to have associated airborne ACD of the face, neck, and forearms due to Compositae allergy. In a different setting, the aromas of chamomile (Matricaria chamomilla) have been reported to cause airborne ACD in a tea drinker.21 Paulsen22 found that ingestion of chamomile tea can induce systemic ACD in sensitized individuals.
Alstroemeriaceae, Liliaceae, and Primulaceae
Florists are exposed to many plant species and have a high prevalence of ACD. Thiboutot et al23 found that 15 of 57 (26%) floral workers experienced hand dermatitis that cleared with time away from work. The Peruvian lily (Alstroemeria, Alstroemeriaceae family), which contains tuliposide A, was found to be the leading cause of sensitization.23 Tulips (Tulipa, Liliaceae family), as the flower name suggests, also contain tuliposide A, which along with mechanical irritation from the course tecta fibers on the bulbs lead to a dermatitis known as tulip fingers.24,25 Poison primrose (Primula obconica, Primulaceae family), cultivated for its highly colorful flowers, contains the contact allergen primin.6 A common clinical presentation of ACD for any of these culprit flowers is localized dermatitis of the thumb and index finger in a florist or gardener.
Plants That Cause Irritant Reactions
Cactuses
Although the long spines of the Cactaceae family of cactuses is a warning for passersby, it is the small and nearly invisible barbed hairs (glochids) that inflict a more dramatic cutaneous reaction. The prickly pear cactus (Opuntia species) is a good example of such a plant, as its glochids cause mechanical irritation but also can become embedded in the skin and result in subcutaneous granulomas known as sabra dermatitis.26
Stinging Nettle
The dermatologic term urticaria owes its namesake to the stinging nettle plant, which comes from the family Urticaceae. The stinging nettle has small hairs on its leaves, referred to as stinging trichomes, which have needlelike tips that pierce the skin and inject a mix of histamine, formic acid, and acetylcholine, causing a pruritic dermatitis that may last up to 12 hours.27 The plant is found worldwide and is a common weed in North America.
Phytophotodermatitis
Lemons and limes (Rutaceae family) are common culprits of phytophotodermatitis, often causing what is known as a margarita burn after outdoor consumption or preparation of this tasty citrus beverage.28 An accidental spray of lime juice on the skin while adding it to a beer, guacamole, salsa, or any other food or beverage also can cause phytophotodermatitis.29-31 Although the juice of lemons and limes contains psoralens, the rind can contain a 6- to 186-fold increased concentration.32 Psoralen is the photoactive agent in Rutaceae plants that intercalates in double-stranded DNA and promotes intrastrand cross-links when exposed to UVA light, which ultimately leads to dermatitis.9 Phytophotodermatitis commonly causes erythema, edema, and painful bullae on sun-exposed areas and classically heals with hyperpigmentation.
Pseudophytodermatitis can occur in grain farmers and harvesters who handle wheat and/or barley and incidentally come in contact with insects and chemicals on the plant material. Pseudophytodermatitis from mites in the wheat and/or barley plant can occur at harvest time when contact with the plant material is high. Insects such as the North American itch mite (Pediculoides ventricosus) can cause petechiae, wheals, and pustules. In addition, insecticides such as malathion and arsenical sprays that are applied to plant leaves can cause pseudophytodermatitis, which may be initially diagnosed as dermatitis to the plant itself.6
Patch Testing to Plants
When a patient presents with recurrent or persistent dermatitis and a plant contact allergen is suspected, patch testing is indicated. Most comprehensive patch test series contain various plant allergens, such as sesquiterpene lactones, Compositae mix, and limonene hydroperoxides, and patch testing to a specialized plant series may be necessary. Poison ivy/oak/sumac allergens typically are not included in patch test series because of the high prevalence of allergic reactions to these chemicals and the likelihood of sensitization when patch testing with urushiol. Compositae contact sensitization can be difficult to diagnose because neither sesquiterpene lactone mix 0.1% nor parthenolide 0.1% are sensitive enough to pick up all Compositae allergies.33,34 Paulsen and Andersen34 proposed that if Compositae sensitization is suspected, testing should include sesquiterpene lactone, parthenolide, and Compositae mix II 2.5%, as well as other potential Compositae allergens based on the patient’s history.34
Because plants can have geographic variability and contain potentially unknown allergens,35 testing to plant components may increase the diagnostic yield of patch testing. Dividing the plant into component parts (ie, stem, bulb, leaf, flower) is helpful, as different components have different allergen concentrations. It is important to consult expert resources before proceeding with plant component patch testing because irritant reactions are frequent and may confound the testing.36
Prevention and Treatment
For all plant dermatoses, the mainstay of prevention is to avoid contact with the offending plant material. Gloves can be an important protective tool for plant dermatitis prevention; the correct material depends on the plant species being handled. Rubber gloves should not be worn to protect against Toxicodendron plants since the catechols in urushiol are soluble in rubber; vinyl gloves should be worn instead.6 Marks37 found that tuliposide A, the allergen in the Peruvian lily (Alstroemeria), penetrates both vinyl and latex gloves; it does not penetrate nitrile gloves. If exposed, the risk of dermatitis can be decreased if the allergen is washed away with soap and water as soon as possible. Some allergens such as Toxicodendron are absorbed quickly and need to be washed off within 10 minutes of exposure.6 Importantly, exposed gardening gloves may continue to perpetuate ACD if the allergen is not also washed off the gloves themselves.
For light-mediated dermatoses, sun avoidance or use of an effective sunscreen can reduce symptoms in an individual who has already been exposed.10 UVA light activates psoralen-mediated dermatitis but not until 30 to 120 minutes after absorption into the skin.38
Barrier creams are thought to be protective against plant ACD through a variety of mechanisms. The cream itself is meant to reduce skin contact to an allergen or irritant. Additionally, barrier creams contain active ingredients such as silicone, hydrocarbons, and aluminum chlorohydrate, which are thought to trap or transform offending agents before contacting the skin. When contact with a Toxicodendron species is anticipated, Marks et al39 found that dermatitis was absent or significantly reduced when 144 patients were pretreated with quaternium-18 bentonite lotion 5% (P<.0001).
Although allergen avoidance and use of gloves and barrier creams are the mainstays of preventing plant dermatoses, treatment often is required to control postexposure symptoms. For all plant dermatoses, topical corticosteroids can be used to reduce inflammation and pruritus. In some cases, systemic steroids may be necessary. To prevent rebound of dermatitis, patients often require a 3-week or longer course of oral steroids to quell the reaction, particularly if the dermatitis is vigorous or an id reaction is present.40 Antihistamines and cold compresses also can provide symptomatic relief.
Final Interpretation
Plants can cause a variety of dermatoses. Although Toxicodendron plants are the most frequent cause of ACD, it is important to keep in mind that florists, gardeners, and farmers are exposed to a large variety of allergens, irritants, and phototoxic agents that cause dermatoses as well. Confirmation of plant-induced ACD involves patch testing against suspected species. Prevention involves use of appropriate barriers and avoidance of implicated plants. Treatment includes topical steroids, antihistamines, and prednisone.
Plants can contribute to a variety of dermatoses. The Toxicodendron genus, which includes poison ivy, poison oak, and poison sumac, is a well-known and common cause of allergic contact dermatitis (ACD), but many other plants can cause direct or airborne contact dermatitis, especially in gardeners, florists, and farmers. This article provides an overview of different plant-related dermatoses and culprit plants as well as how these dermatoses should be diagnosed and treated.
Epidemiology
Plant dermatoses affect more than 50 million individuals each year.1,2 In the United States, the Toxicodendron genus causes ACD in more than 70% of exposed individuals, leading to medical visits.3 An urgent care visit for a plant-related dermatitis is estimated to cost $168, while an emergency department visit can cost 3 times as much.4 Although less common, Compositae plants are another important culprit of plant dermatitis, particularly in gardeners, florists, and farmers. Data from the 2017-2018 North American Contact Dermatitis Group screening series (N=4947) showed sesquiterpene lactones and Compositae to be positive in 0.5% of patch-tested patients.5
Plant Dermatitis Classifications
Plant dermatitis can be classified into 5 main categories: ACD, mechanical irritant contact dermatitis, chemical irritant contact dermatitis, light-mediated dermatitis, and pseudophytodermatitis.6
Allergic contact dermatitis is an immune-mediated type IV delayed hypersensitivity reaction. The common molecular allergens in plants include phenols, α-methylene-γ-butyrolactones, quinones, terpenes, disulfides, isothiocyanates, and polyacetylenic derivatives.6
Plant contact dermatitis due to mechanical and chemical irritants is precipitated by multiple mechanisms, including disruption of the epidermal barrier and subsequent cytokine release from keratinocytes.7 Nonimmunologic contact urticaria from plants is thought to be a type of irritant reaction precipitated by mechanical or chemical trauma.8
Light-mediated dermatitis includes phytophotodermatitis and photoallergic contact dermatitis. Phytophotodermatitis is a phototoxic reaction triggered by exposure to both plant-derived furanocoumarin and UVA light.9 By contrast, photoallergic contact dermatitis is a delayed hypersensitivity reaction from prior sensitization to a light-activated antigen.10
Pseudophytodermatitis, as its name implies, is not truly mediated by an allergen or irritant intrinsic to the plant but rather by dyes, waxes, insecticides, or arthropods that inhabit the plant or are secondarily applied.6
Common Plant Allergens
Anacardiaceae Family
Most of the allergenic plants within the Anacardiaceae family belong to the Toxicodendron genus, which encompasses poison ivy (Toxicodendron radicans), poison oak (Toxicodendron pubescens,Toxicodendron quercifolium, Toxicodendron diversiloum), and poison sumac (Toxicodendron vernix). Poison ivy is the celebrity of the Anacardiaceae family and contributes to most cases of plant-related ACD. It is found in every state in the continental United States. Poison oak is another common culprit found in the western and southeastern United States.11 Plants within the Anacardiaceae family contain an oleoresin called urushiol, which is the primary sensitizing substance. Although poison ivy and poison oak grow well in full sun to partial shade, poison sumac typically is found in damp swampy areas east of the Rocky Mountains. Most cases of ACD related to Anacardiaceae species are due to direct contact with urushiol from a Toxicodendron plant, but burning of brush containing Toxicodendron can cause airborne exposure when urushiol oil is carried by smoke particles.12 Sensitization to Toxicodendron can cause ACD to other Anacardiaceae species such as the Japanese lacquer tree (Toxicodendron vernicifluum), mango tree (Mangifera indica), cashew tree (Anacardium occidentale), and Indian marking nut tree (Semecarpus anacardium).6 Cross-reactions to components of the ginkgo tree (Ginkgo biloba) also are possible.
Toxicodendron plants can be more easily identified and avoided with knowledge of their characteristic leaf patterns. The most dependable way to identify poison ivy and poison oak species is to look for plants with 3 leaves, giving rise to the common saying, “Leaves of three, leave them be.” Poison sumac plants have groups of 7 to 13 leaves arranged as pairs along a central rib. Another helpful finding is a black deposit that Toxicodendron species leave behind following trauma to the leaves. Urushiol oxidizes when exposed to air and turns into a black deposit that can be seen on damaged leaves themselves or can be demonstrated in a black spot test to verify if a plant is a Toxicodendron species. The test is performed by gathering (carefully, without direct contact) a few leaves in a paper towel and crushing them to release sap. Within minutes, the sap will turn black if the plant is indeed a Toxicodendron species.13Pruritic, edematous, erythematous papules, plaques, and eventual vesicles in a linear distribution are suspicious for Toxicodendron exposure. Although your pet will not develop Toxicodendron ACD, oleoresin-contaminated pets can transfer the oils to their owners after coming into contact with these plants. Toxicodendron dermatitis also can be acquired from oleoresin-contaminated fomites such as clothing and shoes worn in the garden or when hiking. Toxicodendron dermatitis can appear at different sites on the body at different times depending on the amount of oleoresin exposure as well as epidermal thickness. For example, the oleoresin can be transferred from the hands to body areas with a thinner stratum corneum (eg, genitalia) and cause subsequent dermatitis.1
Compositae Family
The Compositae family (also known as Asteraceae) is a large plant family with more than 20,000 species, including numerous weeds, wildflowers, and vegetables. The flowers, leaves, stems, and pollens of the Compositae family are coated by cyclic esters called sesquiterpene lactones. Mitchell and Dupuis14 showed that sesquiterpene lactones are the allergens responsible for ACD to various Compositae plants, including ragweed (Ambrosia), sneezeweed (Helenium), and chrysanthemums (Chrysanthemum). Common Compositae vegetables such as lettuce (Lactuca sativa) have been reported to cause ACD in chefs, grocery store produce handlers, gardeners, and even owners of lettuce-eating pet guinea pigs and turtles.15 Similarly, artichokes (Cynara scolymus) can cause ACD in gardeners.16 Exposure to Compositae species also has been implicated in photoallergic reactions, and studies have demonstrated that some patients with chronic actinic dermatitis also have positive patch test reactions to Compositae species and/or sesquiterpene lactones.17,18
In addition to direct contact with Compositae plants, airborne exposure to sesquiterpene lactones can cause ACD.14 The pattern of airborne contact dermatitis typically involves exposed areas such as the eyelids, central face, and/or neck. The beak sign also can be a clue to airborne contact dermatitis, which involves dermatitis of the face that spares the nasal tip and/or nasal ridge. It is thought that the beak sign may result from increased sebaceous gland concentration on the nose, which prevents penetration of allergens and irritants.19 Unlike photoallergic contact dermatitis, which also can involve the face, airborne ACD frequently involves photoprotected areas such as the submandibular chin and the upper lip. Davies and Kersey20 reported the case of a groundsman who was cutting grass with dandelions (Taraxacum officinale) and was found to have associated airborne ACD of the face, neck, and forearms due to Compositae allergy. In a different setting, the aromas of chamomile (Matricaria chamomilla) have been reported to cause airborne ACD in a tea drinker.21 Paulsen22 found that ingestion of chamomile tea can induce systemic ACD in sensitized individuals.
Alstroemeriaceae, Liliaceae, and Primulaceae
Florists are exposed to many plant species and have a high prevalence of ACD. Thiboutot et al23 found that 15 of 57 (26%) floral workers experienced hand dermatitis that cleared with time away from work. The Peruvian lily (Alstroemeria, Alstroemeriaceae family), which contains tuliposide A, was found to be the leading cause of sensitization.23 Tulips (Tulipa, Liliaceae family), as the flower name suggests, also contain tuliposide A, which along with mechanical irritation from the course tecta fibers on the bulbs lead to a dermatitis known as tulip fingers.24,25 Poison primrose (Primula obconica, Primulaceae family), cultivated for its highly colorful flowers, contains the contact allergen primin.6 A common clinical presentation of ACD for any of these culprit flowers is localized dermatitis of the thumb and index finger in a florist or gardener.
Plants That Cause Irritant Reactions
Cactuses
Although the long spines of the Cactaceae family of cactuses is a warning for passersby, it is the small and nearly invisible barbed hairs (glochids) that inflict a more dramatic cutaneous reaction. The prickly pear cactus (Opuntia species) is a good example of such a plant, as its glochids cause mechanical irritation but also can become embedded in the skin and result in subcutaneous granulomas known as sabra dermatitis.26
Stinging Nettle
The dermatologic term urticaria owes its namesake to the stinging nettle plant, which comes from the family Urticaceae. The stinging nettle has small hairs on its leaves, referred to as stinging trichomes, which have needlelike tips that pierce the skin and inject a mix of histamine, formic acid, and acetylcholine, causing a pruritic dermatitis that may last up to 12 hours.27 The plant is found worldwide and is a common weed in North America.
Phytophotodermatitis
Lemons and limes (Rutaceae family) are common culprits of phytophotodermatitis, often causing what is known as a margarita burn after outdoor consumption or preparation of this tasty citrus beverage.28 An accidental spray of lime juice on the skin while adding it to a beer, guacamole, salsa, or any other food or beverage also can cause phytophotodermatitis.29-31 Although the juice of lemons and limes contains psoralens, the rind can contain a 6- to 186-fold increased concentration.32 Psoralen is the photoactive agent in Rutaceae plants that intercalates in double-stranded DNA and promotes intrastrand cross-links when exposed to UVA light, which ultimately leads to dermatitis.9 Phytophotodermatitis commonly causes erythema, edema, and painful bullae on sun-exposed areas and classically heals with hyperpigmentation.
Pseudophytodermatitis can occur in grain farmers and harvesters who handle wheat and/or barley and incidentally come in contact with insects and chemicals on the plant material. Pseudophytodermatitis from mites in the wheat and/or barley plant can occur at harvest time when contact with the plant material is high. Insects such as the North American itch mite (Pediculoides ventricosus) can cause petechiae, wheals, and pustules. In addition, insecticides such as malathion and arsenical sprays that are applied to plant leaves can cause pseudophytodermatitis, which may be initially diagnosed as dermatitis to the plant itself.6
Patch Testing to Plants
When a patient presents with recurrent or persistent dermatitis and a plant contact allergen is suspected, patch testing is indicated. Most comprehensive patch test series contain various plant allergens, such as sesquiterpene lactones, Compositae mix, and limonene hydroperoxides, and patch testing to a specialized plant series may be necessary. Poison ivy/oak/sumac allergens typically are not included in patch test series because of the high prevalence of allergic reactions to these chemicals and the likelihood of sensitization when patch testing with urushiol. Compositae contact sensitization can be difficult to diagnose because neither sesquiterpene lactone mix 0.1% nor parthenolide 0.1% are sensitive enough to pick up all Compositae allergies.33,34 Paulsen and Andersen34 proposed that if Compositae sensitization is suspected, testing should include sesquiterpene lactone, parthenolide, and Compositae mix II 2.5%, as well as other potential Compositae allergens based on the patient’s history.34
Because plants can have geographic variability and contain potentially unknown allergens,35 testing to plant components may increase the diagnostic yield of patch testing. Dividing the plant into component parts (ie, stem, bulb, leaf, flower) is helpful, as different components have different allergen concentrations. It is important to consult expert resources before proceeding with plant component patch testing because irritant reactions are frequent and may confound the testing.36
Prevention and Treatment
For all plant dermatoses, the mainstay of prevention is to avoid contact with the offending plant material. Gloves can be an important protective tool for plant dermatitis prevention; the correct material depends on the plant species being handled. Rubber gloves should not be worn to protect against Toxicodendron plants since the catechols in urushiol are soluble in rubber; vinyl gloves should be worn instead.6 Marks37 found that tuliposide A, the allergen in the Peruvian lily (Alstroemeria), penetrates both vinyl and latex gloves; it does not penetrate nitrile gloves. If exposed, the risk of dermatitis can be decreased if the allergen is washed away with soap and water as soon as possible. Some allergens such as Toxicodendron are absorbed quickly and need to be washed off within 10 minutes of exposure.6 Importantly, exposed gardening gloves may continue to perpetuate ACD if the allergen is not also washed off the gloves themselves.
For light-mediated dermatoses, sun avoidance or use of an effective sunscreen can reduce symptoms in an individual who has already been exposed.10 UVA light activates psoralen-mediated dermatitis but not until 30 to 120 minutes after absorption into the skin.38
Barrier creams are thought to be protective against plant ACD through a variety of mechanisms. The cream itself is meant to reduce skin contact to an allergen or irritant. Additionally, barrier creams contain active ingredients such as silicone, hydrocarbons, and aluminum chlorohydrate, which are thought to trap or transform offending agents before contacting the skin. When contact with a Toxicodendron species is anticipated, Marks et al39 found that dermatitis was absent or significantly reduced when 144 patients were pretreated with quaternium-18 bentonite lotion 5% (P<.0001).
Although allergen avoidance and use of gloves and barrier creams are the mainstays of preventing plant dermatoses, treatment often is required to control postexposure symptoms. For all plant dermatoses, topical corticosteroids can be used to reduce inflammation and pruritus. In some cases, systemic steroids may be necessary. To prevent rebound of dermatitis, patients often require a 3-week or longer course of oral steroids to quell the reaction, particularly if the dermatitis is vigorous or an id reaction is present.40 Antihistamines and cold compresses also can provide symptomatic relief.
Final Interpretation
Plants can cause a variety of dermatoses. Although Toxicodendron plants are the most frequent cause of ACD, it is important to keep in mind that florists, gardeners, and farmers are exposed to a large variety of allergens, irritants, and phototoxic agents that cause dermatoses as well. Confirmation of plant-induced ACD involves patch testing against suspected species. Prevention involves use of appropriate barriers and avoidance of implicated plants. Treatment includes topical steroids, antihistamines, and prednisone.
- Gladman AC. Toxicodendron dermatitis: poison ivy, oak, and sumac. Wilderness Environ Med. 2006;17:120-128.
- Pariser D, Ceilley R, Lefkovits A, et al. Poison ivy, oak and sumac. Derm Insights. 2003;4:26-28.
- Wolff K, Johnson R. Fitzpatrick’s Color Atlas and Synopsis of Clinical Dermatology. 6th ed. McGraw Hill Education; 2009.
- Zomorodi N, Butt M, Maczuga S, et al. Cost and diagnostic characteristics of Toxicodendron dermatitis in the USA: a retrospective cross-sectional analysis. Br J Dermatol. 2020;183:772-773.
- DeKoven JG, Silverberg JI, Warshaw EM, et al. North American Contact Dermatitis Group patch test results: 2017-2018. Dermatitis. 2021;32:111-123.
- Fowler JF, Zirwas MJ. Fisher’s Contact Dermatitis. 7th ed. Contact Dermatitis Institute; 2019.
- Smith HR, Basketter DA, McFadden JP. Irritant dermatitis, irritancy and its role in allergic contact dermatitis. Clin Exp Dermatol. 2002;27:138-146.
- Wakelin SH. Contact urticaria. Clin Exp Dermatol. 2001;26:132-136.
- Ellis CR, Elston DM. Psoralen-induced phytophotodermatitis. Dermatitis. 2021;32:140-143.
- Deleo VA. Photocontact dermatitis. Dermatol Ther. 2004;17:279-288.
- National Institute for Occupational Safety and Health. Poisonous plants. Centers for Disease Control and Prevention website. Updated June 1, 2018. Accessed August 10, 2021. https://www.cdc.gov/niosh/topics/plants/geographic.html
- Schloemer JA, Zirwas MJ, Burkhart CG. Airborne contact dermatitis: common causes in the USA. Int J Dermatol. 2015;54:271-274.
- Guin JD. The black spot test for recognizing poison ivy and related species. J Am Acad Dermatol. 1980;2:332-333.
- Mitchell J, Dupuis G. Allergic contact dermatitis from sesquiterpenoids of the Compositae family of plants. Br J Dermatol. 1971;84:139-150.
- Paulsen E, Andersen KE. Lettuce contact allergy. Contact Dermatitis. 2016;74:67-75.
- Samaran Q, Clark E, Dereure O, et al. Airborne allergic contact dermatitis caused by artichoke. Contact Dermatitis. 2020;82:395-397.
- Du H, Ross JS, Norris PG, et al. Contact and photocontact sensitization in chronic actinic dermatitis: sesquiterpene lactone mix is an important allergen. Br J Dermatol. 1995;132:543-547.
- Wrangsjo K, Marie Ros A, Walhberg JE. Contact allergy to Compositae plants in patients with summer-exacerbated dermatitis. Contact Dermatitis. 1990;22:148-154.
- Staser K, Ezra N, Sheehan MP, et al. The beak sign: a clinical clue to airborne contact dermatitis. Dermatitis. 2014;25:97-98.
- Davies M, Kersey J. Contact allergy to yarrow and dandelion. Contact Dermatitis. 1986;14:256-257.
- Anzai A, Vázquez Herrera NE, Tosti A. Airborne allergic contact dermatitis caused by chamomile tea. Contact Dermatitis. 2015;72:254-255.
- Paulsen E. Systemic allergic dermatitis caused by sesquiterpene lactones. Contact Dermatitis. 2017;76:1-10.
- Thiboutot DM, Hamory BH, Marks JG. Dermatoses among floral shop workers. J Am Acad Dermatol. 1990;22:54-58.
- Hjorth N, Wilkinson DS. Contact dermatitis IV. tulip fingers, hyacinth itch and lily rash. Br J Dermatol. 1968;80:696-698.
- Guin JD, Franks H. Fingertip dermatitis in a retail florist. Cutis. 2001;67:328-330.
- Magro C, Lipner S. Sabra dermatitis: combined features of delayed hypersensitivity and foreign body reaction to implanted glochidia. Dermatol Online J. 2020;26:13030/qt2157f9g0.
- Cummings AJ, Olsen M. Mechanism of action of stinging nettles. Wilderness Environ Med. 2011;22:136-139.
- Maniam G, Light KML, Wilson J. Margarita burn: recognition and treatment of phytophotodermatitis. J Am Board Fam Med. 2021;34:398-401.
- Flugman SL. Mexican beer dermatitis: a unique variant of lime phytophotodermatitis attributable to contemporary beer-drinking practices. Arch Dermatol. 2010;146:1194-1195.
- Kung AC, Stephens MB, Darling T. Phytophotodermatitis: bulla formation and hyperpigmentation during spring break. Mil Med. 2009;174:657-661.
- Smith LG. Phytophotodermatitis. Images Emerg Med. 2017;1:146-147.
- Wagner AM, Wu JJ, Hansen RC, et al. Bullous phytophotodermatitis associated with high natural concentrations of furanocoumarins in limes. Am J Contact Dermat. 2002;13:10-14.
- Green C, Ferguson J. Sesquiterpene lactone mix is not an adequate screen for Compositae allergy. Contact Dermatitis. 1994;31:151-153.
- Paulsen E, Andersen KE. Screening for Compositae contact sensitization with sesquiterpene lactones and Compositae mix 2.5% pet. Contact Dermatitis. 2019;81:368-373.
- Paulsen E, Andersen KE. Patch testing with constituents of Compositae mixes. Contact Dermatitis. 2012;66:241-246.
- Frosch PJ, Geier J, Uter W, et al. Patch testing with the patients’ own products. Contact Dermatitis. 2011:929-941.
- Marks JG. Allergic contact dermatitis to Alstroemeria. Arch Dermatol. 1988;124:914-916.
- Moreau JF, English JC, Gehris RP. Phytophotodermatitis. J Pediatr Adolesc Gynecol. 2014;27:93-94.
- Marks JG, Fowler JF, Sherertz EF, et al. Prevention of poison ivy and poison oak allergic contact dermatitis by quaternium-18 bentonite. J Am Acad Dermatol. 1995;33:212-216.
- Craig K, Meadows SE. What is the best duration of steroid therapy for contact dermatitis (rhus)? J Fam Pract. 2006;55:166-167.
- Gladman AC. Toxicodendron dermatitis: poison ivy, oak, and sumac. Wilderness Environ Med. 2006;17:120-128.
- Pariser D, Ceilley R, Lefkovits A, et al. Poison ivy, oak and sumac. Derm Insights. 2003;4:26-28.
- Wolff K, Johnson R. Fitzpatrick’s Color Atlas and Synopsis of Clinical Dermatology. 6th ed. McGraw Hill Education; 2009.
- Zomorodi N, Butt M, Maczuga S, et al. Cost and diagnostic characteristics of Toxicodendron dermatitis in the USA: a retrospective cross-sectional analysis. Br J Dermatol. 2020;183:772-773.
- DeKoven JG, Silverberg JI, Warshaw EM, et al. North American Contact Dermatitis Group patch test results: 2017-2018. Dermatitis. 2021;32:111-123.
- Fowler JF, Zirwas MJ. Fisher’s Contact Dermatitis. 7th ed. Contact Dermatitis Institute; 2019.
- Smith HR, Basketter DA, McFadden JP. Irritant dermatitis, irritancy and its role in allergic contact dermatitis. Clin Exp Dermatol. 2002;27:138-146.
- Wakelin SH. Contact urticaria. Clin Exp Dermatol. 2001;26:132-136.
- Ellis CR, Elston DM. Psoralen-induced phytophotodermatitis. Dermatitis. 2021;32:140-143.
- Deleo VA. Photocontact dermatitis. Dermatol Ther. 2004;17:279-288.
- National Institute for Occupational Safety and Health. Poisonous plants. Centers for Disease Control and Prevention website. Updated June 1, 2018. Accessed August 10, 2021. https://www.cdc.gov/niosh/topics/plants/geographic.html
- Schloemer JA, Zirwas MJ, Burkhart CG. Airborne contact dermatitis: common causes in the USA. Int J Dermatol. 2015;54:271-274.
- Guin JD. The black spot test for recognizing poison ivy and related species. J Am Acad Dermatol. 1980;2:332-333.
- Mitchell J, Dupuis G. Allergic contact dermatitis from sesquiterpenoids of the Compositae family of plants. Br J Dermatol. 1971;84:139-150.
- Paulsen E, Andersen KE. Lettuce contact allergy. Contact Dermatitis. 2016;74:67-75.
- Samaran Q, Clark E, Dereure O, et al. Airborne allergic contact dermatitis caused by artichoke. Contact Dermatitis. 2020;82:395-397.
- Du H, Ross JS, Norris PG, et al. Contact and photocontact sensitization in chronic actinic dermatitis: sesquiterpene lactone mix is an important allergen. Br J Dermatol. 1995;132:543-547.
- Wrangsjo K, Marie Ros A, Walhberg JE. Contact allergy to Compositae plants in patients with summer-exacerbated dermatitis. Contact Dermatitis. 1990;22:148-154.
- Staser K, Ezra N, Sheehan MP, et al. The beak sign: a clinical clue to airborne contact dermatitis. Dermatitis. 2014;25:97-98.
- Davies M, Kersey J. Contact allergy to yarrow and dandelion. Contact Dermatitis. 1986;14:256-257.
- Anzai A, Vázquez Herrera NE, Tosti A. Airborne allergic contact dermatitis caused by chamomile tea. Contact Dermatitis. 2015;72:254-255.
- Paulsen E. Systemic allergic dermatitis caused by sesquiterpene lactones. Contact Dermatitis. 2017;76:1-10.
- Thiboutot DM, Hamory BH, Marks JG. Dermatoses among floral shop workers. J Am Acad Dermatol. 1990;22:54-58.
- Hjorth N, Wilkinson DS. Contact dermatitis IV. tulip fingers, hyacinth itch and lily rash. Br J Dermatol. 1968;80:696-698.
- Guin JD, Franks H. Fingertip dermatitis in a retail florist. Cutis. 2001;67:328-330.
- Magro C, Lipner S. Sabra dermatitis: combined features of delayed hypersensitivity and foreign body reaction to implanted glochidia. Dermatol Online J. 2020;26:13030/qt2157f9g0.
- Cummings AJ, Olsen M. Mechanism of action of stinging nettles. Wilderness Environ Med. 2011;22:136-139.
- Maniam G, Light KML, Wilson J. Margarita burn: recognition and treatment of phytophotodermatitis. J Am Board Fam Med. 2021;34:398-401.
- Flugman SL. Mexican beer dermatitis: a unique variant of lime phytophotodermatitis attributable to contemporary beer-drinking practices. Arch Dermatol. 2010;146:1194-1195.
- Kung AC, Stephens MB, Darling T. Phytophotodermatitis: bulla formation and hyperpigmentation during spring break. Mil Med. 2009;174:657-661.
- Smith LG. Phytophotodermatitis. Images Emerg Med. 2017;1:146-147.
- Wagner AM, Wu JJ, Hansen RC, et al. Bullous phytophotodermatitis associated with high natural concentrations of furanocoumarins in limes. Am J Contact Dermat. 2002;13:10-14.
- Green C, Ferguson J. Sesquiterpene lactone mix is not an adequate screen for Compositae allergy. Contact Dermatitis. 1994;31:151-153.
- Paulsen E, Andersen KE. Screening for Compositae contact sensitization with sesquiterpene lactones and Compositae mix 2.5% pet. Contact Dermatitis. 2019;81:368-373.
- Paulsen E, Andersen KE. Patch testing with constituents of Compositae mixes. Contact Dermatitis. 2012;66:241-246.
- Frosch PJ, Geier J, Uter W, et al. Patch testing with the patients’ own products. Contact Dermatitis. 2011:929-941.
- Marks JG. Allergic contact dermatitis to Alstroemeria. Arch Dermatol. 1988;124:914-916.
- Moreau JF, English JC, Gehris RP. Phytophotodermatitis. J Pediatr Adolesc Gynecol. 2014;27:93-94.
- Marks JG, Fowler JF, Sherertz EF, et al. Prevention of poison ivy and poison oak allergic contact dermatitis by quaternium-18 bentonite. J Am Acad Dermatol. 1995;33:212-216.
- Craig K, Meadows SE. What is the best duration of steroid therapy for contact dermatitis (rhus)? J Fam Pract. 2006;55:166-167.
Practice Points
- Gardeners, florists, farmers, and outdoor enthusiasts are at risk for various plant dermatoses, which can be classified into 5 main categories: allergic contact dermatitis (ACD), mechanical irritant contact dermatitis, chemical irritant contact dermatitis, light-mediated dermatitis, and pseudophytodermatitis.
- Poison ivy, from the Toxicodendron genus, is the leading cause of plant ACD; however, a myriad of other plants also can cause dermatoses.
- Patch testing can be used to identify the source of immune-mediated type IV delayed hypersensitivity reactions to various plant species in individuals with recurrent or persistent dermatitis.
- Treatment options for all plant dermatoses can include topical steroids, antihistamines, and oral prednisone. Prevention involves avoidance or use of an effective barrier.
Atopic Dermatitis: Evolution and Revolution in Therapy
Atopic dermatitis (AD) is an incredibly common chronic skin disease, affecting up to 25% of children and 7% of adults in the United States.1,2 Despite the prevalence of this disease and its impact on patient quality of life, research and scholarly work in AD has been limited until recent years. A PubMed search of articles indexed for MEDLINE using the term atopic dermatitis showed that there were fewer than 500 articles published in 2000 and 965 in 2010; with our more recent acceleration in research, there were 2168 articles published in 2020 and more than 1300 published in just the first half of 2021 (through June). This new research includes insights into the pathogenesis of AD and study of the disease impact and comorbidities as well as an extensive amount of drug development and clinical trial work for new topical and systemic therapies.
New Agents to Treat AD
The 2016 approval of crisaborole,3 a phosphodiesterase 4 inhibitor, followed by the approval of dupilumab, an IL-4 and IL-13 pathway inhibitor and the first biologic agent approved for AD,4 ushered in a new age of therapy. We currently are awaiting the incorporation of a new set of topical nonsteroidal agents, oral Janus kinase (JAK) inhibitors, and new biologic agents for AD, several of which have completed phase 3 trials and extended safety evaluations. How these new drugs will impact our standard treatment across the spectrum of care for AD is not yet known.
The emergence of new systemic therapies is timely, as the most used systemic medications previously were oral corticosteroids, despite their use being advised against in standard practice guidelines. Other agents such as methotrexate, cyclosporine, azathioprine, and mycophenolate are discussed in the literature and AD treatment guidelines as being potentially useful, though absence of US Food and Drug Administration (FDA) approval and the need for frequent laboratory monitoring, as well as drug-specific side effects and an increased risk of infection, limit their use in the United States, especially in pediatric and adolescent populations.5
The approval of dupilumab as a systemic therapy—initially for adults and subsequently for teenagers (12–17 years of age) and then children (6–11 years of age)—has markedly influenced the standard of care for moderate to severe AD. This agent has been shown to have a considerable impact on disease severity and quality of life, with a good safety profile and the added benefit of not requiring continuous (or any) laboratory monitoring.6-8 Ongoing studies of dupilumab in children (ClinicalTrials.gov identifiers NCT02612454, NCT03346434), including those younger than 1 year,9 raise the question of how commonly this medication might be incorporated into care across the entire age spectrum of patients with AD. What standards will there be for assessment of severity, disease impact, and persistence to warrant use in younger ages? Will early treatment with novel systemic agents change the overall course of the disease and minimize the development of comorbidities? The answers to these questions remain to be seen.
JAK Inhibitors for AD
Additional novel therapeutics currently are undergoing studies for treatment of AD, most notably the oral JAK inhibitors upadacitinib,10 baricitinib,11 and abrocitinib.12 Each of these agents has completed phase 3 trials for AD. Two of these agents—upadacitinib and baricitinib—have prior FDA approval for use in other disease states. Of note, baricitinib is already approved for treatment of moderate to severe AD in adults in more than 40 countries13; however, the use of these agents in other diseases brings about concerns of malignancy, severe infection, and thrombosis. In the clinical trials for AD, many of these events have not been seen, but the number of patients treated is limited, and longer-term safety assessment is important.10,11
How will the oral JAK inhibitors be incorporated into care compared to biologic agents such as dupilumab? Tolerance and more serious potential adverse events are concerns, with nausea, headaches, and acneform eruptions being associated with some of the medications, in addition to potential issues with herpes simplex and zoster infections. However, oral JAK inhibitors have the benefit of not requiring injections, something that many patients may prefer, and data show that these drugs generally are associated with a rapid reduction in pruritus and, depending on the drug, very quick and profound effects on objective signs of AD.10-12 Two head-to-head studies have been completed comparing dupilumab to oral JAK inhibitors in adults: the JADE COMPARE trial examining dupilumab vs abrocitinib12 and the Heads UP trial comparing dupilumab vs upadacitinib.14 Compared to dupilumab, higher-dose abrocitinib showed more rapid responses, superiority in itch response, and similarity or superiority in other outcomes depending on the time point of the evaluation. Adverse event profiles differed; for example, abrocitinib was associated with more nausea, acneform eruptions, and herpes zoster, while dupilumab had higher rates of conjunctivitis.12 Upadacitinib, which was only studied at higher dosing (30 mg daily), showed superiority to dupilumab in itch response and in improvement in AD severity in multiple outcome measures; however, there were increases in serious infections, eczema herpeticum, herpes zoster, and laboratory-related adverse events.14 Dupilumab has the advantage of studies of extended use along with real-world experience, generally with excellent safety and tolerance other than injection-site reactions and conjunctivitis.8 Biologics targeting IL-13—tralokinumab and lebrikizumab—also are to be added to our armamentarium.15,16 The addition of these agents and JAK inhibitors as new systemic treatment options points to the quickly evolving future of AD treatment for patients with extensive disease.
New topical therapies in development provide even more treatment options. New nonsteroidal topicals include topical JAK inhibitors such as ruxolitinib17; tapinarof,18 an aryl hydrocarbon receptor modulator; and phosphodiesterase 4 inhibitors. These agents may be useful either as monotherapy, as studied, potentially without the regional limitations associated with stronger topical corticosteroids, but also should be useful in clinical practice as part of therapeutic regimens with other topical steroid and nonsteroidal agents.
The Microbiome and AD
In addition, research looking at topical microbes as specific interventions that may mediate the microbiome and inflammation of AD are intriguing. A recent phase 1 trial from the University of California San Diego19 indicated that topical bacteriotherapy directed at decreasing Staphylococcus aureus may provide an impact in AD. Observations by Kong et al20 showed that gram-negative microbiome differences are seen in AD patients compared to unaffected individuals, which has fueled studies showing that Roseomonas mucosa, a gram-negative skin commensal, when applied as a topical live biotherapeutic agent has improved disease severity in children and adults with AD.21 Although further studies are underway, these initial data suggest a role for microbiome-modifying therapies as AD treatment.
Chronic Hand Eczema
Chronic hand eczema (CHE), which has considerable overlap with AD in many patients, especially children and adolescents,22-24 is another area of interesting research. This high-prevalence condition is associated with allergic and irritant contact dermatitis24-26—conditions that are both considered alternative diagnoses for and exacerbators of AD27—and is a disease process currently being targeted for new therapies. Delgocitinib (NCT04872101, NCT04871711), the novel JAK inhibitor ARQ-252 (NCT04378569), among other topical agents, as well as systemic therapeutics such as gusacitinib (NCT03728504), are in active trials for CHE. Given CHE’s impact on quality of life28 and its overlap with AD, investigation into this disorder can help drive future AD research as well as lead to better knowledge and treatment of CHE.
Final Thoughts
Despite the promising results of these myriad new therapies in AD, there are many factors that influence how and when we use these drugs, including their approval status, FDA labeling, and the ability of patients to access and afford treatment. Additionally, continued study is needed to evaluate the long-term safety and extended efficacy of newer drugs, such as the oral JAK inhibitors. Despite these hurdles, the current landscape of research and development is rapidly evolving. Compared to the many years when only one main group of therapies was a reasonable option for patients, the future of AD treatment looks bright.
- Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: section 1. diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70:338-351. doi:10.1016/j.jaad.2013.10.010
- Chiesa Fuxench ZC, Block JK, Boguniewicz M, et al. Atopic dermatitis in America study: a cross-sectional study examining the prevalence and disease burden of atopic dermatitis in the US adult population. J Invest Dermatol. 2019;139:583-590. doi:10.1016/j.jid.2018.08.028
- FDA approves Eucrisa for eczema. News release. US Food and Drug Administration; December 14, 2016. Accessed August 16, 2021. https://www.fda.gov/news-events/press-announcements/fda-approves-eucrisa-eczema
- Gooderham MJ, Hong HC, Eshtiaghi P, et al. Dupilumab: a review of its use in the treatment of atopic dermatitis. J Am Acad Dermatol. 2018;78(3 suppl 1):S28-S36. doi:10.1016/j.jaad.2017.12.022
- Sidbury R, Davis DM, Cohen DE, et al. Guidelines of care for the management of atopic dermatitis: section 3. management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;71:327-349. doi:10.1016/j.jaad.2014.03.030
- Paller AS, Siegfried EC, Thaçi D, et al. Efficacy and safety of dupilumab with concomitant topical corticosteroids in children 6 to 11 years old with severe atopic dermatitis: a randomized, double-blinded, placebo-controlled phase 3 trial. J Am Acad Dermatol. 2020;83:1282-1293. doi:10.1016/j.jaad.2020.06.054
- Simpson EL, Paller AS, Siegfried EC, et al. Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis: a phase 3 randomized clinical trial. JAMA Dermatol. 2020;156:44-56. doi:10.1001/jamadermatol.2019.3336
- Deleuran M, Thaçi D, Beck LA, et al. Dupilumab shows long-term safety and efficacy in patients with moderate to severe atopic dermatitis enrolled in a phase 3 open-label extension study. J Am Acad Dermatol. 2020;82:377-388. doi:10.1016/j.jaad.2019.07.074
- Paller AS, Siegfried EC, Simpson EL, et al. A phase 2, open-label study of single-dose dupilumab in children aged 6 months to <6 years with severe uncontrolled atopic dermatitis: pharmacokinetics, safety and efficacy. J Eur Acad Dermatol Venereol. 2021;35:464-475. doi: 10.1111/jdv.16928
- Reich K, Teixeira HD, de Bruin-Weller M, et al. Safety and efficacy of upadacitinib in combination with topical corticosteroids in adolescents and adults with moderate-to-severe atopic dermatitis (AD Up): results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2021;397:2169-2181. doi:10.1016/S0140-6736(21)00589-4
- Simpson EL, Forman S, Silverberg JI, et al. Baricitinib in patients with moderate-to-severe atopic dermatitis: results from a randomized monotherapy phase 3 trial in the United States and Canada (BREEZE-AD5). J Am Acad Dermatol. 2021;85:62-70. doi:10.1016/j.jaad.2021.02.028
- Bieber T, Simpson EL, Silverberg JI, et al. Abrocitinib versus placebo or dupilumab for atopic dermatitis. N Engl J Med. 2021;384:1101-1112. doi:10.1056/NEJMoa2019380
- Lilly and Incyte provide update on supplemental New Drug Application for baricitinib for the treatment of moderate to severe atopic dermatitis. News release. Eli Lilly and Company; July 16, 2021. Accessed August 16, 2021. https://investor.lilly.com/news-releases/news-release-details/lilly-and-incyte-provide-update-supplemental new-drug
- Blauvelt A, Teixeira HD, Simpson EL, et al. Efficacy and safety of upadacitinib vs dupilumab in adults with moderate-to-severe atopic dermatitis: a randomized clinical trial [published online August 4, 2021]. JAMA Dermatol. doi:10.1001/jamadermatol.2021.3023
- Guttman-Yassky E, Blauvelt A, Eichenfield LF, et al. Efficacy and safety of lebrikizumab, a high-affinity interleukin 13 inhibitor, in adults with moderate to severe atopic dermatitis: a phase 2b randomized clinical trial. JAMA Dermatol. 2020;156:411-420. doi:10.1001/jamadermatol.2020.0079
- Silverberg JI, Toth D, Bieber T, et al. Tralokinumab plus topical corticosteroids for the treatment of moderate-to-severe atopic dermatitis: results from the double-blind, randomized, multicentre,placebo-controlled phase III ECZTRA 3 trial. Br J Dermatol. 2021;184:450-463. doi:10.1111/bjd.19573
- Papp K, Szepietowski JC, Kircik L, et al. Efficacy and safety of ruxolitinib cream for the treatment of atopic dermatitis: results from 2 phase 3, randomized, double-blind studies [published online May 4, 2021]. J Am Acad Dermatol. doi:10.1016/j.jaad.2021.04.085
- Paller AS, Stein Gold L, Soung J, et al. Efficacy and patient-reported outcomes from a phase 2b, randomized clinical trial of tapinarof cream for the treatment of adolescents and adults with atopic dermatitis. J Am Acad Dermatol. 2021;84:632-638. doi:10.1016/j.jaad.2020.05.135
- Nakatsuji, T, Hata TR, Tong Y, et al. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial [published online February 22, 2021]. Nat Med. 2021;27:700-709. doi:10.1038/s41591-021-01256-2
- Kong HH, Oh J, Deming C, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012;22:850-859. doi:10.1101/gr.131029.111
- Myles IA, Castillo CR, Barbian KD, et al. Therapeutic responses to Roseomonas mucosa in atopic dermatitis may involve lipid-mediated TNF-related epithelial repair. Sci Transl Med. 2020;12:eaaz8631. doi:10.1126/scitranslmed.aaz8631
- Mortz CG, Lauritsen JM, Bindslev-Jensen C, et al. Prevalence of atopic dermatitis, asthma, allergic rhinitis, and hand and contact dermatitis in adolescents. The Odense Adolescence Cohort Study on Atopic Diseases and Dermatitis. Br J Dermatol. 2001;144:523-532. doi:10.1046/j.1365-2133.2001.04078.x
- Grönhagen C, Lidén C, Wahlgren CF, et al. Hand eczema and atopic dermatitis in adolescents: a prospective cohort study from the BAMSE project. Br J Dermatol. 2015;173:1175-1182. doi:10.1111/bjd.14019
- Mortz CG, Lauritsen JM, Bindslev-Jensen C, et al. Contact allergy and allergic contact dermatitis in adolescents: prevalence measures and associations. The Odense Adolescence Cohort Study on Atopic Diseases and Dermatitis (TOACS). Acta Derm Venereol. 2002;82:352-358. doi:10.1080/000155502320624087
- Isaksson M, Olhardt S, Rådehed J, et al. Children with atopic dermatitis should always be patch-tested if they have hand or foot dermatitis. Acta Derm Venereol. 2015;95:583-586. doi:10.2340/00015555-1995
- Silverberg JI, Warshaw EM, Maibach HI, et al. Hand eczema in children referred for patch testing: North American Contact Dermatitis Group Data, 2000-2016. Br J Dermatol. 2021;185:185-194. doi:10.1111/bjd.19818
- Agner T, Elsner P. Hand eczema: epidemiology, prognosis and prevention. J Eur Acad Dermatol Venereol. 2020;34(suppl 1):4-12. doi:10.1111/jdv.16061
- Cazzaniga S, Ballmer-Weber BK, Gräni N, et al. Medical, psychological and socio-economic implications of chronic hand eczema: a cross-sectional study. J Eur Acad Dermatol Venereol. 2016;30:628-637. doi:10.1111/jdv.13479
Atopic dermatitis (AD) is an incredibly common chronic skin disease, affecting up to 25% of children and 7% of adults in the United States.1,2 Despite the prevalence of this disease and its impact on patient quality of life, research and scholarly work in AD has been limited until recent years. A PubMed search of articles indexed for MEDLINE using the term atopic dermatitis showed that there were fewer than 500 articles published in 2000 and 965 in 2010; with our more recent acceleration in research, there were 2168 articles published in 2020 and more than 1300 published in just the first half of 2021 (through June). This new research includes insights into the pathogenesis of AD and study of the disease impact and comorbidities as well as an extensive amount of drug development and clinical trial work for new topical and systemic therapies.
New Agents to Treat AD
The 2016 approval of crisaborole,3 a phosphodiesterase 4 inhibitor, followed by the approval of dupilumab, an IL-4 and IL-13 pathway inhibitor and the first biologic agent approved for AD,4 ushered in a new age of therapy. We currently are awaiting the incorporation of a new set of topical nonsteroidal agents, oral Janus kinase (JAK) inhibitors, and new biologic agents for AD, several of which have completed phase 3 trials and extended safety evaluations. How these new drugs will impact our standard treatment across the spectrum of care for AD is not yet known.
The emergence of new systemic therapies is timely, as the most used systemic medications previously were oral corticosteroids, despite their use being advised against in standard practice guidelines. Other agents such as methotrexate, cyclosporine, azathioprine, and mycophenolate are discussed in the literature and AD treatment guidelines as being potentially useful, though absence of US Food and Drug Administration (FDA) approval and the need for frequent laboratory monitoring, as well as drug-specific side effects and an increased risk of infection, limit their use in the United States, especially in pediatric and adolescent populations.5
The approval of dupilumab as a systemic therapy—initially for adults and subsequently for teenagers (12–17 years of age) and then children (6–11 years of age)—has markedly influenced the standard of care for moderate to severe AD. This agent has been shown to have a considerable impact on disease severity and quality of life, with a good safety profile and the added benefit of not requiring continuous (or any) laboratory monitoring.6-8 Ongoing studies of dupilumab in children (ClinicalTrials.gov identifiers NCT02612454, NCT03346434), including those younger than 1 year,9 raise the question of how commonly this medication might be incorporated into care across the entire age spectrum of patients with AD. What standards will there be for assessment of severity, disease impact, and persistence to warrant use in younger ages? Will early treatment with novel systemic agents change the overall course of the disease and minimize the development of comorbidities? The answers to these questions remain to be seen.
JAK Inhibitors for AD
Additional novel therapeutics currently are undergoing studies for treatment of AD, most notably the oral JAK inhibitors upadacitinib,10 baricitinib,11 and abrocitinib.12 Each of these agents has completed phase 3 trials for AD. Two of these agents—upadacitinib and baricitinib—have prior FDA approval for use in other disease states. Of note, baricitinib is already approved for treatment of moderate to severe AD in adults in more than 40 countries13; however, the use of these agents in other diseases brings about concerns of malignancy, severe infection, and thrombosis. In the clinical trials for AD, many of these events have not been seen, but the number of patients treated is limited, and longer-term safety assessment is important.10,11
How will the oral JAK inhibitors be incorporated into care compared to biologic agents such as dupilumab? Tolerance and more serious potential adverse events are concerns, with nausea, headaches, and acneform eruptions being associated with some of the medications, in addition to potential issues with herpes simplex and zoster infections. However, oral JAK inhibitors have the benefit of not requiring injections, something that many patients may prefer, and data show that these drugs generally are associated with a rapid reduction in pruritus and, depending on the drug, very quick and profound effects on objective signs of AD.10-12 Two head-to-head studies have been completed comparing dupilumab to oral JAK inhibitors in adults: the JADE COMPARE trial examining dupilumab vs abrocitinib12 and the Heads UP trial comparing dupilumab vs upadacitinib.14 Compared to dupilumab, higher-dose abrocitinib showed more rapid responses, superiority in itch response, and similarity or superiority in other outcomes depending on the time point of the evaluation. Adverse event profiles differed; for example, abrocitinib was associated with more nausea, acneform eruptions, and herpes zoster, while dupilumab had higher rates of conjunctivitis.12 Upadacitinib, which was only studied at higher dosing (30 mg daily), showed superiority to dupilumab in itch response and in improvement in AD severity in multiple outcome measures; however, there were increases in serious infections, eczema herpeticum, herpes zoster, and laboratory-related adverse events.14 Dupilumab has the advantage of studies of extended use along with real-world experience, generally with excellent safety and tolerance other than injection-site reactions and conjunctivitis.8 Biologics targeting IL-13—tralokinumab and lebrikizumab—also are to be added to our armamentarium.15,16 The addition of these agents and JAK inhibitors as new systemic treatment options points to the quickly evolving future of AD treatment for patients with extensive disease.
New topical therapies in development provide even more treatment options. New nonsteroidal topicals include topical JAK inhibitors such as ruxolitinib17; tapinarof,18 an aryl hydrocarbon receptor modulator; and phosphodiesterase 4 inhibitors. These agents may be useful either as monotherapy, as studied, potentially without the regional limitations associated with stronger topical corticosteroids, but also should be useful in clinical practice as part of therapeutic regimens with other topical steroid and nonsteroidal agents.
The Microbiome and AD
In addition, research looking at topical microbes as specific interventions that may mediate the microbiome and inflammation of AD are intriguing. A recent phase 1 trial from the University of California San Diego19 indicated that topical bacteriotherapy directed at decreasing Staphylococcus aureus may provide an impact in AD. Observations by Kong et al20 showed that gram-negative microbiome differences are seen in AD patients compared to unaffected individuals, which has fueled studies showing that Roseomonas mucosa, a gram-negative skin commensal, when applied as a topical live biotherapeutic agent has improved disease severity in children and adults with AD.21 Although further studies are underway, these initial data suggest a role for microbiome-modifying therapies as AD treatment.
Chronic Hand Eczema
Chronic hand eczema (CHE), which has considerable overlap with AD in many patients, especially children and adolescents,22-24 is another area of interesting research. This high-prevalence condition is associated with allergic and irritant contact dermatitis24-26—conditions that are both considered alternative diagnoses for and exacerbators of AD27—and is a disease process currently being targeted for new therapies. Delgocitinib (NCT04872101, NCT04871711), the novel JAK inhibitor ARQ-252 (NCT04378569), among other topical agents, as well as systemic therapeutics such as gusacitinib (NCT03728504), are in active trials for CHE. Given CHE’s impact on quality of life28 and its overlap with AD, investigation into this disorder can help drive future AD research as well as lead to better knowledge and treatment of CHE.
Final Thoughts
Despite the promising results of these myriad new therapies in AD, there are many factors that influence how and when we use these drugs, including their approval status, FDA labeling, and the ability of patients to access and afford treatment. Additionally, continued study is needed to evaluate the long-term safety and extended efficacy of newer drugs, such as the oral JAK inhibitors. Despite these hurdles, the current landscape of research and development is rapidly evolving. Compared to the many years when only one main group of therapies was a reasonable option for patients, the future of AD treatment looks bright.
Atopic dermatitis (AD) is an incredibly common chronic skin disease, affecting up to 25% of children and 7% of adults in the United States.1,2 Despite the prevalence of this disease and its impact on patient quality of life, research and scholarly work in AD has been limited until recent years. A PubMed search of articles indexed for MEDLINE using the term atopic dermatitis showed that there were fewer than 500 articles published in 2000 and 965 in 2010; with our more recent acceleration in research, there were 2168 articles published in 2020 and more than 1300 published in just the first half of 2021 (through June). This new research includes insights into the pathogenesis of AD and study of the disease impact and comorbidities as well as an extensive amount of drug development and clinical trial work for new topical and systemic therapies.
New Agents to Treat AD
The 2016 approval of crisaborole,3 a phosphodiesterase 4 inhibitor, followed by the approval of dupilumab, an IL-4 and IL-13 pathway inhibitor and the first biologic agent approved for AD,4 ushered in a new age of therapy. We currently are awaiting the incorporation of a new set of topical nonsteroidal agents, oral Janus kinase (JAK) inhibitors, and new biologic agents for AD, several of which have completed phase 3 trials and extended safety evaluations. How these new drugs will impact our standard treatment across the spectrum of care for AD is not yet known.
The emergence of new systemic therapies is timely, as the most used systemic medications previously were oral corticosteroids, despite their use being advised against in standard practice guidelines. Other agents such as methotrexate, cyclosporine, azathioprine, and mycophenolate are discussed in the literature and AD treatment guidelines as being potentially useful, though absence of US Food and Drug Administration (FDA) approval and the need for frequent laboratory monitoring, as well as drug-specific side effects and an increased risk of infection, limit their use in the United States, especially in pediatric and adolescent populations.5
The approval of dupilumab as a systemic therapy—initially for adults and subsequently for teenagers (12–17 years of age) and then children (6–11 years of age)—has markedly influenced the standard of care for moderate to severe AD. This agent has been shown to have a considerable impact on disease severity and quality of life, with a good safety profile and the added benefit of not requiring continuous (or any) laboratory monitoring.6-8 Ongoing studies of dupilumab in children (ClinicalTrials.gov identifiers NCT02612454, NCT03346434), including those younger than 1 year,9 raise the question of how commonly this medication might be incorporated into care across the entire age spectrum of patients with AD. What standards will there be for assessment of severity, disease impact, and persistence to warrant use in younger ages? Will early treatment with novel systemic agents change the overall course of the disease and minimize the development of comorbidities? The answers to these questions remain to be seen.
JAK Inhibitors for AD
Additional novel therapeutics currently are undergoing studies for treatment of AD, most notably the oral JAK inhibitors upadacitinib,10 baricitinib,11 and abrocitinib.12 Each of these agents has completed phase 3 trials for AD. Two of these agents—upadacitinib and baricitinib—have prior FDA approval for use in other disease states. Of note, baricitinib is already approved for treatment of moderate to severe AD in adults in more than 40 countries13; however, the use of these agents in other diseases brings about concerns of malignancy, severe infection, and thrombosis. In the clinical trials for AD, many of these events have not been seen, but the number of patients treated is limited, and longer-term safety assessment is important.10,11
How will the oral JAK inhibitors be incorporated into care compared to biologic agents such as dupilumab? Tolerance and more serious potential adverse events are concerns, with nausea, headaches, and acneform eruptions being associated with some of the medications, in addition to potential issues with herpes simplex and zoster infections. However, oral JAK inhibitors have the benefit of not requiring injections, something that many patients may prefer, and data show that these drugs generally are associated with a rapid reduction in pruritus and, depending on the drug, very quick and profound effects on objective signs of AD.10-12 Two head-to-head studies have been completed comparing dupilumab to oral JAK inhibitors in adults: the JADE COMPARE trial examining dupilumab vs abrocitinib12 and the Heads UP trial comparing dupilumab vs upadacitinib.14 Compared to dupilumab, higher-dose abrocitinib showed more rapid responses, superiority in itch response, and similarity or superiority in other outcomes depending on the time point of the evaluation. Adverse event profiles differed; for example, abrocitinib was associated with more nausea, acneform eruptions, and herpes zoster, while dupilumab had higher rates of conjunctivitis.12 Upadacitinib, which was only studied at higher dosing (30 mg daily), showed superiority to dupilumab in itch response and in improvement in AD severity in multiple outcome measures; however, there were increases in serious infections, eczema herpeticum, herpes zoster, and laboratory-related adverse events.14 Dupilumab has the advantage of studies of extended use along with real-world experience, generally with excellent safety and tolerance other than injection-site reactions and conjunctivitis.8 Biologics targeting IL-13—tralokinumab and lebrikizumab—also are to be added to our armamentarium.15,16 The addition of these agents and JAK inhibitors as new systemic treatment options points to the quickly evolving future of AD treatment for patients with extensive disease.
New topical therapies in development provide even more treatment options. New nonsteroidal topicals include topical JAK inhibitors such as ruxolitinib17; tapinarof,18 an aryl hydrocarbon receptor modulator; and phosphodiesterase 4 inhibitors. These agents may be useful either as monotherapy, as studied, potentially without the regional limitations associated with stronger topical corticosteroids, but also should be useful in clinical practice as part of therapeutic regimens with other topical steroid and nonsteroidal agents.
The Microbiome and AD
In addition, research looking at topical microbes as specific interventions that may mediate the microbiome and inflammation of AD are intriguing. A recent phase 1 trial from the University of California San Diego19 indicated that topical bacteriotherapy directed at decreasing Staphylococcus aureus may provide an impact in AD. Observations by Kong et al20 showed that gram-negative microbiome differences are seen in AD patients compared to unaffected individuals, which has fueled studies showing that Roseomonas mucosa, a gram-negative skin commensal, when applied as a topical live biotherapeutic agent has improved disease severity in children and adults with AD.21 Although further studies are underway, these initial data suggest a role for microbiome-modifying therapies as AD treatment.
Chronic Hand Eczema
Chronic hand eczema (CHE), which has considerable overlap with AD in many patients, especially children and adolescents,22-24 is another area of interesting research. This high-prevalence condition is associated with allergic and irritant contact dermatitis24-26—conditions that are both considered alternative diagnoses for and exacerbators of AD27—and is a disease process currently being targeted for new therapies. Delgocitinib (NCT04872101, NCT04871711), the novel JAK inhibitor ARQ-252 (NCT04378569), among other topical agents, as well as systemic therapeutics such as gusacitinib (NCT03728504), are in active trials for CHE. Given CHE’s impact on quality of life28 and its overlap with AD, investigation into this disorder can help drive future AD research as well as lead to better knowledge and treatment of CHE.
Final Thoughts
Despite the promising results of these myriad new therapies in AD, there are many factors that influence how and when we use these drugs, including their approval status, FDA labeling, and the ability of patients to access and afford treatment. Additionally, continued study is needed to evaluate the long-term safety and extended efficacy of newer drugs, such as the oral JAK inhibitors. Despite these hurdles, the current landscape of research and development is rapidly evolving. Compared to the many years when only one main group of therapies was a reasonable option for patients, the future of AD treatment looks bright.
- Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: section 1. diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70:338-351. doi:10.1016/j.jaad.2013.10.010
- Chiesa Fuxench ZC, Block JK, Boguniewicz M, et al. Atopic dermatitis in America study: a cross-sectional study examining the prevalence and disease burden of atopic dermatitis in the US adult population. J Invest Dermatol. 2019;139:583-590. doi:10.1016/j.jid.2018.08.028
- FDA approves Eucrisa for eczema. News release. US Food and Drug Administration; December 14, 2016. Accessed August 16, 2021. https://www.fda.gov/news-events/press-announcements/fda-approves-eucrisa-eczema
- Gooderham MJ, Hong HC, Eshtiaghi P, et al. Dupilumab: a review of its use in the treatment of atopic dermatitis. J Am Acad Dermatol. 2018;78(3 suppl 1):S28-S36. doi:10.1016/j.jaad.2017.12.022
- Sidbury R, Davis DM, Cohen DE, et al. Guidelines of care for the management of atopic dermatitis: section 3. management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;71:327-349. doi:10.1016/j.jaad.2014.03.030
- Paller AS, Siegfried EC, Thaçi D, et al. Efficacy and safety of dupilumab with concomitant topical corticosteroids in children 6 to 11 years old with severe atopic dermatitis: a randomized, double-blinded, placebo-controlled phase 3 trial. J Am Acad Dermatol. 2020;83:1282-1293. doi:10.1016/j.jaad.2020.06.054
- Simpson EL, Paller AS, Siegfried EC, et al. Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis: a phase 3 randomized clinical trial. JAMA Dermatol. 2020;156:44-56. doi:10.1001/jamadermatol.2019.3336
- Deleuran M, Thaçi D, Beck LA, et al. Dupilumab shows long-term safety and efficacy in patients with moderate to severe atopic dermatitis enrolled in a phase 3 open-label extension study. J Am Acad Dermatol. 2020;82:377-388. doi:10.1016/j.jaad.2019.07.074
- Paller AS, Siegfried EC, Simpson EL, et al. A phase 2, open-label study of single-dose dupilumab in children aged 6 months to <6 years with severe uncontrolled atopic dermatitis: pharmacokinetics, safety and efficacy. J Eur Acad Dermatol Venereol. 2021;35:464-475. doi: 10.1111/jdv.16928
- Reich K, Teixeira HD, de Bruin-Weller M, et al. Safety and efficacy of upadacitinib in combination with topical corticosteroids in adolescents and adults with moderate-to-severe atopic dermatitis (AD Up): results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2021;397:2169-2181. doi:10.1016/S0140-6736(21)00589-4
- Simpson EL, Forman S, Silverberg JI, et al. Baricitinib in patients with moderate-to-severe atopic dermatitis: results from a randomized monotherapy phase 3 trial in the United States and Canada (BREEZE-AD5). J Am Acad Dermatol. 2021;85:62-70. doi:10.1016/j.jaad.2021.02.028
- Bieber T, Simpson EL, Silverberg JI, et al. Abrocitinib versus placebo or dupilumab for atopic dermatitis. N Engl J Med. 2021;384:1101-1112. doi:10.1056/NEJMoa2019380
- Lilly and Incyte provide update on supplemental New Drug Application for baricitinib for the treatment of moderate to severe atopic dermatitis. News release. Eli Lilly and Company; July 16, 2021. Accessed August 16, 2021. https://investor.lilly.com/news-releases/news-release-details/lilly-and-incyte-provide-update-supplemental new-drug
- Blauvelt A, Teixeira HD, Simpson EL, et al. Efficacy and safety of upadacitinib vs dupilumab in adults with moderate-to-severe atopic dermatitis: a randomized clinical trial [published online August 4, 2021]. JAMA Dermatol. doi:10.1001/jamadermatol.2021.3023
- Guttman-Yassky E, Blauvelt A, Eichenfield LF, et al. Efficacy and safety of lebrikizumab, a high-affinity interleukin 13 inhibitor, in adults with moderate to severe atopic dermatitis: a phase 2b randomized clinical trial. JAMA Dermatol. 2020;156:411-420. doi:10.1001/jamadermatol.2020.0079
- Silverberg JI, Toth D, Bieber T, et al. Tralokinumab plus topical corticosteroids for the treatment of moderate-to-severe atopic dermatitis: results from the double-blind, randomized, multicentre,placebo-controlled phase III ECZTRA 3 trial. Br J Dermatol. 2021;184:450-463. doi:10.1111/bjd.19573
- Papp K, Szepietowski JC, Kircik L, et al. Efficacy and safety of ruxolitinib cream for the treatment of atopic dermatitis: results from 2 phase 3, randomized, double-blind studies [published online May 4, 2021]. J Am Acad Dermatol. doi:10.1016/j.jaad.2021.04.085
- Paller AS, Stein Gold L, Soung J, et al. Efficacy and patient-reported outcomes from a phase 2b, randomized clinical trial of tapinarof cream for the treatment of adolescents and adults with atopic dermatitis. J Am Acad Dermatol. 2021;84:632-638. doi:10.1016/j.jaad.2020.05.135
- Nakatsuji, T, Hata TR, Tong Y, et al. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial [published online February 22, 2021]. Nat Med. 2021;27:700-709. doi:10.1038/s41591-021-01256-2
- Kong HH, Oh J, Deming C, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012;22:850-859. doi:10.1101/gr.131029.111
- Myles IA, Castillo CR, Barbian KD, et al. Therapeutic responses to Roseomonas mucosa in atopic dermatitis may involve lipid-mediated TNF-related epithelial repair. Sci Transl Med. 2020;12:eaaz8631. doi:10.1126/scitranslmed.aaz8631
- Mortz CG, Lauritsen JM, Bindslev-Jensen C, et al. Prevalence of atopic dermatitis, asthma, allergic rhinitis, and hand and contact dermatitis in adolescents. The Odense Adolescence Cohort Study on Atopic Diseases and Dermatitis. Br J Dermatol. 2001;144:523-532. doi:10.1046/j.1365-2133.2001.04078.x
- Grönhagen C, Lidén C, Wahlgren CF, et al. Hand eczema and atopic dermatitis in adolescents: a prospective cohort study from the BAMSE project. Br J Dermatol. 2015;173:1175-1182. doi:10.1111/bjd.14019
- Mortz CG, Lauritsen JM, Bindslev-Jensen C, et al. Contact allergy and allergic contact dermatitis in adolescents: prevalence measures and associations. The Odense Adolescence Cohort Study on Atopic Diseases and Dermatitis (TOACS). Acta Derm Venereol. 2002;82:352-358. doi:10.1080/000155502320624087
- Isaksson M, Olhardt S, Rådehed J, et al. Children with atopic dermatitis should always be patch-tested if they have hand or foot dermatitis. Acta Derm Venereol. 2015;95:583-586. doi:10.2340/00015555-1995
- Silverberg JI, Warshaw EM, Maibach HI, et al. Hand eczema in children referred for patch testing: North American Contact Dermatitis Group Data, 2000-2016. Br J Dermatol. 2021;185:185-194. doi:10.1111/bjd.19818
- Agner T, Elsner P. Hand eczema: epidemiology, prognosis and prevention. J Eur Acad Dermatol Venereol. 2020;34(suppl 1):4-12. doi:10.1111/jdv.16061
- Cazzaniga S, Ballmer-Weber BK, Gräni N, et al. Medical, psychological and socio-economic implications of chronic hand eczema: a cross-sectional study. J Eur Acad Dermatol Venereol. 2016;30:628-637. doi:10.1111/jdv.13479
- Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: section 1. diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70:338-351. doi:10.1016/j.jaad.2013.10.010
- Chiesa Fuxench ZC, Block JK, Boguniewicz M, et al. Atopic dermatitis in America study: a cross-sectional study examining the prevalence and disease burden of atopic dermatitis in the US adult population. J Invest Dermatol. 2019;139:583-590. doi:10.1016/j.jid.2018.08.028
- FDA approves Eucrisa for eczema. News release. US Food and Drug Administration; December 14, 2016. Accessed August 16, 2021. https://www.fda.gov/news-events/press-announcements/fda-approves-eucrisa-eczema
- Gooderham MJ, Hong HC, Eshtiaghi P, et al. Dupilumab: a review of its use in the treatment of atopic dermatitis. J Am Acad Dermatol. 2018;78(3 suppl 1):S28-S36. doi:10.1016/j.jaad.2017.12.022
- Sidbury R, Davis DM, Cohen DE, et al. Guidelines of care for the management of atopic dermatitis: section 3. management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;71:327-349. doi:10.1016/j.jaad.2014.03.030
- Paller AS, Siegfried EC, Thaçi D, et al. Efficacy and safety of dupilumab with concomitant topical corticosteroids in children 6 to 11 years old with severe atopic dermatitis: a randomized, double-blinded, placebo-controlled phase 3 trial. J Am Acad Dermatol. 2020;83:1282-1293. doi:10.1016/j.jaad.2020.06.054
- Simpson EL, Paller AS, Siegfried EC, et al. Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis: a phase 3 randomized clinical trial. JAMA Dermatol. 2020;156:44-56. doi:10.1001/jamadermatol.2019.3336
- Deleuran M, Thaçi D, Beck LA, et al. Dupilumab shows long-term safety and efficacy in patients with moderate to severe atopic dermatitis enrolled in a phase 3 open-label extension study. J Am Acad Dermatol. 2020;82:377-388. doi:10.1016/j.jaad.2019.07.074
- Paller AS, Siegfried EC, Simpson EL, et al. A phase 2, open-label study of single-dose dupilumab in children aged 6 months to <6 years with severe uncontrolled atopic dermatitis: pharmacokinetics, safety and efficacy. J Eur Acad Dermatol Venereol. 2021;35:464-475. doi: 10.1111/jdv.16928
- Reich K, Teixeira HD, de Bruin-Weller M, et al. Safety and efficacy of upadacitinib in combination with topical corticosteroids in adolescents and adults with moderate-to-severe atopic dermatitis (AD Up): results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2021;397:2169-2181. doi:10.1016/S0140-6736(21)00589-4
- Simpson EL, Forman S, Silverberg JI, et al. Baricitinib in patients with moderate-to-severe atopic dermatitis: results from a randomized monotherapy phase 3 trial in the United States and Canada (BREEZE-AD5). J Am Acad Dermatol. 2021;85:62-70. doi:10.1016/j.jaad.2021.02.028
- Bieber T, Simpson EL, Silverberg JI, et al. Abrocitinib versus placebo or dupilumab for atopic dermatitis. N Engl J Med. 2021;384:1101-1112. doi:10.1056/NEJMoa2019380
- Lilly and Incyte provide update on supplemental New Drug Application for baricitinib for the treatment of moderate to severe atopic dermatitis. News release. Eli Lilly and Company; July 16, 2021. Accessed August 16, 2021. https://investor.lilly.com/news-releases/news-release-details/lilly-and-incyte-provide-update-supplemental new-drug
- Blauvelt A, Teixeira HD, Simpson EL, et al. Efficacy and safety of upadacitinib vs dupilumab in adults with moderate-to-severe atopic dermatitis: a randomized clinical trial [published online August 4, 2021]. JAMA Dermatol. doi:10.1001/jamadermatol.2021.3023
- Guttman-Yassky E, Blauvelt A, Eichenfield LF, et al. Efficacy and safety of lebrikizumab, a high-affinity interleukin 13 inhibitor, in adults with moderate to severe atopic dermatitis: a phase 2b randomized clinical trial. JAMA Dermatol. 2020;156:411-420. doi:10.1001/jamadermatol.2020.0079
- Silverberg JI, Toth D, Bieber T, et al. Tralokinumab plus topical corticosteroids for the treatment of moderate-to-severe atopic dermatitis: results from the double-blind, randomized, multicentre,placebo-controlled phase III ECZTRA 3 trial. Br J Dermatol. 2021;184:450-463. doi:10.1111/bjd.19573
- Papp K, Szepietowski JC, Kircik L, et al. Efficacy and safety of ruxolitinib cream for the treatment of atopic dermatitis: results from 2 phase 3, randomized, double-blind studies [published online May 4, 2021]. J Am Acad Dermatol. doi:10.1016/j.jaad.2021.04.085
- Paller AS, Stein Gold L, Soung J, et al. Efficacy and patient-reported outcomes from a phase 2b, randomized clinical trial of tapinarof cream for the treatment of adolescents and adults with atopic dermatitis. J Am Acad Dermatol. 2021;84:632-638. doi:10.1016/j.jaad.2020.05.135
- Nakatsuji, T, Hata TR, Tong Y, et al. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial [published online February 22, 2021]. Nat Med. 2021;27:700-709. doi:10.1038/s41591-021-01256-2
- Kong HH, Oh J, Deming C, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012;22:850-859. doi:10.1101/gr.131029.111
- Myles IA, Castillo CR, Barbian KD, et al. Therapeutic responses to Roseomonas mucosa in atopic dermatitis may involve lipid-mediated TNF-related epithelial repair. Sci Transl Med. 2020;12:eaaz8631. doi:10.1126/scitranslmed.aaz8631
- Mortz CG, Lauritsen JM, Bindslev-Jensen C, et al. Prevalence of atopic dermatitis, asthma, allergic rhinitis, and hand and contact dermatitis in adolescents. The Odense Adolescence Cohort Study on Atopic Diseases and Dermatitis. Br J Dermatol. 2001;144:523-532. doi:10.1046/j.1365-2133.2001.04078.x
- Grönhagen C, Lidén C, Wahlgren CF, et al. Hand eczema and atopic dermatitis in adolescents: a prospective cohort study from the BAMSE project. Br J Dermatol. 2015;173:1175-1182. doi:10.1111/bjd.14019
- Mortz CG, Lauritsen JM, Bindslev-Jensen C, et al. Contact allergy and allergic contact dermatitis in adolescents: prevalence measures and associations. The Odense Adolescence Cohort Study on Atopic Diseases and Dermatitis (TOACS). Acta Derm Venereol. 2002;82:352-358. doi:10.1080/000155502320624087
- Isaksson M, Olhardt S, Rådehed J, et al. Children with atopic dermatitis should always be patch-tested if they have hand or foot dermatitis. Acta Derm Venereol. 2015;95:583-586. doi:10.2340/00015555-1995
- Silverberg JI, Warshaw EM, Maibach HI, et al. Hand eczema in children referred for patch testing: North American Contact Dermatitis Group Data, 2000-2016. Br J Dermatol. 2021;185:185-194. doi:10.1111/bjd.19818
- Agner T, Elsner P. Hand eczema: epidemiology, prognosis and prevention. J Eur Acad Dermatol Venereol. 2020;34(suppl 1):4-12. doi:10.1111/jdv.16061
- Cazzaniga S, Ballmer-Weber BK, Gräni N, et al. Medical, psychological and socio-economic implications of chronic hand eczema: a cross-sectional study. J Eur Acad Dermatol Venereol. 2016;30:628-637. doi:10.1111/jdv.13479
Lesions on the Thigh After an Organ Transplant
The Diagnosis: Microcystic Lymphatic Malformation
The shave biopsy demonstrated numerous thin-walled vascular spaces filled with lymphatic fluid within the dermis (Figure), consistent with a diagnosis of microcystic lymphatic malformation (LM). Lymphatic malformations represent a class of benign vascular lesions consisting of anomalous or dilated lymphatic vessels, which can be broadly categorized as macrocystic (formerly cavernous lymphangioma or cystic hygroma), microcystic (formerly lymphangioma circumscriptum), or mixed.1 Patients often will present with pruritus, crusting, secondary infection, edema, or oozing.2 The superficial blebs of microcystic LMs resemble frog spawn and range in color from clear to pink, brawny, or deep maroon.3 Although the lymphatic vessels involved in microcystic LMs appear disconnected from the major lymphatic circulation,3 systemic fluid overload could plausibly promote lesional swelling and tenderness; we attributed our patient's worsening symptoms to the cumulative 7.8 L of intravenous fluid he received intraoperatively during his cardiac transplant. The excess fluid allowed communication between lymphatic cisterns and thin-walled vesicles on the skin surface through dilated channels. Overall, LMs represent roughly 26% of pediatric benign vascular tumors and approximately 4% of all vascular tumors.4
Although microcystic LMs may appear especially vascular or verrucous, the differential diagnosis for our patient's LM included condyloma acuminatum,5,6 condyloma lata,7 epidermal nevus, and lymphangiosarcoma. Epidermal nevi are congenital lesions, varying in appearance from velvety to verrucous patches and plaques that often evolve during puberty and become thicker, more verrucous, and hyperpigmented. Keratinocytic epidermal nevus syndromes and other entities such as nevus sebaceous have been associated with somatic mutations affecting proteins in the fibroblast growth factor receptor signaling pathway (eg, FGFR3, HRAS).8 Although the clinical appearance alone may be similar, lymphangiosarcoma can be distinguished from LM via biopsy.
There are several methods to diagnose LM. Duplex sonography is possibly the best noninvasive method to identify the flow between venous valves. Magnetic resonance imaging can detect larger occurrences of LM, and lymphangiography can be utilized to confirm a normal or abnormal lymphatic network.4 Treatment options are broad, including surgical excision, laser ablation, and topical sirolimus. Hypertonic saline sclerotherapy can be injected into the afflicted lymphatic channels to decrease inflammation, erythema, and hyperpigmentation without further treatment or major side effects.4
However, the benefits of sclerotherapy alone in the treatment of LM often come gradually, and radiofrequency ablation may need to be utilized to achieve more immediate results.2 Overall, outcomes are highly variable, but favorable outcomes often can be difficult to obtain due to a high recurrence rate.2,8 Our patient's symptoms improved during his postoperative recovery, and he declined further intervention.
- Elluru RG, Balakrishnan K, Padua HM. Lymphatic malformations: diagnosis and management. Semin Pediatr Surg. 2014;23:178-185. doi:10.1053/j.sempedsurg.2014.07.002
- Niti K, Manish P. Microcystic lymphatic malformation (lymphangioma circumscriptum) treated using a minimally invasive technique of radiofrequency ablation and sclerotherapy. Dermatol Surg. 2010;36:1711-1717. doi:10.1111/j.1524-4725.2010.01723.x
- Patel GA, Schwartz RA. Cutaneous lymphangioma circumscriptum: frog spawn on the skin. Int J Dermatol. 2009;48:1290-1295. doi:10.1111 /j.1365-4632.2009.04226.x
- Bikowski JB, Dumont AM. Lymphangioma circumscriptum: treatment with hypertonic saline sclerotherapy. J Am Acad Dermatol. 2005;53:442-444. doi:10.1016/j.jaad.2005.04.086
- Costa-Silva M, Fernandes I, Rodrigues AG, et al. Anogenital warts in pediatric population. An Bras Dermatol. 2017;92:675-681. doi:10.1590 /abd1806-4841.201756411
- Darmstadt GL. Perianal lymphangioma circumscriptum mistaken for genital warts. Pediatrics 1996;98;461.
- Bruins FG, van Deudekom FJA, de Vries HJC. Syphilitic condylomata lata mimicking anogenital warts. BMJ. 2015;350:h1259. doi:10.1136 /bmj.h1259
- Asch S, Sugarman JL. Epidermal nevus syndromes: new insights into whorls and swirls. Pediatr Dermatol. 2018;35:21-29. doi:10.1111 /pde.13273
The Diagnosis: Microcystic Lymphatic Malformation
The shave biopsy demonstrated numerous thin-walled vascular spaces filled with lymphatic fluid within the dermis (Figure), consistent with a diagnosis of microcystic lymphatic malformation (LM). Lymphatic malformations represent a class of benign vascular lesions consisting of anomalous or dilated lymphatic vessels, which can be broadly categorized as macrocystic (formerly cavernous lymphangioma or cystic hygroma), microcystic (formerly lymphangioma circumscriptum), or mixed.1 Patients often will present with pruritus, crusting, secondary infection, edema, or oozing.2 The superficial blebs of microcystic LMs resemble frog spawn and range in color from clear to pink, brawny, or deep maroon.3 Although the lymphatic vessels involved in microcystic LMs appear disconnected from the major lymphatic circulation,3 systemic fluid overload could plausibly promote lesional swelling and tenderness; we attributed our patient's worsening symptoms to the cumulative 7.8 L of intravenous fluid he received intraoperatively during his cardiac transplant. The excess fluid allowed communication between lymphatic cisterns and thin-walled vesicles on the skin surface through dilated channels. Overall, LMs represent roughly 26% of pediatric benign vascular tumors and approximately 4% of all vascular tumors.4
Although microcystic LMs may appear especially vascular or verrucous, the differential diagnosis for our patient's LM included condyloma acuminatum,5,6 condyloma lata,7 epidermal nevus, and lymphangiosarcoma. Epidermal nevi are congenital lesions, varying in appearance from velvety to verrucous patches and plaques that often evolve during puberty and become thicker, more verrucous, and hyperpigmented. Keratinocytic epidermal nevus syndromes and other entities such as nevus sebaceous have been associated with somatic mutations affecting proteins in the fibroblast growth factor receptor signaling pathway (eg, FGFR3, HRAS).8 Although the clinical appearance alone may be similar, lymphangiosarcoma can be distinguished from LM via biopsy.
There are several methods to diagnose LM. Duplex sonography is possibly the best noninvasive method to identify the flow between venous valves. Magnetic resonance imaging can detect larger occurrences of LM, and lymphangiography can be utilized to confirm a normal or abnormal lymphatic network.4 Treatment options are broad, including surgical excision, laser ablation, and topical sirolimus. Hypertonic saline sclerotherapy can be injected into the afflicted lymphatic channels to decrease inflammation, erythema, and hyperpigmentation without further treatment or major side effects.4
However, the benefits of sclerotherapy alone in the treatment of LM often come gradually, and radiofrequency ablation may need to be utilized to achieve more immediate results.2 Overall, outcomes are highly variable, but favorable outcomes often can be difficult to obtain due to a high recurrence rate.2,8 Our patient's symptoms improved during his postoperative recovery, and he declined further intervention.
The Diagnosis: Microcystic Lymphatic Malformation
The shave biopsy demonstrated numerous thin-walled vascular spaces filled with lymphatic fluid within the dermis (Figure), consistent with a diagnosis of microcystic lymphatic malformation (LM). Lymphatic malformations represent a class of benign vascular lesions consisting of anomalous or dilated lymphatic vessels, which can be broadly categorized as macrocystic (formerly cavernous lymphangioma or cystic hygroma), microcystic (formerly lymphangioma circumscriptum), or mixed.1 Patients often will present with pruritus, crusting, secondary infection, edema, or oozing.2 The superficial blebs of microcystic LMs resemble frog spawn and range in color from clear to pink, brawny, or deep maroon.3 Although the lymphatic vessels involved in microcystic LMs appear disconnected from the major lymphatic circulation,3 systemic fluid overload could plausibly promote lesional swelling and tenderness; we attributed our patient's worsening symptoms to the cumulative 7.8 L of intravenous fluid he received intraoperatively during his cardiac transplant. The excess fluid allowed communication between lymphatic cisterns and thin-walled vesicles on the skin surface through dilated channels. Overall, LMs represent roughly 26% of pediatric benign vascular tumors and approximately 4% of all vascular tumors.4
Although microcystic LMs may appear especially vascular or verrucous, the differential diagnosis for our patient's LM included condyloma acuminatum,5,6 condyloma lata,7 epidermal nevus, and lymphangiosarcoma. Epidermal nevi are congenital lesions, varying in appearance from velvety to verrucous patches and plaques that often evolve during puberty and become thicker, more verrucous, and hyperpigmented. Keratinocytic epidermal nevus syndromes and other entities such as nevus sebaceous have been associated with somatic mutations affecting proteins in the fibroblast growth factor receptor signaling pathway (eg, FGFR3, HRAS).8 Although the clinical appearance alone may be similar, lymphangiosarcoma can be distinguished from LM via biopsy.
There are several methods to diagnose LM. Duplex sonography is possibly the best noninvasive method to identify the flow between venous valves. Magnetic resonance imaging can detect larger occurrences of LM, and lymphangiography can be utilized to confirm a normal or abnormal lymphatic network.4 Treatment options are broad, including surgical excision, laser ablation, and topical sirolimus. Hypertonic saline sclerotherapy can be injected into the afflicted lymphatic channels to decrease inflammation, erythema, and hyperpigmentation without further treatment or major side effects.4
However, the benefits of sclerotherapy alone in the treatment of LM often come gradually, and radiofrequency ablation may need to be utilized to achieve more immediate results.2 Overall, outcomes are highly variable, but favorable outcomes often can be difficult to obtain due to a high recurrence rate.2,8 Our patient's symptoms improved during his postoperative recovery, and he declined further intervention.
- Elluru RG, Balakrishnan K, Padua HM. Lymphatic malformations: diagnosis and management. Semin Pediatr Surg. 2014;23:178-185. doi:10.1053/j.sempedsurg.2014.07.002
- Niti K, Manish P. Microcystic lymphatic malformation (lymphangioma circumscriptum) treated using a minimally invasive technique of radiofrequency ablation and sclerotherapy. Dermatol Surg. 2010;36:1711-1717. doi:10.1111/j.1524-4725.2010.01723.x
- Patel GA, Schwartz RA. Cutaneous lymphangioma circumscriptum: frog spawn on the skin. Int J Dermatol. 2009;48:1290-1295. doi:10.1111 /j.1365-4632.2009.04226.x
- Bikowski JB, Dumont AM. Lymphangioma circumscriptum: treatment with hypertonic saline sclerotherapy. J Am Acad Dermatol. 2005;53:442-444. doi:10.1016/j.jaad.2005.04.086
- Costa-Silva M, Fernandes I, Rodrigues AG, et al. Anogenital warts in pediatric population. An Bras Dermatol. 2017;92:675-681. doi:10.1590 /abd1806-4841.201756411
- Darmstadt GL. Perianal lymphangioma circumscriptum mistaken for genital warts. Pediatrics 1996;98;461.
- Bruins FG, van Deudekom FJA, de Vries HJC. Syphilitic condylomata lata mimicking anogenital warts. BMJ. 2015;350:h1259. doi:10.1136 /bmj.h1259
- Asch S, Sugarman JL. Epidermal nevus syndromes: new insights into whorls and swirls. Pediatr Dermatol. 2018;35:21-29. doi:10.1111 /pde.13273
- Elluru RG, Balakrishnan K, Padua HM. Lymphatic malformations: diagnosis and management. Semin Pediatr Surg. 2014;23:178-185. doi:10.1053/j.sempedsurg.2014.07.002
- Niti K, Manish P. Microcystic lymphatic malformation (lymphangioma circumscriptum) treated using a minimally invasive technique of radiofrequency ablation and sclerotherapy. Dermatol Surg. 2010;36:1711-1717. doi:10.1111/j.1524-4725.2010.01723.x
- Patel GA, Schwartz RA. Cutaneous lymphangioma circumscriptum: frog spawn on the skin. Int J Dermatol. 2009;48:1290-1295. doi:10.1111 /j.1365-4632.2009.04226.x
- Bikowski JB, Dumont AM. Lymphangioma circumscriptum: treatment with hypertonic saline sclerotherapy. J Am Acad Dermatol. 2005;53:442-444. doi:10.1016/j.jaad.2005.04.086
- Costa-Silva M, Fernandes I, Rodrigues AG, et al. Anogenital warts in pediatric population. An Bras Dermatol. 2017;92:675-681. doi:10.1590 /abd1806-4841.201756411
- Darmstadt GL. Perianal lymphangioma circumscriptum mistaken for genital warts. Pediatrics 1996;98;461.
- Bruins FG, van Deudekom FJA, de Vries HJC. Syphilitic condylomata lata mimicking anogenital warts. BMJ. 2015;350:h1259. doi:10.1136 /bmj.h1259
- Asch S, Sugarman JL. Epidermal nevus syndromes: new insights into whorls and swirls. Pediatr Dermatol. 2018;35:21-29. doi:10.1111 /pde.13273
A 17-year-old adolescent boy presented with increasingly painful genital warts on the right thigh, groin, and scrotum that had been present since birth. The patient had a medical history of cardiac transplantation in the months prior to presentation and was on immunosuppressive therapy. The lesions had become more swollen and bothersome in the weeks following the transplantation and now prevented him from ambulating due to discomfort. He denied any history of sexual contact or oral lesions. Physical examination revealed numerous translucent and hemorrhagic vesicles clustered and linearly distributed on the right medial thigh. A shave biopsy of a vesicle was performed.
A Severe Presentation of Plasma Cell Cheilitis
Plasma cell cheilitis (PCC), also known as plasmocytosis circumorificialis and plasmocytosis mucosae,1 is a poorly understood, uncommon inflammatory condition characterized by dense infiltration of mature plasma cells in the mucosal dermis of the lip.2-5 The etiology of PCC is unknown but is thought to be a reactive immune process triggered by infection, mechanical friction, trauma, or solar damage.1,5,6
The most common presentation of PCC is a slowly evolving, red-brown patch or plaque on the lower lip in older individuals.2,3,5,7 Secondary changes with disease progression can include erosion, ulceration, fissures, edema, bleeding, or crusting.5 The diagnosis of PCC is challenging because it can mimic neoplastic, infectious, and inflammatory conditions.8,9
Treatment strategies for PCC described in the literature vary, as does therapeutic response. Resolution of PCC has been documented after systemic steroids, intralesional steroids, systemic griseofulvin, and topical calcineurin inhibitors, among other agents.6,7,10-16
We present the case of a patient with a lip lesion who ultimately was diagnosed with PCC after it progressed to an advanced necrotic stage.
Case Report
An 80-year-old male veteran of the Armed Services initially presented to our institution via teledermatology with redness and crusting of the lower lip (Figure 1). He had a history of myelodysplastic syndrome and anemia requiring iron transfusion. The process appeared to be consistent with actinic cheilitis vs squamous cell carcinoma. In-person dermatology consultation was recommended; however, the patient did not follow through with that appointment.
Five months later, additional photographs of the lesion were taken by the patient's primary care physician and sent through teledermatology, revealing progression to an erythematous, yellow-crusted erosion (Figure 2). The medical record indicated that a punch biopsy performed by the patient’s primary care physician showed hyperkeratosis and fungal organisms on periodic acid–Schiff staining. He subsequently applied ketoconazole and terbinafine cream to the lower lip without improvement. Prompt in-person evaluation by dermatology was again recommended.
Ten days later, the patient was seen in our dermatology clinic, at which point his condition had rapidly progressed. The lower lip displayed a 3.0×2.5-cm, yellow and black, crusted, ulcerated plaque (Figure 3). He reported severe burning and pain of the lip as well as spontaneous bleeding. He had lost approximately 10 pounds over the last month due to poor oral intake. A second punch biopsy showed benign mucosa with extensive ulceration and formation of full-thickness granulation tissue. No fungi or bacteria were identified.
Consultation and Histologic Analysis
Dermatopathology was consulted and recommended a third punch biopsy for additional testing. A repeat biopsy demonstrated ulceration with lateral elements of retained epidermis and a dense submucosal chronic inflammatory infiltrate comprising plasma cells and lymphocytes (Figures 4 and 5). Immunohistochemical staining demonstrated a mixed inflammatory infiltrate with CD3+ T cells and CD20+ B cells. In situ hybridization studies demonstrated numerous lambda-positive and kappa-positive plasma cells without chain restriction. Periodic acid–Schiff with diastase and Grocott-Gomori methenamine-silver staining demonstrated no fungi. Findings were interpreted to be most consistent with a diagnosis of PCC.
Treatment and Follow-up
The patient was treated with clobetasol ointment 0.05% twice daily for 6 weeks and topical lidocaine as needed for pain. At 6-week follow-up, he displayed substantial improvement, with normal-appearing lips and complete resolution of symptoms.
Comment
The diagnosis and management of PCC is difficult because the condition is uncommon (though its true incidence is unknown) and the presentation is nonspecific, invoking a wide differential diagnosis. In the literature, PCC presents as a slowly progressive, red-brown patch or plaque on the lower lip in older individuals.2,3,5,7 The lesion can progress to become eroded, ulcerated, fissured, or edematous.5
Differential Diagnosis
The clinical differential diagnosis of PCC is broad and includes inflammatory, infectious, and neoplastic causes, such as actinic cheilitis, allergic contact cheilitis, exfoliative cheilitis, granulomatous cheilitis, lichen planus, candidiasis, syphilis, and squamous cell carcinoma of the lip.7,9 The histologic differential diagnosis includes allergic contact cheilitis, secondary syphilis, actinic cheilitis, squamous cell carcinoma, cheilitis granulomatosa, and plasmacytoma.17-19
Histopathology
On biopsy, PCC usually is characterized by plasma cells in a bandlike pattern in the upper submucosa or even more diffusely throughout the submucosa.20 In earlier studies, polyclonality of plasma cells with kappa and lambda light chains has been demonstrated5; in this case, such polyclonality militated against a plasma cell dyscrasia. There have been reports of a various number of eosinophils in PCC,5,20 but eosinophils were not a prominent feature in our case.
Treatment
As reported in the literature, treatment of PCC has been attempted using a broad range of strategies; however, the optimal regimen has yet to be elucidated.15 Numerous therapies, including excision, radiation, electrocauterization, cryotherapy, steroids, systemic griseofulvin, topical fusidic acid, and topical calcineurin inhibitors, have yielded variable success.6,7,10-16
The success of topical corticosteroids, as demonstrated in our case, has been unpredictable; the reported response has ranged from complete resolution to failure.9 This variability is thought to be related to epithelial width and the degree of acanthosis, with ulcerative lesions demonstrating a superior response to topical corticosteroids.9
Conclusion
Our case highlights the challenges of diagnosing and managing PCC, especially through teledermatology. Initial photographs of the lesion (Figure 1) that were submitted demonstrated a nonspecific erosion, which was concerning for any of several infectious, inflammatory, and malignant causes. Prompt in-person evaluation was warranted; regrettably, the patient’s condition worsened rapidly in the 10 days it took for him to be seen in-person by dermatology.
Furthermore, this case necessitated 3 separate biopsies because the pathology on the first 2 biopsies initially was equivocal, demonstrating ulceration and granulation tissue. The diagnosis was finally made after a third biopsy was recommended by a dermatopathologist, who eventually identified a bandlike distribution of polyclonal plasma cells in the upper submucosa, consistent with a diagnosis of PCC. Our patient’s final disease presentation (Figure 3) was exuberant and may represent the end point of untreated PCC.
- Senol M, Ozcan A, Aydin NE, et al. Intertriginous plasmacytosis with plasmoacanthoma: report of a typical case and review of the literature. Int J Dermatol. 2008;47:265-268. doi:10.1111/j.1365-4632.2008.03385.x
- Rocha N, Mota F, Horta M, et al. Plasma cell cheilitis. J Eur Acad Dermatol Venereol. 2004;18:96-98. doi:10.1111/j.1468-3083.2004.00791.x
- Farrier JN, Perkins CS. Plasma cell cheilitis. Br J Oral Maxillofac Surg. 2008;46:679-680. doi:10.1016/j.bjoms.2008.03.009
- Baughman RD, Berger P, Pringle WM. Plasma cell cheilitis. Arch Dermatol. 1974;110:725-726.
- Lee JY, Kim KH, Hahm JE, et al. Plasma cell cheilitis: a clinicopathological and immunohistochemical study of 13 cases. Ann Dermatol. 2017;29:536-542. doi:10.5021/ad.2017.29.5.536
- da Cunha Filho RR, Tochetto LB, Tochetto BB, et al. “Angular” plasma cell cheilitis. Dermatol Online J. 2014;20:doj_21759.
- Yang JH, Lee UH, Jang SJ, et al. Plasma cell cheilitis treated with intralesional injection of corticosteroids. J Dermatol. 2005;32:987-990. doi:10.1111/j.1346-8138.2005.tb00887.x
- Solomon LW, Wein RO, Rosenwald I, et al. Plasma cell mucositis of the oral cavity: report of a case and review of the literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2008;106:853-860. doi:10.1016/j.tripleo.2008.08.016
- Dos Santos HT, Cunha JLS, Santana LAM, et al. Plasma cell cheilitis: the diagnosis of a disorder mimicking lip cancer. Autops Case Rep. 2019;9:e2018075. doi:10.4322/acr.2018.075
- Fujimura T, Furudate S, Ishibashi M, et al. Successful treatment of plasmacytosis circumorificialis with topical tacrolimus: two case reports and an immunohistochemical study. Case Rep Dermatol. 2013;5:79-83. doi:10.1159/000350184
- Tamaki K, Osada A, Tsukamoto K, et al. Treatment of plasma cell cheilitis with griseofulvin. J Am Acad Dermatol. 1994;30:789-790. doi:10.1016/s0190-9622(08)81515-0
- Choi JW, Choi M, Cho KH. Successful treatment of plasma cell cheilitis with topical calcineurin inhibitors. J Dermatol. 2009;36:669-671. doi:10.1111/j.1346-8138.2009.00733.x
- Hanami Y, Motoki Y, Yamamoto T. Successful treatment of plasma cell cheilitis with topical tacrolimus: report of two cases. Dermatol Online J. 2011;17:6.
- Jin SP, Cho KH, Huh CH. Plasma cell cheilitis, successfully treated with topical 0.03% tacrolimus ointment. J Dermatolog Treat. 2010;21:130-132. doi:10.1080/09546630903200620
- Tseng JT-P, Cheng C-J, Lee W-R, et al. Plasma-cell cheilitis: successful treatment with intralesional injections of corticosteroids. Clin Exp Dermatol. 2009;34:174-177. doi:10.1111/j.1365-2230.2008.02765.x
- Yoshimura K, Nakano S, Tsuruta D, et al. Successful treatment with 308-nm monochromatic excimer light and subsequent tacrolimus 0.03% ointment in refractory plasma cell cheilitis. J Dermatol. 2013;40:471-474. doi:10.1111/1346-8138.12152
- Fujimura Y, Natsuga K, Abe R, et al. Plasma cell cheilitis extending beyond vermillion border. J Dermatol. 2015;42:935-936. doi:10.1111/1346-8138.12985
- White JW Jr, Olsen KD, Banks PM. Plasma cell orificial mucositis. report of a case and review of the literature. Arch Dermatol. 1986;122:1321-1324. doi:10.1001/archderm.122.11.1321
- Román CC, Yuste CM, Gonzalez MA, et al. Plasma cell gingivitis. Cutis. 2002;69:41-45.
- Choe HC, Park HJ, Oh ST, et al. Clinicopathologic study of 8 patients with plasma cell cheilitis. Korean J Dermatol. 2003;41:174-178.
Plasma cell cheilitis (PCC), also known as plasmocytosis circumorificialis and plasmocytosis mucosae,1 is a poorly understood, uncommon inflammatory condition characterized by dense infiltration of mature plasma cells in the mucosal dermis of the lip.2-5 The etiology of PCC is unknown but is thought to be a reactive immune process triggered by infection, mechanical friction, trauma, or solar damage.1,5,6
The most common presentation of PCC is a slowly evolving, red-brown patch or plaque on the lower lip in older individuals.2,3,5,7 Secondary changes with disease progression can include erosion, ulceration, fissures, edema, bleeding, or crusting.5 The diagnosis of PCC is challenging because it can mimic neoplastic, infectious, and inflammatory conditions.8,9
Treatment strategies for PCC described in the literature vary, as does therapeutic response. Resolution of PCC has been documented after systemic steroids, intralesional steroids, systemic griseofulvin, and topical calcineurin inhibitors, among other agents.6,7,10-16
We present the case of a patient with a lip lesion who ultimately was diagnosed with PCC after it progressed to an advanced necrotic stage.
Case Report
An 80-year-old male veteran of the Armed Services initially presented to our institution via teledermatology with redness and crusting of the lower lip (Figure 1). He had a history of myelodysplastic syndrome and anemia requiring iron transfusion. The process appeared to be consistent with actinic cheilitis vs squamous cell carcinoma. In-person dermatology consultation was recommended; however, the patient did not follow through with that appointment.
Five months later, additional photographs of the lesion were taken by the patient's primary care physician and sent through teledermatology, revealing progression to an erythematous, yellow-crusted erosion (Figure 2). The medical record indicated that a punch biopsy performed by the patient’s primary care physician showed hyperkeratosis and fungal organisms on periodic acid–Schiff staining. He subsequently applied ketoconazole and terbinafine cream to the lower lip without improvement. Prompt in-person evaluation by dermatology was again recommended.
Ten days later, the patient was seen in our dermatology clinic, at which point his condition had rapidly progressed. The lower lip displayed a 3.0×2.5-cm, yellow and black, crusted, ulcerated plaque (Figure 3). He reported severe burning and pain of the lip as well as spontaneous bleeding. He had lost approximately 10 pounds over the last month due to poor oral intake. A second punch biopsy showed benign mucosa with extensive ulceration and formation of full-thickness granulation tissue. No fungi or bacteria were identified.
Consultation and Histologic Analysis
Dermatopathology was consulted and recommended a third punch biopsy for additional testing. A repeat biopsy demonstrated ulceration with lateral elements of retained epidermis and a dense submucosal chronic inflammatory infiltrate comprising plasma cells and lymphocytes (Figures 4 and 5). Immunohistochemical staining demonstrated a mixed inflammatory infiltrate with CD3+ T cells and CD20+ B cells. In situ hybridization studies demonstrated numerous lambda-positive and kappa-positive plasma cells without chain restriction. Periodic acid–Schiff with diastase and Grocott-Gomori methenamine-silver staining demonstrated no fungi. Findings were interpreted to be most consistent with a diagnosis of PCC.
Treatment and Follow-up
The patient was treated with clobetasol ointment 0.05% twice daily for 6 weeks and topical lidocaine as needed for pain. At 6-week follow-up, he displayed substantial improvement, with normal-appearing lips and complete resolution of symptoms.
Comment
The diagnosis and management of PCC is difficult because the condition is uncommon (though its true incidence is unknown) and the presentation is nonspecific, invoking a wide differential diagnosis. In the literature, PCC presents as a slowly progressive, red-brown patch or plaque on the lower lip in older individuals.2,3,5,7 The lesion can progress to become eroded, ulcerated, fissured, or edematous.5
Differential Diagnosis
The clinical differential diagnosis of PCC is broad and includes inflammatory, infectious, and neoplastic causes, such as actinic cheilitis, allergic contact cheilitis, exfoliative cheilitis, granulomatous cheilitis, lichen planus, candidiasis, syphilis, and squamous cell carcinoma of the lip.7,9 The histologic differential diagnosis includes allergic contact cheilitis, secondary syphilis, actinic cheilitis, squamous cell carcinoma, cheilitis granulomatosa, and plasmacytoma.17-19
Histopathology
On biopsy, PCC usually is characterized by plasma cells in a bandlike pattern in the upper submucosa or even more diffusely throughout the submucosa.20 In earlier studies, polyclonality of plasma cells with kappa and lambda light chains has been demonstrated5; in this case, such polyclonality militated against a plasma cell dyscrasia. There have been reports of a various number of eosinophils in PCC,5,20 but eosinophils were not a prominent feature in our case.
Treatment
As reported in the literature, treatment of PCC has been attempted using a broad range of strategies; however, the optimal regimen has yet to be elucidated.15 Numerous therapies, including excision, radiation, electrocauterization, cryotherapy, steroids, systemic griseofulvin, topical fusidic acid, and topical calcineurin inhibitors, have yielded variable success.6,7,10-16
The success of topical corticosteroids, as demonstrated in our case, has been unpredictable; the reported response has ranged from complete resolution to failure.9 This variability is thought to be related to epithelial width and the degree of acanthosis, with ulcerative lesions demonstrating a superior response to topical corticosteroids.9
Conclusion
Our case highlights the challenges of diagnosing and managing PCC, especially through teledermatology. Initial photographs of the lesion (Figure 1) that were submitted demonstrated a nonspecific erosion, which was concerning for any of several infectious, inflammatory, and malignant causes. Prompt in-person evaluation was warranted; regrettably, the patient’s condition worsened rapidly in the 10 days it took for him to be seen in-person by dermatology.
Furthermore, this case necessitated 3 separate biopsies because the pathology on the first 2 biopsies initially was equivocal, demonstrating ulceration and granulation tissue. The diagnosis was finally made after a third biopsy was recommended by a dermatopathologist, who eventually identified a bandlike distribution of polyclonal plasma cells in the upper submucosa, consistent with a diagnosis of PCC. Our patient’s final disease presentation (Figure 3) was exuberant and may represent the end point of untreated PCC.
Plasma cell cheilitis (PCC), also known as plasmocytosis circumorificialis and plasmocytosis mucosae,1 is a poorly understood, uncommon inflammatory condition characterized by dense infiltration of mature plasma cells in the mucosal dermis of the lip.2-5 The etiology of PCC is unknown but is thought to be a reactive immune process triggered by infection, mechanical friction, trauma, or solar damage.1,5,6
The most common presentation of PCC is a slowly evolving, red-brown patch or plaque on the lower lip in older individuals.2,3,5,7 Secondary changes with disease progression can include erosion, ulceration, fissures, edema, bleeding, or crusting.5 The diagnosis of PCC is challenging because it can mimic neoplastic, infectious, and inflammatory conditions.8,9
Treatment strategies for PCC described in the literature vary, as does therapeutic response. Resolution of PCC has been documented after systemic steroids, intralesional steroids, systemic griseofulvin, and topical calcineurin inhibitors, among other agents.6,7,10-16
We present the case of a patient with a lip lesion who ultimately was diagnosed with PCC after it progressed to an advanced necrotic stage.
Case Report
An 80-year-old male veteran of the Armed Services initially presented to our institution via teledermatology with redness and crusting of the lower lip (Figure 1). He had a history of myelodysplastic syndrome and anemia requiring iron transfusion. The process appeared to be consistent with actinic cheilitis vs squamous cell carcinoma. In-person dermatology consultation was recommended; however, the patient did not follow through with that appointment.
Five months later, additional photographs of the lesion were taken by the patient's primary care physician and sent through teledermatology, revealing progression to an erythematous, yellow-crusted erosion (Figure 2). The medical record indicated that a punch biopsy performed by the patient’s primary care physician showed hyperkeratosis and fungal organisms on periodic acid–Schiff staining. He subsequently applied ketoconazole and terbinafine cream to the lower lip without improvement. Prompt in-person evaluation by dermatology was again recommended.
Ten days later, the patient was seen in our dermatology clinic, at which point his condition had rapidly progressed. The lower lip displayed a 3.0×2.5-cm, yellow and black, crusted, ulcerated plaque (Figure 3). He reported severe burning and pain of the lip as well as spontaneous bleeding. He had lost approximately 10 pounds over the last month due to poor oral intake. A second punch biopsy showed benign mucosa with extensive ulceration and formation of full-thickness granulation tissue. No fungi or bacteria were identified.
Consultation and Histologic Analysis
Dermatopathology was consulted and recommended a third punch biopsy for additional testing. A repeat biopsy demonstrated ulceration with lateral elements of retained epidermis and a dense submucosal chronic inflammatory infiltrate comprising plasma cells and lymphocytes (Figures 4 and 5). Immunohistochemical staining demonstrated a mixed inflammatory infiltrate with CD3+ T cells and CD20+ B cells. In situ hybridization studies demonstrated numerous lambda-positive and kappa-positive plasma cells without chain restriction. Periodic acid–Schiff with diastase and Grocott-Gomori methenamine-silver staining demonstrated no fungi. Findings were interpreted to be most consistent with a diagnosis of PCC.
Treatment and Follow-up
The patient was treated with clobetasol ointment 0.05% twice daily for 6 weeks and topical lidocaine as needed for pain. At 6-week follow-up, he displayed substantial improvement, with normal-appearing lips and complete resolution of symptoms.
Comment
The diagnosis and management of PCC is difficult because the condition is uncommon (though its true incidence is unknown) and the presentation is nonspecific, invoking a wide differential diagnosis. In the literature, PCC presents as a slowly progressive, red-brown patch or plaque on the lower lip in older individuals.2,3,5,7 The lesion can progress to become eroded, ulcerated, fissured, or edematous.5
Differential Diagnosis
The clinical differential diagnosis of PCC is broad and includes inflammatory, infectious, and neoplastic causes, such as actinic cheilitis, allergic contact cheilitis, exfoliative cheilitis, granulomatous cheilitis, lichen planus, candidiasis, syphilis, and squamous cell carcinoma of the lip.7,9 The histologic differential diagnosis includes allergic contact cheilitis, secondary syphilis, actinic cheilitis, squamous cell carcinoma, cheilitis granulomatosa, and plasmacytoma.17-19
Histopathology
On biopsy, PCC usually is characterized by plasma cells in a bandlike pattern in the upper submucosa or even more diffusely throughout the submucosa.20 In earlier studies, polyclonality of plasma cells with kappa and lambda light chains has been demonstrated5; in this case, such polyclonality militated against a plasma cell dyscrasia. There have been reports of a various number of eosinophils in PCC,5,20 but eosinophils were not a prominent feature in our case.
Treatment
As reported in the literature, treatment of PCC has been attempted using a broad range of strategies; however, the optimal regimen has yet to be elucidated.15 Numerous therapies, including excision, radiation, electrocauterization, cryotherapy, steroids, systemic griseofulvin, topical fusidic acid, and topical calcineurin inhibitors, have yielded variable success.6,7,10-16
The success of topical corticosteroids, as demonstrated in our case, has been unpredictable; the reported response has ranged from complete resolution to failure.9 This variability is thought to be related to epithelial width and the degree of acanthosis, with ulcerative lesions demonstrating a superior response to topical corticosteroids.9
Conclusion
Our case highlights the challenges of diagnosing and managing PCC, especially through teledermatology. Initial photographs of the lesion (Figure 1) that were submitted demonstrated a nonspecific erosion, which was concerning for any of several infectious, inflammatory, and malignant causes. Prompt in-person evaluation was warranted; regrettably, the patient’s condition worsened rapidly in the 10 days it took for him to be seen in-person by dermatology.
Furthermore, this case necessitated 3 separate biopsies because the pathology on the first 2 biopsies initially was equivocal, demonstrating ulceration and granulation tissue. The diagnosis was finally made after a third biopsy was recommended by a dermatopathologist, who eventually identified a bandlike distribution of polyclonal plasma cells in the upper submucosa, consistent with a diagnosis of PCC. Our patient’s final disease presentation (Figure 3) was exuberant and may represent the end point of untreated PCC.
- Senol M, Ozcan A, Aydin NE, et al. Intertriginous plasmacytosis with plasmoacanthoma: report of a typical case and review of the literature. Int J Dermatol. 2008;47:265-268. doi:10.1111/j.1365-4632.2008.03385.x
- Rocha N, Mota F, Horta M, et al. Plasma cell cheilitis. J Eur Acad Dermatol Venereol. 2004;18:96-98. doi:10.1111/j.1468-3083.2004.00791.x
- Farrier JN, Perkins CS. Plasma cell cheilitis. Br J Oral Maxillofac Surg. 2008;46:679-680. doi:10.1016/j.bjoms.2008.03.009
- Baughman RD, Berger P, Pringle WM. Plasma cell cheilitis. Arch Dermatol. 1974;110:725-726.
- Lee JY, Kim KH, Hahm JE, et al. Plasma cell cheilitis: a clinicopathological and immunohistochemical study of 13 cases. Ann Dermatol. 2017;29:536-542. doi:10.5021/ad.2017.29.5.536
- da Cunha Filho RR, Tochetto LB, Tochetto BB, et al. “Angular” plasma cell cheilitis. Dermatol Online J. 2014;20:doj_21759.
- Yang JH, Lee UH, Jang SJ, et al. Plasma cell cheilitis treated with intralesional injection of corticosteroids. J Dermatol. 2005;32:987-990. doi:10.1111/j.1346-8138.2005.tb00887.x
- Solomon LW, Wein RO, Rosenwald I, et al. Plasma cell mucositis of the oral cavity: report of a case and review of the literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2008;106:853-860. doi:10.1016/j.tripleo.2008.08.016
- Dos Santos HT, Cunha JLS, Santana LAM, et al. Plasma cell cheilitis: the diagnosis of a disorder mimicking lip cancer. Autops Case Rep. 2019;9:e2018075. doi:10.4322/acr.2018.075
- Fujimura T, Furudate S, Ishibashi M, et al. Successful treatment of plasmacytosis circumorificialis with topical tacrolimus: two case reports and an immunohistochemical study. Case Rep Dermatol. 2013;5:79-83. doi:10.1159/000350184
- Tamaki K, Osada A, Tsukamoto K, et al. Treatment of plasma cell cheilitis with griseofulvin. J Am Acad Dermatol. 1994;30:789-790. doi:10.1016/s0190-9622(08)81515-0
- Choi JW, Choi M, Cho KH. Successful treatment of plasma cell cheilitis with topical calcineurin inhibitors. J Dermatol. 2009;36:669-671. doi:10.1111/j.1346-8138.2009.00733.x
- Hanami Y, Motoki Y, Yamamoto T. Successful treatment of plasma cell cheilitis with topical tacrolimus: report of two cases. Dermatol Online J. 2011;17:6.
- Jin SP, Cho KH, Huh CH. Plasma cell cheilitis, successfully treated with topical 0.03% tacrolimus ointment. J Dermatolog Treat. 2010;21:130-132. doi:10.1080/09546630903200620
- Tseng JT-P, Cheng C-J, Lee W-R, et al. Plasma-cell cheilitis: successful treatment with intralesional injections of corticosteroids. Clin Exp Dermatol. 2009;34:174-177. doi:10.1111/j.1365-2230.2008.02765.x
- Yoshimura K, Nakano S, Tsuruta D, et al. Successful treatment with 308-nm monochromatic excimer light and subsequent tacrolimus 0.03% ointment in refractory plasma cell cheilitis. J Dermatol. 2013;40:471-474. doi:10.1111/1346-8138.12152
- Fujimura Y, Natsuga K, Abe R, et al. Plasma cell cheilitis extending beyond vermillion border. J Dermatol. 2015;42:935-936. doi:10.1111/1346-8138.12985
- White JW Jr, Olsen KD, Banks PM. Plasma cell orificial mucositis. report of a case and review of the literature. Arch Dermatol. 1986;122:1321-1324. doi:10.1001/archderm.122.11.1321
- Román CC, Yuste CM, Gonzalez MA, et al. Plasma cell gingivitis. Cutis. 2002;69:41-45.
- Choe HC, Park HJ, Oh ST, et al. Clinicopathologic study of 8 patients with plasma cell cheilitis. Korean J Dermatol. 2003;41:174-178.
- Senol M, Ozcan A, Aydin NE, et al. Intertriginous plasmacytosis with plasmoacanthoma: report of a typical case and review of the literature. Int J Dermatol. 2008;47:265-268. doi:10.1111/j.1365-4632.2008.03385.x
- Rocha N, Mota F, Horta M, et al. Plasma cell cheilitis. J Eur Acad Dermatol Venereol. 2004;18:96-98. doi:10.1111/j.1468-3083.2004.00791.x
- Farrier JN, Perkins CS. Plasma cell cheilitis. Br J Oral Maxillofac Surg. 2008;46:679-680. doi:10.1016/j.bjoms.2008.03.009
- Baughman RD, Berger P, Pringle WM. Plasma cell cheilitis. Arch Dermatol. 1974;110:725-726.
- Lee JY, Kim KH, Hahm JE, et al. Plasma cell cheilitis: a clinicopathological and immunohistochemical study of 13 cases. Ann Dermatol. 2017;29:536-542. doi:10.5021/ad.2017.29.5.536
- da Cunha Filho RR, Tochetto LB, Tochetto BB, et al. “Angular” plasma cell cheilitis. Dermatol Online J. 2014;20:doj_21759.
- Yang JH, Lee UH, Jang SJ, et al. Plasma cell cheilitis treated with intralesional injection of corticosteroids. J Dermatol. 2005;32:987-990. doi:10.1111/j.1346-8138.2005.tb00887.x
- Solomon LW, Wein RO, Rosenwald I, et al. Plasma cell mucositis of the oral cavity: report of a case and review of the literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2008;106:853-860. doi:10.1016/j.tripleo.2008.08.016
- Dos Santos HT, Cunha JLS, Santana LAM, et al. Plasma cell cheilitis: the diagnosis of a disorder mimicking lip cancer. Autops Case Rep. 2019;9:e2018075. doi:10.4322/acr.2018.075
- Fujimura T, Furudate S, Ishibashi M, et al. Successful treatment of plasmacytosis circumorificialis with topical tacrolimus: two case reports and an immunohistochemical study. Case Rep Dermatol. 2013;5:79-83. doi:10.1159/000350184
- Tamaki K, Osada A, Tsukamoto K, et al. Treatment of plasma cell cheilitis with griseofulvin. J Am Acad Dermatol. 1994;30:789-790. doi:10.1016/s0190-9622(08)81515-0
- Choi JW, Choi M, Cho KH. Successful treatment of plasma cell cheilitis with topical calcineurin inhibitors. J Dermatol. 2009;36:669-671. doi:10.1111/j.1346-8138.2009.00733.x
- Hanami Y, Motoki Y, Yamamoto T. Successful treatment of plasma cell cheilitis with topical tacrolimus: report of two cases. Dermatol Online J. 2011;17:6.
- Jin SP, Cho KH, Huh CH. Plasma cell cheilitis, successfully treated with topical 0.03% tacrolimus ointment. J Dermatolog Treat. 2010;21:130-132. doi:10.1080/09546630903200620
- Tseng JT-P, Cheng C-J, Lee W-R, et al. Plasma-cell cheilitis: successful treatment with intralesional injections of corticosteroids. Clin Exp Dermatol. 2009;34:174-177. doi:10.1111/j.1365-2230.2008.02765.x
- Yoshimura K, Nakano S, Tsuruta D, et al. Successful treatment with 308-nm monochromatic excimer light and subsequent tacrolimus 0.03% ointment in refractory plasma cell cheilitis. J Dermatol. 2013;40:471-474. doi:10.1111/1346-8138.12152
- Fujimura Y, Natsuga K, Abe R, et al. Plasma cell cheilitis extending beyond vermillion border. J Dermatol. 2015;42:935-936. doi:10.1111/1346-8138.12985
- White JW Jr, Olsen KD, Banks PM. Plasma cell orificial mucositis. report of a case and review of the literature. Arch Dermatol. 1986;122:1321-1324. doi:10.1001/archderm.122.11.1321
- Román CC, Yuste CM, Gonzalez MA, et al. Plasma cell gingivitis. Cutis. 2002;69:41-45.
- Choe HC, Park HJ, Oh ST, et al. Clinicopathologic study of 8 patients with plasma cell cheilitis. Korean J Dermatol. 2003;41:174-178.
PRACTICE POINTS
- Plasma cell cheilitis (PCC) is a benign condition that affects the lower lip in older individuals, presenting as a nonspecific, red-brown patch or plaque that can progress slowly to erosions and edema.
- Our patient with PCC experienced full resolution of symptoms with application of a class I topical corticosteroid.
Verruca Vulgaris Arising Within the Red Portion of a Multicolored Tattoo
To the Editor:
The art of tattooing continues to gain popularity in the 21st century, albeit with accompanying hazards.1 Reported adverse reactions to tattoos include infections, tumors, and hypersensitivity and granulomatous reactions.2 Various infectious agents may involve tattoos, including human papillomavirus (HPV), molluscum contagiosum, herpes simplex virus, hepatitis C virus, tuberculoid and nontuberculoid mycobacteria, and Staphylococcus aureus.2 Verruca vulgaris infrequently has been reported to develop in tattoos.3,4 Previously reported cases of verruca in tattoos suggest a predilection for blue or black pigment.1-5 We report a case of verruca vulgaris occurring within the red-inked areas of a tattoo that first appeared approximately 18 years after the initial tattoo placement.
A 44-year-old woman presented with erythema, induration, and irritation of a tattoo on the left leg of 2 years’ duration. The tattoo initially was inscribed more than 20 years prior. The patient had a history of type 2 diabetes mellitus and chronic obstructive pulmonary disease. She reported no prior trauma to the area, prior rash or irritation, or similar changes to her other tattoos, including those with red ink. The affected tattoo was inscribed at a separate time from the other tattoos. Physical examination of the irritated tattoo revealed hyperkeratotic papules with firm scaling in the zone of dermal red pigment (Figure 1). Notable nodularity or deep induration was not present. The clinical differential diagnosis included a hypersensitivity reaction to red tattoo ink, sarcoidosis, and an infectious process, such as an atypical mycobacterial infection. A punch biopsy demonstrated papillomatous epidermal hyperplasia with hyperkeratosis, focal parakeratosis, and frequent vacuolization of keratinocytes with enlarged keratohyalin granules, diagnostic of verruca vulgaris (Figure 2). Of note, the patient did not have clinically apparent viral warts elsewhere on physical examination. The patient was successfully managedwith a combination of 2 treatments of intralesional Candida antigen and 3 treatments of cryotherapy with resolution of most lesions over the course of 8 months. Over the following several months, the patient applied topical salicylic acid, which led to the resolution of the remaining lesions. The verrucae had not recurred 19 months after the initial presentation.
The development of verruca vulgaris within a tattoo may occur secondary to various mechanisms of HPV inoculation, including introduction of the virus through contaminated ink, the tattoo artist’s saliva, autoinoculation, or koebnerization of a pre-existing verruca vulgaris.4 Local immune system dysregulation secondary to tattoo ink also has been proposed as a mechanism for HPV infection in this setting.1,5 The contents of darker tattoo pigments may promote formation of reactive oxygen species inducing local immunocompromise.5
The pathogenic mechanism was elusive in our patient. Although the localization of verruca vulgaris to the zones of red pigment may be merely coincidental, this phenomenon raised suspicion for direct inoculation via contaminated red ink. The patient’s other red ink–containing tattoos that were inscribed separately were spared, compatible with contamination of the red ink used for the affected tattoo. However, the delayed onset of nearly 2 decades was exceptional, given the shorter previously reported latencies ranging from months to 10 years.4 Autoinoculation or koebnerization is plausible, though greater involvement of nonred pigments would be expected as well as a briefer latency. Finally, the possibility of local immune dysregulation seemed feasible, given the slow evolution of the lesions largely restricted to one pigment type.
We report a case of verruca vulgaris within the red area of a multicolored tattoo that occurred approximately 18 years after tattoo placement. This case highlights a rare presentation of an infectious agent that may complicate tattoos. Both predilection for red pigment rather than black or blue pigment and the long latency period raised interesting questions regarding pathogenesis. Confirmatory biopsy enables effective management of this tattoo complication.
- Huynh TN, Jackson JD, Brodell RT. Tattoo and vaccination sites: possible nest for opportunistic infections, tumors, and dysimmune reactions. Clin Dermatol. 2014;32:678-684.
- Wenzel SM, Rittmann I, Landthaler M, et al. Adverse reactions after tattooing: review of the literature and comparison to results of a survey. Dermatology. 2013;226:138-147.
- Trefzer U, Schmollack K, Stockfleth E, et al. Verrucae in a multicolored decorative tattoo. J Am Acad Dermatol. 2004;50:478-479.
- Wanat KA, Tyring S, Rady P, et al. Human papillomavirus type 27 associated with multiple verruca within a tattoo: report of a case and review of the literature. Int J Dermatol. 2014;53:882-884.
- Ramey K, Ibrahim J, Brodell RT. Verruca localization predominately in black tattoo ink: a retrospective case series. J Eur Acad Dermatol Venereol. 2016;30:E34-E36.
To the Editor:
The art of tattooing continues to gain popularity in the 21st century, albeit with accompanying hazards.1 Reported adverse reactions to tattoos include infections, tumors, and hypersensitivity and granulomatous reactions.2 Various infectious agents may involve tattoos, including human papillomavirus (HPV), molluscum contagiosum, herpes simplex virus, hepatitis C virus, tuberculoid and nontuberculoid mycobacteria, and Staphylococcus aureus.2 Verruca vulgaris infrequently has been reported to develop in tattoos.3,4 Previously reported cases of verruca in tattoos suggest a predilection for blue or black pigment.1-5 We report a case of verruca vulgaris occurring within the red-inked areas of a tattoo that first appeared approximately 18 years after the initial tattoo placement.
A 44-year-old woman presented with erythema, induration, and irritation of a tattoo on the left leg of 2 years’ duration. The tattoo initially was inscribed more than 20 years prior. The patient had a history of type 2 diabetes mellitus and chronic obstructive pulmonary disease. She reported no prior trauma to the area, prior rash or irritation, or similar changes to her other tattoos, including those with red ink. The affected tattoo was inscribed at a separate time from the other tattoos. Physical examination of the irritated tattoo revealed hyperkeratotic papules with firm scaling in the zone of dermal red pigment (Figure 1). Notable nodularity or deep induration was not present. The clinical differential diagnosis included a hypersensitivity reaction to red tattoo ink, sarcoidosis, and an infectious process, such as an atypical mycobacterial infection. A punch biopsy demonstrated papillomatous epidermal hyperplasia with hyperkeratosis, focal parakeratosis, and frequent vacuolization of keratinocytes with enlarged keratohyalin granules, diagnostic of verruca vulgaris (Figure 2). Of note, the patient did not have clinically apparent viral warts elsewhere on physical examination. The patient was successfully managedwith a combination of 2 treatments of intralesional Candida antigen and 3 treatments of cryotherapy with resolution of most lesions over the course of 8 months. Over the following several months, the patient applied topical salicylic acid, which led to the resolution of the remaining lesions. The verrucae had not recurred 19 months after the initial presentation.
The development of verruca vulgaris within a tattoo may occur secondary to various mechanisms of HPV inoculation, including introduction of the virus through contaminated ink, the tattoo artist’s saliva, autoinoculation, or koebnerization of a pre-existing verruca vulgaris.4 Local immune system dysregulation secondary to tattoo ink also has been proposed as a mechanism for HPV infection in this setting.1,5 The contents of darker tattoo pigments may promote formation of reactive oxygen species inducing local immunocompromise.5
The pathogenic mechanism was elusive in our patient. Although the localization of verruca vulgaris to the zones of red pigment may be merely coincidental, this phenomenon raised suspicion for direct inoculation via contaminated red ink. The patient’s other red ink–containing tattoos that were inscribed separately were spared, compatible with contamination of the red ink used for the affected tattoo. However, the delayed onset of nearly 2 decades was exceptional, given the shorter previously reported latencies ranging from months to 10 years.4 Autoinoculation or koebnerization is plausible, though greater involvement of nonred pigments would be expected as well as a briefer latency. Finally, the possibility of local immune dysregulation seemed feasible, given the slow evolution of the lesions largely restricted to one pigment type.
We report a case of verruca vulgaris within the red area of a multicolored tattoo that occurred approximately 18 years after tattoo placement. This case highlights a rare presentation of an infectious agent that may complicate tattoos. Both predilection for red pigment rather than black or blue pigment and the long latency period raised interesting questions regarding pathogenesis. Confirmatory biopsy enables effective management of this tattoo complication.
To the Editor:
The art of tattooing continues to gain popularity in the 21st century, albeit with accompanying hazards.1 Reported adverse reactions to tattoos include infections, tumors, and hypersensitivity and granulomatous reactions.2 Various infectious agents may involve tattoos, including human papillomavirus (HPV), molluscum contagiosum, herpes simplex virus, hepatitis C virus, tuberculoid and nontuberculoid mycobacteria, and Staphylococcus aureus.2 Verruca vulgaris infrequently has been reported to develop in tattoos.3,4 Previously reported cases of verruca in tattoos suggest a predilection for blue or black pigment.1-5 We report a case of verruca vulgaris occurring within the red-inked areas of a tattoo that first appeared approximately 18 years after the initial tattoo placement.
A 44-year-old woman presented with erythema, induration, and irritation of a tattoo on the left leg of 2 years’ duration. The tattoo initially was inscribed more than 20 years prior. The patient had a history of type 2 diabetes mellitus and chronic obstructive pulmonary disease. She reported no prior trauma to the area, prior rash or irritation, or similar changes to her other tattoos, including those with red ink. The affected tattoo was inscribed at a separate time from the other tattoos. Physical examination of the irritated tattoo revealed hyperkeratotic papules with firm scaling in the zone of dermal red pigment (Figure 1). Notable nodularity or deep induration was not present. The clinical differential diagnosis included a hypersensitivity reaction to red tattoo ink, sarcoidosis, and an infectious process, such as an atypical mycobacterial infection. A punch biopsy demonstrated papillomatous epidermal hyperplasia with hyperkeratosis, focal parakeratosis, and frequent vacuolization of keratinocytes with enlarged keratohyalin granules, diagnostic of verruca vulgaris (Figure 2). Of note, the patient did not have clinically apparent viral warts elsewhere on physical examination. The patient was successfully managedwith a combination of 2 treatments of intralesional Candida antigen and 3 treatments of cryotherapy with resolution of most lesions over the course of 8 months. Over the following several months, the patient applied topical salicylic acid, which led to the resolution of the remaining lesions. The verrucae had not recurred 19 months after the initial presentation.
The development of verruca vulgaris within a tattoo may occur secondary to various mechanisms of HPV inoculation, including introduction of the virus through contaminated ink, the tattoo artist’s saliva, autoinoculation, or koebnerization of a pre-existing verruca vulgaris.4 Local immune system dysregulation secondary to tattoo ink also has been proposed as a mechanism for HPV infection in this setting.1,5 The contents of darker tattoo pigments may promote formation of reactive oxygen species inducing local immunocompromise.5
The pathogenic mechanism was elusive in our patient. Although the localization of verruca vulgaris to the zones of red pigment may be merely coincidental, this phenomenon raised suspicion for direct inoculation via contaminated red ink. The patient’s other red ink–containing tattoos that were inscribed separately were spared, compatible with contamination of the red ink used for the affected tattoo. However, the delayed onset of nearly 2 decades was exceptional, given the shorter previously reported latencies ranging from months to 10 years.4 Autoinoculation or koebnerization is plausible, though greater involvement of nonred pigments would be expected as well as a briefer latency. Finally, the possibility of local immune dysregulation seemed feasible, given the slow evolution of the lesions largely restricted to one pigment type.
We report a case of verruca vulgaris within the red area of a multicolored tattoo that occurred approximately 18 years after tattoo placement. This case highlights a rare presentation of an infectious agent that may complicate tattoos. Both predilection for red pigment rather than black or blue pigment and the long latency period raised interesting questions regarding pathogenesis. Confirmatory biopsy enables effective management of this tattoo complication.
- Huynh TN, Jackson JD, Brodell RT. Tattoo and vaccination sites: possible nest for opportunistic infections, tumors, and dysimmune reactions. Clin Dermatol. 2014;32:678-684.
- Wenzel SM, Rittmann I, Landthaler M, et al. Adverse reactions after tattooing: review of the literature and comparison to results of a survey. Dermatology. 2013;226:138-147.
- Trefzer U, Schmollack K, Stockfleth E, et al. Verrucae in a multicolored decorative tattoo. J Am Acad Dermatol. 2004;50:478-479.
- Wanat KA, Tyring S, Rady P, et al. Human papillomavirus type 27 associated with multiple verruca within a tattoo: report of a case and review of the literature. Int J Dermatol. 2014;53:882-884.
- Ramey K, Ibrahim J, Brodell RT. Verruca localization predominately in black tattoo ink: a retrospective case series. J Eur Acad Dermatol Venereol. 2016;30:E34-E36.
- Huynh TN, Jackson JD, Brodell RT. Tattoo and vaccination sites: possible nest for opportunistic infections, tumors, and dysimmune reactions. Clin Dermatol. 2014;32:678-684.
- Wenzel SM, Rittmann I, Landthaler M, et al. Adverse reactions after tattooing: review of the literature and comparison to results of a survey. Dermatology. 2013;226:138-147.
- Trefzer U, Schmollack K, Stockfleth E, et al. Verrucae in a multicolored decorative tattoo. J Am Acad Dermatol. 2004;50:478-479.
- Wanat KA, Tyring S, Rady P, et al. Human papillomavirus type 27 associated with multiple verruca within a tattoo: report of a case and review of the literature. Int J Dermatol. 2014;53:882-884.
- Ramey K, Ibrahim J, Brodell RT. Verruca localization predominately in black tattoo ink: a retrospective case series. J Eur Acad Dermatol Venereol. 2016;30:E34-E36.
Practice Points
- Various adverse reactions and infectious agents may involve tattoos.
- Verruca vulgaris may affect tattoos in a color-restricted manner and demonstrate latency of many years after tattoo placement.
- Timely diagnosis of the tattoo-involving process, confirmed by biopsy, allows for appropriate management.
Pediatric-Onset Refractory Lupus Erythematosus Panniculitis Treated With Rituximab
To the Editor:
Lupus erythematosus panniculitis (LEP) is rare in the pediatric population. It can be difficult to manage, as patients may not respond to conventional treatments including hydroxychloroquine and prednisone. We report the use of rituximab in the treatment of a 20-year-old woman with LEP of the face, legs, and arms that was refractory to standard treatments. She also had a history of hemophagocytic lymphohistiocytosis (HLH). Further studies are warranted to determine the role of rituximab in the treatment of pediatric patients with LEP.
A 20-year-old woman with history of LEP and HLH initially presented with migratory violaceous nodules on the face 16 years prior to the current presentation. A skin biopsy 3 years after that initial presentation suggested a diagnosis of cutaneous lupus erythematosus. Six years later, numerous asymptomatic lesions appeared on the legs, predominantly on the calves; she was successfully treated with hydroxychloroquine and high-dose prednisone. Four years prior to the current presentation, a febrile illness prompted discontinuation of hydroxychloroquine and hospitalization, where she was first was diagnosed with HLH; she achieved remission with cyclosporine. At the current presentation, she continued to have persistent violaceous lesions on the face, lower arms, and legs with underlying nodularity (Figure 1). Skin biopsies revealed LEP and were less suggestive of HLH. She was restarted on hydroxychloroquine, which did not adequately control the disease. Rheumatologic workup was only notable for an antinuclear antibody titer of 1:80 (reference range, <1:80) in a speckled pattern.
Due to the refractory nature of her condition, continued lesion development despite standard treatment, and concerns of possible scarring, we considered a trial of rituximab. Because HLH and LEP can mimic subcutaneous T-cell lymphoma, another skin biopsy was performed, which revealed a deep dermal and subcutaneous lymphohistiocytic infiltrate composed of predominantly CD3+ T cells with a mixed population of CD4+ and CD8+ cells (Figure 2). There was no evidence of transformation into lymphoma. Pathologic findings were most compatible with LEP rather than an HLH-associated panniculitis due to the lack of definitive phagocytosis. She received rituximab using body surface area–based dosing at 375 mg/m2. CD19 levels decreased to undetectable levels after the first dose. Rituximab was dosed based on clinical response; she tolerated treatment well and experienced considerable improvement in the number of lesions following completion of 4 doses at weeks 0, 1, 5, and 7 (Figure 3). She developed a flare at 7 months and improved again after another dose of rituximab.
Lupus erythematosus panniculitis is a rare variant of lupus erythematosus with an average age of presentation between 30 and 60 years.1 In children, LEP presents as recurrent subcutaneous nodules and plaques, commonly involving the face and upper arms.1,2 Long-term sequelae include local swelling and skin atrophy.3 Conventional treatment options for pediatric patients include hydroxychloroquine and corticosteroids.1 Management can be challenging due to the lack of response to conventional treatments as well as the chronic progressive nature of LEP.2 In refractory cases, cyclosporine, azathioprine, sulfones, thalidomide, mycophenolate mofetil, and cyclophosphamide are alternative treatment options.1-4
Rituximab, a chimeric monoclonal antibody targeting B-cell surface marker CD20, results in depletion of mature B cells. Use of rituximab for LEP has been described in multiple case reports involving an 8-year-old boy, 22-year-old girl, and 2 middle-aged women.2-4 In addition, a recently published case series of 4 patients with childhood-onset refractory LEP described improvement of disease activity with rituximab.5 It is important to rule out subcutaneous T-cell lymphoma before treatment with rituximab, as its histopathology can closely resemble that seen in LEP and HLH-associated cytophagic histiocytic panniculitis.1,6
Rituximab may be an effective treatment option in pediatric patients with refractory LEP. Larger studies on the use of rituximab in the pediatric population are necessary.
- Weingartner JS, Zedek DC, Burkhart CN, et al. Lupus erythematosus panniculitis in children: report of three cases and review of previously reported cases. Pediatr Dermatol. 2011;29:169-176.
- Moreno-Suárez F, Pulpillo-Ruiz Á. Rituximab for the treatment of lupus erythematosus panniculitis. Dermatol Ther. 2013;26:415-418.
- Guissa VR, Trudes G, Jesus AA, et al. Lupus erythematosus panniculitis in children and adolescents. Acta Reumatol Port. 2012;37:82-85.
- Mcardle A, Baker JF. A case of “refractory” lupus erythematosus profundus responsive to rituximab. Clin Rheumatol. 2009;28:745-746.
- Correll CK, Miller DD, Maguiness SM. Treatment of childhood-onset lupus erythematosus panniculitis with rituximab. JAMA Dermatol. 2020;156:566-569.
- Aronson IK, Worobec SM. Cytophagic histiocytic panniculitis and hemophagocytic lymphohistiocytosis: an overview. Dermatol Ther. 2010;23:389-402.
To the Editor:
Lupus erythematosus panniculitis (LEP) is rare in the pediatric population. It can be difficult to manage, as patients may not respond to conventional treatments including hydroxychloroquine and prednisone. We report the use of rituximab in the treatment of a 20-year-old woman with LEP of the face, legs, and arms that was refractory to standard treatments. She also had a history of hemophagocytic lymphohistiocytosis (HLH). Further studies are warranted to determine the role of rituximab in the treatment of pediatric patients with LEP.
A 20-year-old woman with history of LEP and HLH initially presented with migratory violaceous nodules on the face 16 years prior to the current presentation. A skin biopsy 3 years after that initial presentation suggested a diagnosis of cutaneous lupus erythematosus. Six years later, numerous asymptomatic lesions appeared on the legs, predominantly on the calves; she was successfully treated with hydroxychloroquine and high-dose prednisone. Four years prior to the current presentation, a febrile illness prompted discontinuation of hydroxychloroquine and hospitalization, where she was first was diagnosed with HLH; she achieved remission with cyclosporine. At the current presentation, she continued to have persistent violaceous lesions on the face, lower arms, and legs with underlying nodularity (Figure 1). Skin biopsies revealed LEP and were less suggestive of HLH. She was restarted on hydroxychloroquine, which did not adequately control the disease. Rheumatologic workup was only notable for an antinuclear antibody titer of 1:80 (reference range, <1:80) in a speckled pattern.
Due to the refractory nature of her condition, continued lesion development despite standard treatment, and concerns of possible scarring, we considered a trial of rituximab. Because HLH and LEP can mimic subcutaneous T-cell lymphoma, another skin biopsy was performed, which revealed a deep dermal and subcutaneous lymphohistiocytic infiltrate composed of predominantly CD3+ T cells with a mixed population of CD4+ and CD8+ cells (Figure 2). There was no evidence of transformation into lymphoma. Pathologic findings were most compatible with LEP rather than an HLH-associated panniculitis due to the lack of definitive phagocytosis. She received rituximab using body surface area–based dosing at 375 mg/m2. CD19 levels decreased to undetectable levels after the first dose. Rituximab was dosed based on clinical response; she tolerated treatment well and experienced considerable improvement in the number of lesions following completion of 4 doses at weeks 0, 1, 5, and 7 (Figure 3). She developed a flare at 7 months and improved again after another dose of rituximab.
Lupus erythematosus panniculitis is a rare variant of lupus erythematosus with an average age of presentation between 30 and 60 years.1 In children, LEP presents as recurrent subcutaneous nodules and plaques, commonly involving the face and upper arms.1,2 Long-term sequelae include local swelling and skin atrophy.3 Conventional treatment options for pediatric patients include hydroxychloroquine and corticosteroids.1 Management can be challenging due to the lack of response to conventional treatments as well as the chronic progressive nature of LEP.2 In refractory cases, cyclosporine, azathioprine, sulfones, thalidomide, mycophenolate mofetil, and cyclophosphamide are alternative treatment options.1-4
Rituximab, a chimeric monoclonal antibody targeting B-cell surface marker CD20, results in depletion of mature B cells. Use of rituximab for LEP has been described in multiple case reports involving an 8-year-old boy, 22-year-old girl, and 2 middle-aged women.2-4 In addition, a recently published case series of 4 patients with childhood-onset refractory LEP described improvement of disease activity with rituximab.5 It is important to rule out subcutaneous T-cell lymphoma before treatment with rituximab, as its histopathology can closely resemble that seen in LEP and HLH-associated cytophagic histiocytic panniculitis.1,6
Rituximab may be an effective treatment option in pediatric patients with refractory LEP. Larger studies on the use of rituximab in the pediatric population are necessary.
To the Editor:
Lupus erythematosus panniculitis (LEP) is rare in the pediatric population. It can be difficult to manage, as patients may not respond to conventional treatments including hydroxychloroquine and prednisone. We report the use of rituximab in the treatment of a 20-year-old woman with LEP of the face, legs, and arms that was refractory to standard treatments. She also had a history of hemophagocytic lymphohistiocytosis (HLH). Further studies are warranted to determine the role of rituximab in the treatment of pediatric patients with LEP.
A 20-year-old woman with history of LEP and HLH initially presented with migratory violaceous nodules on the face 16 years prior to the current presentation. A skin biopsy 3 years after that initial presentation suggested a diagnosis of cutaneous lupus erythematosus. Six years later, numerous asymptomatic lesions appeared on the legs, predominantly on the calves; she was successfully treated with hydroxychloroquine and high-dose prednisone. Four years prior to the current presentation, a febrile illness prompted discontinuation of hydroxychloroquine and hospitalization, where she was first was diagnosed with HLH; she achieved remission with cyclosporine. At the current presentation, she continued to have persistent violaceous lesions on the face, lower arms, and legs with underlying nodularity (Figure 1). Skin biopsies revealed LEP and were less suggestive of HLH. She was restarted on hydroxychloroquine, which did not adequately control the disease. Rheumatologic workup was only notable for an antinuclear antibody titer of 1:80 (reference range, <1:80) in a speckled pattern.
Due to the refractory nature of her condition, continued lesion development despite standard treatment, and concerns of possible scarring, we considered a trial of rituximab. Because HLH and LEP can mimic subcutaneous T-cell lymphoma, another skin biopsy was performed, which revealed a deep dermal and subcutaneous lymphohistiocytic infiltrate composed of predominantly CD3+ T cells with a mixed population of CD4+ and CD8+ cells (Figure 2). There was no evidence of transformation into lymphoma. Pathologic findings were most compatible with LEP rather than an HLH-associated panniculitis due to the lack of definitive phagocytosis. She received rituximab using body surface area–based dosing at 375 mg/m2. CD19 levels decreased to undetectable levels after the first dose. Rituximab was dosed based on clinical response; she tolerated treatment well and experienced considerable improvement in the number of lesions following completion of 4 doses at weeks 0, 1, 5, and 7 (Figure 3). She developed a flare at 7 months and improved again after another dose of rituximab.
Lupus erythematosus panniculitis is a rare variant of lupus erythematosus with an average age of presentation between 30 and 60 years.1 In children, LEP presents as recurrent subcutaneous nodules and plaques, commonly involving the face and upper arms.1,2 Long-term sequelae include local swelling and skin atrophy.3 Conventional treatment options for pediatric patients include hydroxychloroquine and corticosteroids.1 Management can be challenging due to the lack of response to conventional treatments as well as the chronic progressive nature of LEP.2 In refractory cases, cyclosporine, azathioprine, sulfones, thalidomide, mycophenolate mofetil, and cyclophosphamide are alternative treatment options.1-4
Rituximab, a chimeric monoclonal antibody targeting B-cell surface marker CD20, results in depletion of mature B cells. Use of rituximab for LEP has been described in multiple case reports involving an 8-year-old boy, 22-year-old girl, and 2 middle-aged women.2-4 In addition, a recently published case series of 4 patients with childhood-onset refractory LEP described improvement of disease activity with rituximab.5 It is important to rule out subcutaneous T-cell lymphoma before treatment with rituximab, as its histopathology can closely resemble that seen in LEP and HLH-associated cytophagic histiocytic panniculitis.1,6
Rituximab may be an effective treatment option in pediatric patients with refractory LEP. Larger studies on the use of rituximab in the pediatric population are necessary.
- Weingartner JS, Zedek DC, Burkhart CN, et al. Lupus erythematosus panniculitis in children: report of three cases and review of previously reported cases. Pediatr Dermatol. 2011;29:169-176.
- Moreno-Suárez F, Pulpillo-Ruiz Á. Rituximab for the treatment of lupus erythematosus panniculitis. Dermatol Ther. 2013;26:415-418.
- Guissa VR, Trudes G, Jesus AA, et al. Lupus erythematosus panniculitis in children and adolescents. Acta Reumatol Port. 2012;37:82-85.
- Mcardle A, Baker JF. A case of “refractory” lupus erythematosus profundus responsive to rituximab. Clin Rheumatol. 2009;28:745-746.
- Correll CK, Miller DD, Maguiness SM. Treatment of childhood-onset lupus erythematosus panniculitis with rituximab. JAMA Dermatol. 2020;156:566-569.
- Aronson IK, Worobec SM. Cytophagic histiocytic panniculitis and hemophagocytic lymphohistiocytosis: an overview. Dermatol Ther. 2010;23:389-402.
- Weingartner JS, Zedek DC, Burkhart CN, et al. Lupus erythematosus panniculitis in children: report of three cases and review of previously reported cases. Pediatr Dermatol. 2011;29:169-176.
- Moreno-Suárez F, Pulpillo-Ruiz Á. Rituximab for the treatment of lupus erythematosus panniculitis. Dermatol Ther. 2013;26:415-418.
- Guissa VR, Trudes G, Jesus AA, et al. Lupus erythematosus panniculitis in children and adolescents. Acta Reumatol Port. 2012;37:82-85.
- Mcardle A, Baker JF. A case of “refractory” lupus erythematosus profundus responsive to rituximab. Clin Rheumatol. 2009;28:745-746.
- Correll CK, Miller DD, Maguiness SM. Treatment of childhood-onset lupus erythematosus panniculitis with rituximab. JAMA Dermatol. 2020;156:566-569.
- Aronson IK, Worobec SM. Cytophagic histiocytic panniculitis and hemophagocytic lymphohistiocytosis: an overview. Dermatol Ther. 2010;23:389-402.
Practice Points
- Lupus erythematosus panniculitis (LEP) is rare in the pediatric population and often is difficult to treat.
- Rituximab can be an effective treatment option for refractory LEP.
- Before the initiation of rituximab, a biopsy is warranted to rule out subcutaneous T-cell lymphoma, which can mimic LEP and hemophagocytic lymphohistiocytosis–associated panniculitis.
Volunteer Opportunities Within Dermatology: More than Skin Deep
The adage “so much to do, so little time” aptly describes the daily challenges facing dermatologists and dermatology residents. The time and attention required by direct patient care, writing notes, navigating electronic health records, and engaging in education and research as well as family commitments can drain even the most tireless clinician. In addition, dermatologists are expected to play a critical role in clinic and practice management to successfully curate an online presence and adapt their skills to successfully manage a teledermatology practice. Coupled with the time spent socializing with friends or colleagues and time for personal hobbies or exercise, it’s easy to see how sleep deprivation is common in many of our colleagues.
What’s being left out of these jam-packed schedules? Increasingly, it is the time and expertise dedicated to volunteering in our local communities. Two recent research letters highlighted how a dramatic increase in the number of research projects and publications is not mirrored by a similar increase in volunteer experiences as dermatology residency selection becomes more competitive.1,2
Although the rate of volunteerism among practicing dermatologists has yet to be studied, a brief review suggests a component of unmet dermatology need within our communities. It’s estimated that approximately 5% to 10% of all emergency department visits are for dermatologic concerns.3-5 In many cases, the reason for the visit is nonurgent and instead reflects a lack of other options for care. However, the need for dermatologists extends beyond the emergency department setting. A review of the prevalence of patients presenting for care to a group of regional free clinics found that 8% (N=5553) of all visitors sought care for dermatologic concerns.6 The benefit is not just for those seated on the examination table; research has shown that while many of the underlying factors resulting in physician burnout stem from systemic issues, participating in volunteer opportunities helps combat burnout in ourselves and our colleagues.7-9 Herein, opportunities that exist for dermatologists to reconnect with their communities, advocate for causes distinctive to the specialty, and care for neighbors most in need are highlighted.
Camp Wonder
Every year, children from across the United States living with chronic and debilitating skin conditions get the opportunity to join fellow campers and spend a week just being kids without the constant focus on being a patient. Camp Wonder’s founder and director, Francesca Tenconi, describes the camp as a place where kids “can form a community and can feel free to be themselves, without judgment, without stares. They get the chance to forget about their skin disease and be themselves” (oral communication, June 18, 2021). Tenconi and the camp’s cofounders and medical directors, Drs. Jenny Kim and Stefani Takahashi, envisioned the camp as a place for all campers regardless of their skin condition to feel safe and welcome. This overall mission guides camp leadership and staff every year over the course of the camp week where campers participate in a mix of traditional and nontraditional summer activities that are safe and accessible for all, from spending time in the pool to arts and crafts and a ropes course.
Camp Wonder is in its 21st year of hosting children and adolescents from across North America at its camp in Livermore, California. This year, Tenconi expects about 100 campers during the last week in July. Camp Wonder relies on medical staff volunteers to make the camp setting safe, inclusive, and fun. “Our dermatology residents and dermatology volunteers are a huge part of why we’re able to have camp,” said Tenconi. “A lot of our kids require very specific medical care throughout the week. We are able to provide this camp experience for them because we have this medical support system available, this specialized dermatology knowledge.” She also noted the benefit to the volunteers themselves, saying,“The feedback we get a lot from residents and dermatologists is that camp gave them a chance to understand the true-life impact of some of the skin diseases these kids and families are living with. Kids will open up to them and tell them how their disease has impacted them personally” (oral communication, June 18, 2021).
Volunteer medical providers help manage the medical needs of the campers beginning at check-in and work shifts in the infirmary as well as help with dispensing and administering medications, changing dressings, and applying ointments or other topical medications. When not assisting with medical care, medical staff can get to know the campers; help out with arts and crafts, games, sports, and other camp activities; and put on skits and plays for campers at nightly camp hangouts (Figure 1).
How to Get Involved
Visit the website (https://www.csdf.org/camp-wonder) for information on becoming a medical volunteer for 2022. Donations to help keep the camp running also are greatly appreciated, as attendance, including travel costs, is free for families through the Children’s Skin Disease Foundation. Finally, dermatologists can help by keeping their young patients with skin disease in mind as future campers. The camp welcomes kids from across the United States and Canada and invites questions from dermatologists and families on how to become a camper and what the experience is like.
Native American Health Services Rotation
Located in the southwestern United States, the Navajo Nation is North America’s largest Native American tribe by enrollment and resides on the largest reservation in the United States.10 Comprised of 27,000 square miles within portions of Arizona, New Mexico, and Utah, the reservation’s total area is greater than that of Massachusetts, Vermont, and New Hampshire combined.11 The reservation is home to an estimated 180,000 Navajo people, a population roughly the size of Salt Lake City, Utah. Yet, many homes on the reservation are without electricity, running water, telephones, or broadband access, and many roads on the reservation remain unpaved. Prior to the COVID-19 pandemic, 4 dermatology residents were selected each year to travel to this unique and remote location to work with the staff of the Chinle Comprehensive Health Care Facility (Chinle, Arizona), an Indian Health Service facility, as part of the American Academy of Dermatology (AAD)–sponsored Native American Health Services Resident Rotation (NAHSRR).
Dr. Lucinda Kohn, Assistant Professor of Dermatology at the University of Colorado and the director of the NAHSRR program discovered the value of this rotation firsthand as a dermatology resident. In 2017, she traveled to the area to spend 2 weeks serving within the community. “I went because of a personal connection. My husband is Native American, although not Navajo. I wanted to experience what it was like to provide dermatologic care for Native Americans. I found the Navajo people to be so friendly and so grateful for our care. The clinicians we worked with at Chinle were excited to have us share our expertise and to pass on their knowledge to us,” said Dr. Kohn (personal communication, June 24, 2021).
Rotating residents provide dermatologic care for the Navajo people and share their unique medical skill set to local primary care clinicians serving as preceptors. They also may have an opportunity to learn from Native healers about traditional Navajo beliefs and ceremonies used as part of a holistic approach to healing.
The program, similar to volunteer programs across the country, was put on hold during the height of the COVID-19 pandemic. “The Navajo nation witnessed a really tragic surge of COVID cases that required that limited medical resources be diverted to help cope with the pandemic,” says Dr. Kohn. “It really wasn’t safe for residents to travel to the reservation either, so the rotation had to be put on hold.” However, in April 2021, the health care staff of the Chinle Comprehensive Care Facility reached out to revive the program, which is now pending the green light from the AAD. It is unclear if or when AAD leadership will allow this rotation to restart. Dr. Kohn hopes to be able to start accepting new applications soon. “This rotation provides a wealth of benefits to all those involved, from the residents who get the chance to work with a unique population in need to the clinicians who gain a diverse understanding of dermatology treatment techniques. And of course, for the patients, who are so appreciative of the care they receive from our volunteers” (personal communication, June 25, 2021).
How to Get Involved
Dr. Kohn is happy to field questions regarding the rotation and requests for more information via email (lucinda.kohn@cuanschutz.edu). Residents interested in this program also may reach out to the AAD’s Education and Volunteers Abroad Committee to express interest in the NAHSRR program’s reinstatement.
Destination Healthy Skin
Since 2017, the Skin Cancer Foundation’s Destination Healthy Skin (DHS) RV has been the setting for more than 3800 free skin cancer screenings provided by volunteers within underserved populations across the United States (Figure 2). After a year hiatus due to the pandemic, DHS hit the road again, starting in New York City on August 1 to 3, 2021. From there, the DHS RV will traverse the country in one large loop, starting with visits to large and small cities in the Midwest and the West Coast. Following a visit to San Diego, California, in early October, the RV will turn east, with stops in Arizona, Texas, and several southern states before ending in Philadelphia, Pennsylvania. Dr. Elizabeth Hale, Senior Vice President of the Skin Cancer Foundation, feels that increasing awareness of the importance of regular skin cancer screening for those at risk is more important than ever. “We know that many people in the past year put routine cancer screening on the back burner, but we’re beginning to appreciate that this has led to significant delays in skin cancer diagnosis and potentially more significant disease when cases are diagnosed.” Dr. Hale noted that as the country continues to return to a degree of normalcy, the backlog of patients now seeking their routine screening has led to longer wait times. She expects DHS may offer some relief. “There are no appointments necessary. If the RV is close to their hometown, patients have an advantage in being able to be seen first come, first served, without having to wait for an appointment or make sure their insurance is accepted. It’s a free screening that can increase access to dermatologists” (personal communication, June 21, 2021).
The program’s organizers acknowledge that DHS is not a long-term solution for improving dermatology access in the United States and recognize that more needs to be done to raise awareness, both of the value that screenings can provide and the importance of sun-protective behavior. “This is an important first step,” says Dr. Hale. “It’s important that we disseminate that no one is immune to skin cancer. It’s about education, and this is a tool to educate patients that everyone should have a skin check once a year, regardless of where you live or what your skin type is” (personal communication, June 21, 2021).
Volunteer dermatologists are needed to assist with screenings when the DHS RV arrives in their community. Providers complete a screening form identifying any concerning lesions and can document specific lesions using the patient’s cell phone. Following the screenings, participating dermatologists are welcome to invite participants to make appointments at their practices or suggest local clinics for follow-up care.
How to Get Involved
The schedule for this year’s screening events can be found online (https://www.skincancer.org/early-detection/destination-healthy-skin/). Consider volunteering (https://www.skincancer.org/early-detection/destination-healthy-skin/physician-volunteers/) or helping to raise awareness by reaching out to local dermatology societies or free clinics in your area. Residents and physician’s assistants are welcome to volunteer as well, as long as they are under the on-site supervision of a board-certified dermatologist.
Final Thoughts
As medical professionals, we all recognize there are valuable contributions we can make to groups and organizations that need our help. The stresses and pressure of work and everyday life can make finding the time to offer that help seem impossible. Although it may seem counterintuitive, volunteering our time to help others can help us better navigate the professional burnout that many medical professionals experience today.
- Ezekor M, Pona A, Cline A, et al. An increasing trend in the number of publications and research projects among dermatology residency applicants. J Am Acad Dermatol. 2020;83:214-216.
- Atluri S, Seivright JR, Shi VY, et al. Volunteer and work experiences among dermatology residency applicants. J Am Acad Dermatol. 2021;84:E97-E98.
- Abokwidir M, Davis SA, Fleischer AB, et al. Use of the emergency department for dermatologic care in the United States by ethnic group. J Dermatolog Treat. 2015;26:392-394.
- Uscher-Pines L, Pines J, Kellermann A, et al. Emergency department visits for nonurgent conditions: systematic literature review. Am J Manag Care. 2013;19:47-59.
- Jack AR, Spence AA, Nichols BJ, et al. Cutaneous conditions leading to dermatology consultations in the emergency department. West J Emerg Med. 2011;12:551-555.
- Ayoubi N, Mirza A-S, Swanson J, et al. Dermatologic care of uninsured patients managed at free clinics. J Am Acad Dermatol. 2019;81:433-437.
- Wright AA, Katz IT. Beyond burnout—redesigning care to restore meaning and sanity for physicians. N Engl J Med. 2018;378:309-311.
- Bull C, Aucoin JB. Voluntary association participation and life satisfaction: a replication note. J Gerontol. 1975;30:73-76.
- Iserson KV. Burnout syndrome: global medicine volunteering as a possible treatment strategy. J Emerg Med. 2018;54:516-521.
- Romero S. Navajo Nation becomes largest tribe in U.S. after pandemic enrollment surge. New York Times. May 21, 2021. Accessed August 19, 2021. https://www.nytimes.com/2021/05/21/us/navajo-cherokee-population.html
- Moore GR, Benally J, Tuttle S. The Navajo Nation: quick facts. University of Arizona website. Accessed August 19, 2021. https://extension.arizona.edu/sites/extension.arizona.edu/files/pubs/az1471.pdf
The adage “so much to do, so little time” aptly describes the daily challenges facing dermatologists and dermatology residents. The time and attention required by direct patient care, writing notes, navigating electronic health records, and engaging in education and research as well as family commitments can drain even the most tireless clinician. In addition, dermatologists are expected to play a critical role in clinic and practice management to successfully curate an online presence and adapt their skills to successfully manage a teledermatology practice. Coupled with the time spent socializing with friends or colleagues and time for personal hobbies or exercise, it’s easy to see how sleep deprivation is common in many of our colleagues.
What’s being left out of these jam-packed schedules? Increasingly, it is the time and expertise dedicated to volunteering in our local communities. Two recent research letters highlighted how a dramatic increase in the number of research projects and publications is not mirrored by a similar increase in volunteer experiences as dermatology residency selection becomes more competitive.1,2
Although the rate of volunteerism among practicing dermatologists has yet to be studied, a brief review suggests a component of unmet dermatology need within our communities. It’s estimated that approximately 5% to 10% of all emergency department visits are for dermatologic concerns.3-5 In many cases, the reason for the visit is nonurgent and instead reflects a lack of other options for care. However, the need for dermatologists extends beyond the emergency department setting. A review of the prevalence of patients presenting for care to a group of regional free clinics found that 8% (N=5553) of all visitors sought care for dermatologic concerns.6 The benefit is not just for those seated on the examination table; research has shown that while many of the underlying factors resulting in physician burnout stem from systemic issues, participating in volunteer opportunities helps combat burnout in ourselves and our colleagues.7-9 Herein, opportunities that exist for dermatologists to reconnect with their communities, advocate for causes distinctive to the specialty, and care for neighbors most in need are highlighted.
Camp Wonder
Every year, children from across the United States living with chronic and debilitating skin conditions get the opportunity to join fellow campers and spend a week just being kids without the constant focus on being a patient. Camp Wonder’s founder and director, Francesca Tenconi, describes the camp as a place where kids “can form a community and can feel free to be themselves, without judgment, without stares. They get the chance to forget about their skin disease and be themselves” (oral communication, June 18, 2021). Tenconi and the camp’s cofounders and medical directors, Drs. Jenny Kim and Stefani Takahashi, envisioned the camp as a place for all campers regardless of their skin condition to feel safe and welcome. This overall mission guides camp leadership and staff every year over the course of the camp week where campers participate in a mix of traditional and nontraditional summer activities that are safe and accessible for all, from spending time in the pool to arts and crafts and a ropes course.
Camp Wonder is in its 21st year of hosting children and adolescents from across North America at its camp in Livermore, California. This year, Tenconi expects about 100 campers during the last week in July. Camp Wonder relies on medical staff volunteers to make the camp setting safe, inclusive, and fun. “Our dermatology residents and dermatology volunteers are a huge part of why we’re able to have camp,” said Tenconi. “A lot of our kids require very specific medical care throughout the week. We are able to provide this camp experience for them because we have this medical support system available, this specialized dermatology knowledge.” She also noted the benefit to the volunteers themselves, saying,“The feedback we get a lot from residents and dermatologists is that camp gave them a chance to understand the true-life impact of some of the skin diseases these kids and families are living with. Kids will open up to them and tell them how their disease has impacted them personally” (oral communication, June 18, 2021).
Volunteer medical providers help manage the medical needs of the campers beginning at check-in and work shifts in the infirmary as well as help with dispensing and administering medications, changing dressings, and applying ointments or other topical medications. When not assisting with medical care, medical staff can get to know the campers; help out with arts and crafts, games, sports, and other camp activities; and put on skits and plays for campers at nightly camp hangouts (Figure 1).
How to Get Involved
Visit the website (https://www.csdf.org/camp-wonder) for information on becoming a medical volunteer for 2022. Donations to help keep the camp running also are greatly appreciated, as attendance, including travel costs, is free for families through the Children’s Skin Disease Foundation. Finally, dermatologists can help by keeping their young patients with skin disease in mind as future campers. The camp welcomes kids from across the United States and Canada and invites questions from dermatologists and families on how to become a camper and what the experience is like.
Native American Health Services Rotation
Located in the southwestern United States, the Navajo Nation is North America’s largest Native American tribe by enrollment and resides on the largest reservation in the United States.10 Comprised of 27,000 square miles within portions of Arizona, New Mexico, and Utah, the reservation’s total area is greater than that of Massachusetts, Vermont, and New Hampshire combined.11 The reservation is home to an estimated 180,000 Navajo people, a population roughly the size of Salt Lake City, Utah. Yet, many homes on the reservation are without electricity, running water, telephones, or broadband access, and many roads on the reservation remain unpaved. Prior to the COVID-19 pandemic, 4 dermatology residents were selected each year to travel to this unique and remote location to work with the staff of the Chinle Comprehensive Health Care Facility (Chinle, Arizona), an Indian Health Service facility, as part of the American Academy of Dermatology (AAD)–sponsored Native American Health Services Resident Rotation (NAHSRR).
Dr. Lucinda Kohn, Assistant Professor of Dermatology at the University of Colorado and the director of the NAHSRR program discovered the value of this rotation firsthand as a dermatology resident. In 2017, she traveled to the area to spend 2 weeks serving within the community. “I went because of a personal connection. My husband is Native American, although not Navajo. I wanted to experience what it was like to provide dermatologic care for Native Americans. I found the Navajo people to be so friendly and so grateful for our care. The clinicians we worked with at Chinle were excited to have us share our expertise and to pass on their knowledge to us,” said Dr. Kohn (personal communication, June 24, 2021).
Rotating residents provide dermatologic care for the Navajo people and share their unique medical skill set to local primary care clinicians serving as preceptors. They also may have an opportunity to learn from Native healers about traditional Navajo beliefs and ceremonies used as part of a holistic approach to healing.
The program, similar to volunteer programs across the country, was put on hold during the height of the COVID-19 pandemic. “The Navajo nation witnessed a really tragic surge of COVID cases that required that limited medical resources be diverted to help cope with the pandemic,” says Dr. Kohn. “It really wasn’t safe for residents to travel to the reservation either, so the rotation had to be put on hold.” However, in April 2021, the health care staff of the Chinle Comprehensive Care Facility reached out to revive the program, which is now pending the green light from the AAD. It is unclear if or when AAD leadership will allow this rotation to restart. Dr. Kohn hopes to be able to start accepting new applications soon. “This rotation provides a wealth of benefits to all those involved, from the residents who get the chance to work with a unique population in need to the clinicians who gain a diverse understanding of dermatology treatment techniques. And of course, for the patients, who are so appreciative of the care they receive from our volunteers” (personal communication, June 25, 2021).
How to Get Involved
Dr. Kohn is happy to field questions regarding the rotation and requests for more information via email (lucinda.kohn@cuanschutz.edu). Residents interested in this program also may reach out to the AAD’s Education and Volunteers Abroad Committee to express interest in the NAHSRR program’s reinstatement.
Destination Healthy Skin
Since 2017, the Skin Cancer Foundation’s Destination Healthy Skin (DHS) RV has been the setting for more than 3800 free skin cancer screenings provided by volunteers within underserved populations across the United States (Figure 2). After a year hiatus due to the pandemic, DHS hit the road again, starting in New York City on August 1 to 3, 2021. From there, the DHS RV will traverse the country in one large loop, starting with visits to large and small cities in the Midwest and the West Coast. Following a visit to San Diego, California, in early October, the RV will turn east, with stops in Arizona, Texas, and several southern states before ending in Philadelphia, Pennsylvania. Dr. Elizabeth Hale, Senior Vice President of the Skin Cancer Foundation, feels that increasing awareness of the importance of regular skin cancer screening for those at risk is more important than ever. “We know that many people in the past year put routine cancer screening on the back burner, but we’re beginning to appreciate that this has led to significant delays in skin cancer diagnosis and potentially more significant disease when cases are diagnosed.” Dr. Hale noted that as the country continues to return to a degree of normalcy, the backlog of patients now seeking their routine screening has led to longer wait times. She expects DHS may offer some relief. “There are no appointments necessary. If the RV is close to their hometown, patients have an advantage in being able to be seen first come, first served, without having to wait for an appointment or make sure their insurance is accepted. It’s a free screening that can increase access to dermatologists” (personal communication, June 21, 2021).
The program’s organizers acknowledge that DHS is not a long-term solution for improving dermatology access in the United States and recognize that more needs to be done to raise awareness, both of the value that screenings can provide and the importance of sun-protective behavior. “This is an important first step,” says Dr. Hale. “It’s important that we disseminate that no one is immune to skin cancer. It’s about education, and this is a tool to educate patients that everyone should have a skin check once a year, regardless of where you live or what your skin type is” (personal communication, June 21, 2021).
Volunteer dermatologists are needed to assist with screenings when the DHS RV arrives in their community. Providers complete a screening form identifying any concerning lesions and can document specific lesions using the patient’s cell phone. Following the screenings, participating dermatologists are welcome to invite participants to make appointments at their practices or suggest local clinics for follow-up care.
How to Get Involved
The schedule for this year’s screening events can be found online (https://www.skincancer.org/early-detection/destination-healthy-skin/). Consider volunteering (https://www.skincancer.org/early-detection/destination-healthy-skin/physician-volunteers/) or helping to raise awareness by reaching out to local dermatology societies or free clinics in your area. Residents and physician’s assistants are welcome to volunteer as well, as long as they are under the on-site supervision of a board-certified dermatologist.
Final Thoughts
As medical professionals, we all recognize there are valuable contributions we can make to groups and organizations that need our help. The stresses and pressure of work and everyday life can make finding the time to offer that help seem impossible. Although it may seem counterintuitive, volunteering our time to help others can help us better navigate the professional burnout that many medical professionals experience today.
The adage “so much to do, so little time” aptly describes the daily challenges facing dermatologists and dermatology residents. The time and attention required by direct patient care, writing notes, navigating electronic health records, and engaging in education and research as well as family commitments can drain even the most tireless clinician. In addition, dermatologists are expected to play a critical role in clinic and practice management to successfully curate an online presence and adapt their skills to successfully manage a teledermatology practice. Coupled with the time spent socializing with friends or colleagues and time for personal hobbies or exercise, it’s easy to see how sleep deprivation is common in many of our colleagues.
What’s being left out of these jam-packed schedules? Increasingly, it is the time and expertise dedicated to volunteering in our local communities. Two recent research letters highlighted how a dramatic increase in the number of research projects and publications is not mirrored by a similar increase in volunteer experiences as dermatology residency selection becomes more competitive.1,2
Although the rate of volunteerism among practicing dermatologists has yet to be studied, a brief review suggests a component of unmet dermatology need within our communities. It’s estimated that approximately 5% to 10% of all emergency department visits are for dermatologic concerns.3-5 In many cases, the reason for the visit is nonurgent and instead reflects a lack of other options for care. However, the need for dermatologists extends beyond the emergency department setting. A review of the prevalence of patients presenting for care to a group of regional free clinics found that 8% (N=5553) of all visitors sought care for dermatologic concerns.6 The benefit is not just for those seated on the examination table; research has shown that while many of the underlying factors resulting in physician burnout stem from systemic issues, participating in volunteer opportunities helps combat burnout in ourselves and our colleagues.7-9 Herein, opportunities that exist for dermatologists to reconnect with their communities, advocate for causes distinctive to the specialty, and care for neighbors most in need are highlighted.
Camp Wonder
Every year, children from across the United States living with chronic and debilitating skin conditions get the opportunity to join fellow campers and spend a week just being kids without the constant focus on being a patient. Camp Wonder’s founder and director, Francesca Tenconi, describes the camp as a place where kids “can form a community and can feel free to be themselves, without judgment, without stares. They get the chance to forget about their skin disease and be themselves” (oral communication, June 18, 2021). Tenconi and the camp’s cofounders and medical directors, Drs. Jenny Kim and Stefani Takahashi, envisioned the camp as a place for all campers regardless of their skin condition to feel safe and welcome. This overall mission guides camp leadership and staff every year over the course of the camp week where campers participate in a mix of traditional and nontraditional summer activities that are safe and accessible for all, from spending time in the pool to arts and crafts and a ropes course.
Camp Wonder is in its 21st year of hosting children and adolescents from across North America at its camp in Livermore, California. This year, Tenconi expects about 100 campers during the last week in July. Camp Wonder relies on medical staff volunteers to make the camp setting safe, inclusive, and fun. “Our dermatology residents and dermatology volunteers are a huge part of why we’re able to have camp,” said Tenconi. “A lot of our kids require very specific medical care throughout the week. We are able to provide this camp experience for them because we have this medical support system available, this specialized dermatology knowledge.” She also noted the benefit to the volunteers themselves, saying,“The feedback we get a lot from residents and dermatologists is that camp gave them a chance to understand the true-life impact of some of the skin diseases these kids and families are living with. Kids will open up to them and tell them how their disease has impacted them personally” (oral communication, June 18, 2021).
Volunteer medical providers help manage the medical needs of the campers beginning at check-in and work shifts in the infirmary as well as help with dispensing and administering medications, changing dressings, and applying ointments or other topical medications. When not assisting with medical care, medical staff can get to know the campers; help out with arts and crafts, games, sports, and other camp activities; and put on skits and plays for campers at nightly camp hangouts (Figure 1).
How to Get Involved
Visit the website (https://www.csdf.org/camp-wonder) for information on becoming a medical volunteer for 2022. Donations to help keep the camp running also are greatly appreciated, as attendance, including travel costs, is free for families through the Children’s Skin Disease Foundation. Finally, dermatologists can help by keeping their young patients with skin disease in mind as future campers. The camp welcomes kids from across the United States and Canada and invites questions from dermatologists and families on how to become a camper and what the experience is like.
Native American Health Services Rotation
Located in the southwestern United States, the Navajo Nation is North America’s largest Native American tribe by enrollment and resides on the largest reservation in the United States.10 Comprised of 27,000 square miles within portions of Arizona, New Mexico, and Utah, the reservation’s total area is greater than that of Massachusetts, Vermont, and New Hampshire combined.11 The reservation is home to an estimated 180,000 Navajo people, a population roughly the size of Salt Lake City, Utah. Yet, many homes on the reservation are without electricity, running water, telephones, or broadband access, and many roads on the reservation remain unpaved. Prior to the COVID-19 pandemic, 4 dermatology residents were selected each year to travel to this unique and remote location to work with the staff of the Chinle Comprehensive Health Care Facility (Chinle, Arizona), an Indian Health Service facility, as part of the American Academy of Dermatology (AAD)–sponsored Native American Health Services Resident Rotation (NAHSRR).
Dr. Lucinda Kohn, Assistant Professor of Dermatology at the University of Colorado and the director of the NAHSRR program discovered the value of this rotation firsthand as a dermatology resident. In 2017, she traveled to the area to spend 2 weeks serving within the community. “I went because of a personal connection. My husband is Native American, although not Navajo. I wanted to experience what it was like to provide dermatologic care for Native Americans. I found the Navajo people to be so friendly and so grateful for our care. The clinicians we worked with at Chinle were excited to have us share our expertise and to pass on their knowledge to us,” said Dr. Kohn (personal communication, June 24, 2021).
Rotating residents provide dermatologic care for the Navajo people and share their unique medical skill set to local primary care clinicians serving as preceptors. They also may have an opportunity to learn from Native healers about traditional Navajo beliefs and ceremonies used as part of a holistic approach to healing.
The program, similar to volunteer programs across the country, was put on hold during the height of the COVID-19 pandemic. “The Navajo nation witnessed a really tragic surge of COVID cases that required that limited medical resources be diverted to help cope with the pandemic,” says Dr. Kohn. “It really wasn’t safe for residents to travel to the reservation either, so the rotation had to be put on hold.” However, in April 2021, the health care staff of the Chinle Comprehensive Care Facility reached out to revive the program, which is now pending the green light from the AAD. It is unclear if or when AAD leadership will allow this rotation to restart. Dr. Kohn hopes to be able to start accepting new applications soon. “This rotation provides a wealth of benefits to all those involved, from the residents who get the chance to work with a unique population in need to the clinicians who gain a diverse understanding of dermatology treatment techniques. And of course, for the patients, who are so appreciative of the care they receive from our volunteers” (personal communication, June 25, 2021).
How to Get Involved
Dr. Kohn is happy to field questions regarding the rotation and requests for more information via email (lucinda.kohn@cuanschutz.edu). Residents interested in this program also may reach out to the AAD’s Education and Volunteers Abroad Committee to express interest in the NAHSRR program’s reinstatement.
Destination Healthy Skin
Since 2017, the Skin Cancer Foundation’s Destination Healthy Skin (DHS) RV has been the setting for more than 3800 free skin cancer screenings provided by volunteers within underserved populations across the United States (Figure 2). After a year hiatus due to the pandemic, DHS hit the road again, starting in New York City on August 1 to 3, 2021. From there, the DHS RV will traverse the country in one large loop, starting with visits to large and small cities in the Midwest and the West Coast. Following a visit to San Diego, California, in early October, the RV will turn east, with stops in Arizona, Texas, and several southern states before ending in Philadelphia, Pennsylvania. Dr. Elizabeth Hale, Senior Vice President of the Skin Cancer Foundation, feels that increasing awareness of the importance of regular skin cancer screening for those at risk is more important than ever. “We know that many people in the past year put routine cancer screening on the back burner, but we’re beginning to appreciate that this has led to significant delays in skin cancer diagnosis and potentially more significant disease when cases are diagnosed.” Dr. Hale noted that as the country continues to return to a degree of normalcy, the backlog of patients now seeking their routine screening has led to longer wait times. She expects DHS may offer some relief. “There are no appointments necessary. If the RV is close to their hometown, patients have an advantage in being able to be seen first come, first served, without having to wait for an appointment or make sure their insurance is accepted. It’s a free screening that can increase access to dermatologists” (personal communication, June 21, 2021).
The program’s organizers acknowledge that DHS is not a long-term solution for improving dermatology access in the United States and recognize that more needs to be done to raise awareness, both of the value that screenings can provide and the importance of sun-protective behavior. “This is an important first step,” says Dr. Hale. “It’s important that we disseminate that no one is immune to skin cancer. It’s about education, and this is a tool to educate patients that everyone should have a skin check once a year, regardless of where you live or what your skin type is” (personal communication, June 21, 2021).
Volunteer dermatologists are needed to assist with screenings when the DHS RV arrives in their community. Providers complete a screening form identifying any concerning lesions and can document specific lesions using the patient’s cell phone. Following the screenings, participating dermatologists are welcome to invite participants to make appointments at their practices or suggest local clinics for follow-up care.
How to Get Involved
The schedule for this year’s screening events can be found online (https://www.skincancer.org/early-detection/destination-healthy-skin/). Consider volunteering (https://www.skincancer.org/early-detection/destination-healthy-skin/physician-volunteers/) or helping to raise awareness by reaching out to local dermatology societies or free clinics in your area. Residents and physician’s assistants are welcome to volunteer as well, as long as they are under the on-site supervision of a board-certified dermatologist.
Final Thoughts
As medical professionals, we all recognize there are valuable contributions we can make to groups and organizations that need our help. The stresses and pressure of work and everyday life can make finding the time to offer that help seem impossible. Although it may seem counterintuitive, volunteering our time to help others can help us better navigate the professional burnout that many medical professionals experience today.
- Ezekor M, Pona A, Cline A, et al. An increasing trend in the number of publications and research projects among dermatology residency applicants. J Am Acad Dermatol. 2020;83:214-216.
- Atluri S, Seivright JR, Shi VY, et al. Volunteer and work experiences among dermatology residency applicants. J Am Acad Dermatol. 2021;84:E97-E98.
- Abokwidir M, Davis SA, Fleischer AB, et al. Use of the emergency department for dermatologic care in the United States by ethnic group. J Dermatolog Treat. 2015;26:392-394.
- Uscher-Pines L, Pines J, Kellermann A, et al. Emergency department visits for nonurgent conditions: systematic literature review. Am J Manag Care. 2013;19:47-59.
- Jack AR, Spence AA, Nichols BJ, et al. Cutaneous conditions leading to dermatology consultations in the emergency department. West J Emerg Med. 2011;12:551-555.
- Ayoubi N, Mirza A-S, Swanson J, et al. Dermatologic care of uninsured patients managed at free clinics. J Am Acad Dermatol. 2019;81:433-437.
- Wright AA, Katz IT. Beyond burnout—redesigning care to restore meaning and sanity for physicians. N Engl J Med. 2018;378:309-311.
- Bull C, Aucoin JB. Voluntary association participation and life satisfaction: a replication note. J Gerontol. 1975;30:73-76.
- Iserson KV. Burnout syndrome: global medicine volunteering as a possible treatment strategy. J Emerg Med. 2018;54:516-521.
- Romero S. Navajo Nation becomes largest tribe in U.S. after pandemic enrollment surge. New York Times. May 21, 2021. Accessed August 19, 2021. https://www.nytimes.com/2021/05/21/us/navajo-cherokee-population.html
- Moore GR, Benally J, Tuttle S. The Navajo Nation: quick facts. University of Arizona website. Accessed August 19, 2021. https://extension.arizona.edu/sites/extension.arizona.edu/files/pubs/az1471.pdf
- Ezekor M, Pona A, Cline A, et al. An increasing trend in the number of publications and research projects among dermatology residency applicants. J Am Acad Dermatol. 2020;83:214-216.
- Atluri S, Seivright JR, Shi VY, et al. Volunteer and work experiences among dermatology residency applicants. J Am Acad Dermatol. 2021;84:E97-E98.
- Abokwidir M, Davis SA, Fleischer AB, et al. Use of the emergency department for dermatologic care in the United States by ethnic group. J Dermatolog Treat. 2015;26:392-394.
- Uscher-Pines L, Pines J, Kellermann A, et al. Emergency department visits for nonurgent conditions: systematic literature review. Am J Manag Care. 2013;19:47-59.
- Jack AR, Spence AA, Nichols BJ, et al. Cutaneous conditions leading to dermatology consultations in the emergency department. West J Emerg Med. 2011;12:551-555.
- Ayoubi N, Mirza A-S, Swanson J, et al. Dermatologic care of uninsured patients managed at free clinics. J Am Acad Dermatol. 2019;81:433-437.
- Wright AA, Katz IT. Beyond burnout—redesigning care to restore meaning and sanity for physicians. N Engl J Med. 2018;378:309-311.
- Bull C, Aucoin JB. Voluntary association participation and life satisfaction: a replication note. J Gerontol. 1975;30:73-76.
- Iserson KV. Burnout syndrome: global medicine volunteering as a possible treatment strategy. J Emerg Med. 2018;54:516-521.
- Romero S. Navajo Nation becomes largest tribe in U.S. after pandemic enrollment surge. New York Times. May 21, 2021. Accessed August 19, 2021. https://www.nytimes.com/2021/05/21/us/navajo-cherokee-population.html
- Moore GR, Benally J, Tuttle S. The Navajo Nation: quick facts. University of Arizona website. Accessed August 19, 2021. https://extension.arizona.edu/sites/extension.arizona.edu/files/pubs/az1471.pdf
Resident Pearl
- Volunteerism rates among dermatology residents seem to be decreasing. We should work to combat this trend by finding ways to give back to our communities and spur our colleagues to do the same.
Verrucous Scalp Plaque and Widespread Eruption
The Diagnosis: Pemphigus Foliaceous
Laboratory workup including a complete blood cell count with differential, comprehensive metabolic panel, antinuclear antibodies, Sjögren syndrome A and B antibodies, hepatitis profile, rapid plasma reagin, HIV screen, aldolase, anti–Jo-1, T-Spot TB test (Quest Diagnostics), and tissue cultures was unremarkable. Two 4-mm punch biopsies were obtained from the left cheek and upper back, both of which demonstrated intragranular acantholysis suggestive of pemphigus foliaceous (Figure 1A). A subsequent punch biopsy from the right lower abdomen sent for direct immunofluorescence demonstrated netlike positivity of IgG and C3 in the upper epidermis (Figure 1B), and serum sent for indirect immunofluorescence demonstrated intercellular IgG antibodies to desmoglein (Dsg) 1 on monkey esophagus and positive Dsg-1 antibodies on enzyme-linked immunosorbent assay, confirming the diagnosis.
The patient was started on a 60-mg prednisone taper as well as dapsone 50 mg daily; the dapsone was titrated up to 100 mg daily. After tapering down to 10 mg daily of prednisone over 2 months and continuing dapsone with minimal improvement, he was given 2 infusions of rituximab 1000 mg 2 weeks apart. The scalp plaque was dramatically improved at 3-month follow-up (Figure 2), with partial improvement of the cheek plaques (Figure 3). Dapsone was increased to 150 mg daily, and he was encouraged to use triamcinolone acetonide ointment 0.1% twice daily, which led to further improvement.
Pemphigus foliaceus is an autoimmune blistering disease that most commonly occurs in middle-aged adults. It generally is less common than pemphigus vulgaris, except in Finland, Tunisia, and Brazil, where there is an endemic condition with an identical clinical and histological presentation known as fogo selvagem.1
The pathogenesis of pemphigus foliaceous is characterized by IgG autoantibodies against Dsg-1, a transmembrane glycoprotein involved in the cellular adhesion of keratinocytes, which is preferentially expressed in the superficial epidermis.2-7 Dysfunction of Dsg-1 results in the separation of superficial epidermal cells, resulting in intraepidermal blisters.2,7 In contrast to pemphigus vulgaris, there typically is a lack of oral mucosal involvement due to compensation by Dsg-3 in the mucosa.4 Potential triggers for pemphigus foliaceous include exposure to UV radiation; radiotherapy; pregnancy; physiologic stress; and drugs, most commonly captopril, penicillamine, and thiols.8
Pemphigus foliaceous lesions clinically appear as eroded and crusted lesions on an erythematous base, commonly in a seborrheic distribution on the face, scalp, and trunk with sparing of the oral mucosa,2,6 but lesions can progress to a widespread and more severe exfoliative dermatitis.7 Lesions also can appear as psoriasiform plaques and often are initially misdiagnosed as psoriasis, particularly in patients with skin of color.9,10
Diagnosis of pemphigus foliaceous typically is made using a combination of histology as well as both direct and indirect immunofluorescence. Histologically, pemphigus foliaceus presents with subcorneal acantholysis, which is most prominent in the granular layer and occasionally the presence of neutrophils and eosinophils in the blister cavity.7 Direct immunofluorescence demonstrates netlike intercellular IgG and C3 in the upper portion of the epidermis.11 Indirect immunofluorescence can help detect circulating IgG antibodies to Dsg-1, with guinea pig esophagus being the ideal substrate.11,12
First-line treatment of pemphigus foliaceus consists of systemic glucocorticoid therapy, often administered with azathioprine, methotrexate, or mycophenolate mofetil.2,6,13 Although first-line treatment is effective in 60% to 80% of patients,2 relapsing cases can be treated with cyclophosphamide, intravenous immunoglobulin, immunoadsorption, plasmapheresis, or rituximab.2
Rituximab is a chimeric monoclonal antibody targeting CD20+ B cells, leading to decreased antibody production, which has been shown to be effective in treating severe and refractory cases of pemphigus foliaceus.6,13Rituximab with short-course prednisone has been found to be more effective in achieving complete remission at 24 months than prednisone alone.14 In patients with contraindications to systemic glucocorticoid therapy, rituximab has been shown as an effective first-line therapy.15 One-quarter of patients treated with rituximab relapsed within 2 years of treatment6 (average time to relapse, 6–26 months).16 High-dose rituximab regimens, along with a higher number of rituximab treatment cycles, have been shown to prolong time to relapse.6 Further, higher baseline levels of Dsg-1 antibody have been correlated to earlier relapse and can be used following rituximab therapy to monitor disease progression.6,16
The differential diagnosis for pemphigus foliaceous includes disseminated blastomycosis, hypertrophic lupus erythematosus, sebopsoriasis, and secondary syphilis. Disseminated blastomycosis presents with cutaneous manifestations such as nodules, papules, or pustules evolving over weeks to months into ulcers with subsequent scarring.17 Hypertrophic lupus erythematosus presents with papules and nodules with associated keratotic scaling on the face, palms, and extensor surfaces of the limbs.18 Sebopsoriasis is characterized by well-defined lesions with an overlying scale distributed on the scalp, face, and chest.19 Secondary syphilis presents as early hyperpigmented macules transitioning to acral papulosquamous lesions involving the palms and soles.20
- Hans-Filho G, Aoki V, Hans Bittner NR, et al. Fogo selvagem: endemic pemphigus foliaceus. An Bras Dermatol. 2018;93:638-650.
- Jenson KK, Burr DM, Edwards BC. Case report: reatment of refractory pemphigus foliaceus with rituximab. Practical Dermatology. February 2016:33-36. Accessed August 27, 2021. https://practicaldermatology.com/articles/2016-feb/case-report -treatment-of-refractory-pemphigus-foliaceus-with-rituximab -financial-matters-aad-asds-resources
- Amagai M, Hashimoto T, Green KJ, et al. Antigen-specific immunoadsorption of pathogenic autoantibodies in pemphigus foliaceus. J Invest Dermatol. 1995;104:895-901.
- Mahoney MG, Wang Z, Rothenberger K, et al. Explanations for the clinical and microscopic localization of lesions in pemphigus foliaceus and vulgaris. J Clin Invest. 1999;103:461-468.
- Oktarina DAM, Sokol E, Kramer D, et al. Endocytosis of IgG, desmoglein 1, and plakoglobin in pemphigus foliaceus patient skin. Front Immunol. 2019;10:1-12.
- Kraft M, Worm M. Pemphigus foliaceus-repeated treatment with rituximab 7 years after initial response: a case report. Front Med. 2018;5:315.
- Hale EK. Pemphigus foliaceous. Dermatol Online J. 2002;8:9.
- Tavakolpour S. Pemphigus trigger factors: special focus on pemphigus vulgaris and pemphigus foliaceus. Arch Dermatol Res. 2018;310:95-106.
- A boobaker J, Morar N, Ramdial PK, et al. Pemphigus in South Africa. Int J Dermatol. 2001;40:115-119.
- Austin E, Millsop JW, Ely H, et al. Psoriasiform pemphigus foliaceus in an African American female: an important clinical manifestation. J Drugs Dermatol. 2018;17:471.
- Arbache ST, Nogueira TG, Delgado L, et al. Immunofluorescence testing in the diagnosis of autoimmune blistering diseases: overview of 10-year experience. An Bras Dermatol. 2014;89:885-889.
- Sabolinski ML, Beutner EH, Krasny S, et al. Substrate specificity of antiepithelial antibodies of pemphigus vulgaris and pemphigus foliaceus sera in immunofluorescence tests on monkey and guinea pig esophagus sections. J Invest Dermatol. 1987;88:545-549.
- Palacios-Álvarez I, Riquelme-McLoughlin C, Curto-Barredo L, et al. Rituximab treatment of pemphigus foliaceus: a retrospective study of 12 patients. J Am Acad Dermatol. 2021;85:484-486.
- Murrell DF, Sprecher E. Rituximab and short-course prednisone as the new gold standard for new-onset pemphigus vulgaris and pemphigus foliaceus. Br J Dermatol. 2017;177:1143-1144.
- Gregoriou S, Efthymiou O, Stefanaki C, et al. Management of pemphigus vulgaris: challenges and solutions. Clin Cosmet Investig Dermatol. 2015;8:521-527.
- Saleh MA. A prospective study comparing patients with early and late relapsing pemphigus treated with rituximab. J Am Acad Dermatol. 2018;79:97-103.
- Castillo CG, Kauffman CA, Miceli MH. Blastomycosis. Infect Dis Clin North Am. 2016;30:247-264.
- Herzum A, Gasparini G, Emanuele C, et al. Atypical and rare forms of cutaneous lupus erythematosus: the importance of the diagnosis for the best management of patients. Dermatology. 2013;1-10.
- Tull TJ, Noy M, Bunker CB, et al. Sebopsoriasis in patients with HIV: a case series of 20 patients. Br J Dermatol. 2016; 173:813-815.
- Balagula Y, Mattei P, Wisco OJ, et al. The great imitator revised: the spectrum of atypical cutaneous manifestations of secondary syphilis. Int J Dermatol. 2014;53:1434-1441.
The Diagnosis: Pemphigus Foliaceous
Laboratory workup including a complete blood cell count with differential, comprehensive metabolic panel, antinuclear antibodies, Sjögren syndrome A and B antibodies, hepatitis profile, rapid plasma reagin, HIV screen, aldolase, anti–Jo-1, T-Spot TB test (Quest Diagnostics), and tissue cultures was unremarkable. Two 4-mm punch biopsies were obtained from the left cheek and upper back, both of which demonstrated intragranular acantholysis suggestive of pemphigus foliaceous (Figure 1A). A subsequent punch biopsy from the right lower abdomen sent for direct immunofluorescence demonstrated netlike positivity of IgG and C3 in the upper epidermis (Figure 1B), and serum sent for indirect immunofluorescence demonstrated intercellular IgG antibodies to desmoglein (Dsg) 1 on monkey esophagus and positive Dsg-1 antibodies on enzyme-linked immunosorbent assay, confirming the diagnosis.
The patient was started on a 60-mg prednisone taper as well as dapsone 50 mg daily; the dapsone was titrated up to 100 mg daily. After tapering down to 10 mg daily of prednisone over 2 months and continuing dapsone with minimal improvement, he was given 2 infusions of rituximab 1000 mg 2 weeks apart. The scalp plaque was dramatically improved at 3-month follow-up (Figure 2), with partial improvement of the cheek plaques (Figure 3). Dapsone was increased to 150 mg daily, and he was encouraged to use triamcinolone acetonide ointment 0.1% twice daily, which led to further improvement.
Pemphigus foliaceus is an autoimmune blistering disease that most commonly occurs in middle-aged adults. It generally is less common than pemphigus vulgaris, except in Finland, Tunisia, and Brazil, where there is an endemic condition with an identical clinical and histological presentation known as fogo selvagem.1
The pathogenesis of pemphigus foliaceous is characterized by IgG autoantibodies against Dsg-1, a transmembrane glycoprotein involved in the cellular adhesion of keratinocytes, which is preferentially expressed in the superficial epidermis.2-7 Dysfunction of Dsg-1 results in the separation of superficial epidermal cells, resulting in intraepidermal blisters.2,7 In contrast to pemphigus vulgaris, there typically is a lack of oral mucosal involvement due to compensation by Dsg-3 in the mucosa.4 Potential triggers for pemphigus foliaceous include exposure to UV radiation; radiotherapy; pregnancy; physiologic stress; and drugs, most commonly captopril, penicillamine, and thiols.8
Pemphigus foliaceous lesions clinically appear as eroded and crusted lesions on an erythematous base, commonly in a seborrheic distribution on the face, scalp, and trunk with sparing of the oral mucosa,2,6 but lesions can progress to a widespread and more severe exfoliative dermatitis.7 Lesions also can appear as psoriasiform plaques and often are initially misdiagnosed as psoriasis, particularly in patients with skin of color.9,10
Diagnosis of pemphigus foliaceous typically is made using a combination of histology as well as both direct and indirect immunofluorescence. Histologically, pemphigus foliaceus presents with subcorneal acantholysis, which is most prominent in the granular layer and occasionally the presence of neutrophils and eosinophils in the blister cavity.7 Direct immunofluorescence demonstrates netlike intercellular IgG and C3 in the upper portion of the epidermis.11 Indirect immunofluorescence can help detect circulating IgG antibodies to Dsg-1, with guinea pig esophagus being the ideal substrate.11,12
First-line treatment of pemphigus foliaceus consists of systemic glucocorticoid therapy, often administered with azathioprine, methotrexate, or mycophenolate mofetil.2,6,13 Although first-line treatment is effective in 60% to 80% of patients,2 relapsing cases can be treated with cyclophosphamide, intravenous immunoglobulin, immunoadsorption, plasmapheresis, or rituximab.2
Rituximab is a chimeric monoclonal antibody targeting CD20+ B cells, leading to decreased antibody production, which has been shown to be effective in treating severe and refractory cases of pemphigus foliaceus.6,13Rituximab with short-course prednisone has been found to be more effective in achieving complete remission at 24 months than prednisone alone.14 In patients with contraindications to systemic glucocorticoid therapy, rituximab has been shown as an effective first-line therapy.15 One-quarter of patients treated with rituximab relapsed within 2 years of treatment6 (average time to relapse, 6–26 months).16 High-dose rituximab regimens, along with a higher number of rituximab treatment cycles, have been shown to prolong time to relapse.6 Further, higher baseline levels of Dsg-1 antibody have been correlated to earlier relapse and can be used following rituximab therapy to monitor disease progression.6,16
The differential diagnosis for pemphigus foliaceous includes disseminated blastomycosis, hypertrophic lupus erythematosus, sebopsoriasis, and secondary syphilis. Disseminated blastomycosis presents with cutaneous manifestations such as nodules, papules, or pustules evolving over weeks to months into ulcers with subsequent scarring.17 Hypertrophic lupus erythematosus presents with papules and nodules with associated keratotic scaling on the face, palms, and extensor surfaces of the limbs.18 Sebopsoriasis is characterized by well-defined lesions with an overlying scale distributed on the scalp, face, and chest.19 Secondary syphilis presents as early hyperpigmented macules transitioning to acral papulosquamous lesions involving the palms and soles.20
The Diagnosis: Pemphigus Foliaceous
Laboratory workup including a complete blood cell count with differential, comprehensive metabolic panel, antinuclear antibodies, Sjögren syndrome A and B antibodies, hepatitis profile, rapid plasma reagin, HIV screen, aldolase, anti–Jo-1, T-Spot TB test (Quest Diagnostics), and tissue cultures was unremarkable. Two 4-mm punch biopsies were obtained from the left cheek and upper back, both of which demonstrated intragranular acantholysis suggestive of pemphigus foliaceous (Figure 1A). A subsequent punch biopsy from the right lower abdomen sent for direct immunofluorescence demonstrated netlike positivity of IgG and C3 in the upper epidermis (Figure 1B), and serum sent for indirect immunofluorescence demonstrated intercellular IgG antibodies to desmoglein (Dsg) 1 on monkey esophagus and positive Dsg-1 antibodies on enzyme-linked immunosorbent assay, confirming the diagnosis.
The patient was started on a 60-mg prednisone taper as well as dapsone 50 mg daily; the dapsone was titrated up to 100 mg daily. After tapering down to 10 mg daily of prednisone over 2 months and continuing dapsone with minimal improvement, he was given 2 infusions of rituximab 1000 mg 2 weeks apart. The scalp plaque was dramatically improved at 3-month follow-up (Figure 2), with partial improvement of the cheek plaques (Figure 3). Dapsone was increased to 150 mg daily, and he was encouraged to use triamcinolone acetonide ointment 0.1% twice daily, which led to further improvement.
Pemphigus foliaceus is an autoimmune blistering disease that most commonly occurs in middle-aged adults. It generally is less common than pemphigus vulgaris, except in Finland, Tunisia, and Brazil, where there is an endemic condition with an identical clinical and histological presentation known as fogo selvagem.1
The pathogenesis of pemphigus foliaceous is characterized by IgG autoantibodies against Dsg-1, a transmembrane glycoprotein involved in the cellular adhesion of keratinocytes, which is preferentially expressed in the superficial epidermis.2-7 Dysfunction of Dsg-1 results in the separation of superficial epidermal cells, resulting in intraepidermal blisters.2,7 In contrast to pemphigus vulgaris, there typically is a lack of oral mucosal involvement due to compensation by Dsg-3 in the mucosa.4 Potential triggers for pemphigus foliaceous include exposure to UV radiation; radiotherapy; pregnancy; physiologic stress; and drugs, most commonly captopril, penicillamine, and thiols.8
Pemphigus foliaceous lesions clinically appear as eroded and crusted lesions on an erythematous base, commonly in a seborrheic distribution on the face, scalp, and trunk with sparing of the oral mucosa,2,6 but lesions can progress to a widespread and more severe exfoliative dermatitis.7 Lesions also can appear as psoriasiform plaques and often are initially misdiagnosed as psoriasis, particularly in patients with skin of color.9,10
Diagnosis of pemphigus foliaceous typically is made using a combination of histology as well as both direct and indirect immunofluorescence. Histologically, pemphigus foliaceus presents with subcorneal acantholysis, which is most prominent in the granular layer and occasionally the presence of neutrophils and eosinophils in the blister cavity.7 Direct immunofluorescence demonstrates netlike intercellular IgG and C3 in the upper portion of the epidermis.11 Indirect immunofluorescence can help detect circulating IgG antibodies to Dsg-1, with guinea pig esophagus being the ideal substrate.11,12
First-line treatment of pemphigus foliaceus consists of systemic glucocorticoid therapy, often administered with azathioprine, methotrexate, or mycophenolate mofetil.2,6,13 Although first-line treatment is effective in 60% to 80% of patients,2 relapsing cases can be treated with cyclophosphamide, intravenous immunoglobulin, immunoadsorption, plasmapheresis, or rituximab.2
Rituximab is a chimeric monoclonal antibody targeting CD20+ B cells, leading to decreased antibody production, which has been shown to be effective in treating severe and refractory cases of pemphigus foliaceus.6,13Rituximab with short-course prednisone has been found to be more effective in achieving complete remission at 24 months than prednisone alone.14 In patients with contraindications to systemic glucocorticoid therapy, rituximab has been shown as an effective first-line therapy.15 One-quarter of patients treated with rituximab relapsed within 2 years of treatment6 (average time to relapse, 6–26 months).16 High-dose rituximab regimens, along with a higher number of rituximab treatment cycles, have been shown to prolong time to relapse.6 Further, higher baseline levels of Dsg-1 antibody have been correlated to earlier relapse and can be used following rituximab therapy to monitor disease progression.6,16
The differential diagnosis for pemphigus foliaceous includes disseminated blastomycosis, hypertrophic lupus erythematosus, sebopsoriasis, and secondary syphilis. Disseminated blastomycosis presents with cutaneous manifestations such as nodules, papules, or pustules evolving over weeks to months into ulcers with subsequent scarring.17 Hypertrophic lupus erythematosus presents with papules and nodules with associated keratotic scaling on the face, palms, and extensor surfaces of the limbs.18 Sebopsoriasis is characterized by well-defined lesions with an overlying scale distributed on the scalp, face, and chest.19 Secondary syphilis presents as early hyperpigmented macules transitioning to acral papulosquamous lesions involving the palms and soles.20
- Hans-Filho G, Aoki V, Hans Bittner NR, et al. Fogo selvagem: endemic pemphigus foliaceus. An Bras Dermatol. 2018;93:638-650.
- Jenson KK, Burr DM, Edwards BC. Case report: reatment of refractory pemphigus foliaceus with rituximab. Practical Dermatology. February 2016:33-36. Accessed August 27, 2021. https://practicaldermatology.com/articles/2016-feb/case-report -treatment-of-refractory-pemphigus-foliaceus-with-rituximab -financial-matters-aad-asds-resources
- Amagai M, Hashimoto T, Green KJ, et al. Antigen-specific immunoadsorption of pathogenic autoantibodies in pemphigus foliaceus. J Invest Dermatol. 1995;104:895-901.
- Mahoney MG, Wang Z, Rothenberger K, et al. Explanations for the clinical and microscopic localization of lesions in pemphigus foliaceus and vulgaris. J Clin Invest. 1999;103:461-468.
- Oktarina DAM, Sokol E, Kramer D, et al. Endocytosis of IgG, desmoglein 1, and plakoglobin in pemphigus foliaceus patient skin. Front Immunol. 2019;10:1-12.
- Kraft M, Worm M. Pemphigus foliaceus-repeated treatment with rituximab 7 years after initial response: a case report. Front Med. 2018;5:315.
- Hale EK. Pemphigus foliaceous. Dermatol Online J. 2002;8:9.
- Tavakolpour S. Pemphigus trigger factors: special focus on pemphigus vulgaris and pemphigus foliaceus. Arch Dermatol Res. 2018;310:95-106.
- A boobaker J, Morar N, Ramdial PK, et al. Pemphigus in South Africa. Int J Dermatol. 2001;40:115-119.
- Austin E, Millsop JW, Ely H, et al. Psoriasiform pemphigus foliaceus in an African American female: an important clinical manifestation. J Drugs Dermatol. 2018;17:471.
- Arbache ST, Nogueira TG, Delgado L, et al. Immunofluorescence testing in the diagnosis of autoimmune blistering diseases: overview of 10-year experience. An Bras Dermatol. 2014;89:885-889.
- Sabolinski ML, Beutner EH, Krasny S, et al. Substrate specificity of antiepithelial antibodies of pemphigus vulgaris and pemphigus foliaceus sera in immunofluorescence tests on monkey and guinea pig esophagus sections. J Invest Dermatol. 1987;88:545-549.
- Palacios-Álvarez I, Riquelme-McLoughlin C, Curto-Barredo L, et al. Rituximab treatment of pemphigus foliaceus: a retrospective study of 12 patients. J Am Acad Dermatol. 2021;85:484-486.
- Murrell DF, Sprecher E. Rituximab and short-course prednisone as the new gold standard for new-onset pemphigus vulgaris and pemphigus foliaceus. Br J Dermatol. 2017;177:1143-1144.
- Gregoriou S, Efthymiou O, Stefanaki C, et al. Management of pemphigus vulgaris: challenges and solutions. Clin Cosmet Investig Dermatol. 2015;8:521-527.
- Saleh MA. A prospective study comparing patients with early and late relapsing pemphigus treated with rituximab. J Am Acad Dermatol. 2018;79:97-103.
- Castillo CG, Kauffman CA, Miceli MH. Blastomycosis. Infect Dis Clin North Am. 2016;30:247-264.
- Herzum A, Gasparini G, Emanuele C, et al. Atypical and rare forms of cutaneous lupus erythematosus: the importance of the diagnosis for the best management of patients. Dermatology. 2013;1-10.
- Tull TJ, Noy M, Bunker CB, et al. Sebopsoriasis in patients with HIV: a case series of 20 patients. Br J Dermatol. 2016; 173:813-815.
- Balagula Y, Mattei P, Wisco OJ, et al. The great imitator revised: the spectrum of atypical cutaneous manifestations of secondary syphilis. Int J Dermatol. 2014;53:1434-1441.
- Hans-Filho G, Aoki V, Hans Bittner NR, et al. Fogo selvagem: endemic pemphigus foliaceus. An Bras Dermatol. 2018;93:638-650.
- Jenson KK, Burr DM, Edwards BC. Case report: reatment of refractory pemphigus foliaceus with rituximab. Practical Dermatology. February 2016:33-36. Accessed August 27, 2021. https://practicaldermatology.com/articles/2016-feb/case-report -treatment-of-refractory-pemphigus-foliaceus-with-rituximab -financial-matters-aad-asds-resources
- Amagai M, Hashimoto T, Green KJ, et al. Antigen-specific immunoadsorption of pathogenic autoantibodies in pemphigus foliaceus. J Invest Dermatol. 1995;104:895-901.
- Mahoney MG, Wang Z, Rothenberger K, et al. Explanations for the clinical and microscopic localization of lesions in pemphigus foliaceus and vulgaris. J Clin Invest. 1999;103:461-468.
- Oktarina DAM, Sokol E, Kramer D, et al. Endocytosis of IgG, desmoglein 1, and plakoglobin in pemphigus foliaceus patient skin. Front Immunol. 2019;10:1-12.
- Kraft M, Worm M. Pemphigus foliaceus-repeated treatment with rituximab 7 years after initial response: a case report. Front Med. 2018;5:315.
- Hale EK. Pemphigus foliaceous. Dermatol Online J. 2002;8:9.
- Tavakolpour S. Pemphigus trigger factors: special focus on pemphigus vulgaris and pemphigus foliaceus. Arch Dermatol Res. 2018;310:95-106.
- A boobaker J, Morar N, Ramdial PK, et al. Pemphigus in South Africa. Int J Dermatol. 2001;40:115-119.
- Austin E, Millsop JW, Ely H, et al. Psoriasiform pemphigus foliaceus in an African American female: an important clinical manifestation. J Drugs Dermatol. 2018;17:471.
- Arbache ST, Nogueira TG, Delgado L, et al. Immunofluorescence testing in the diagnosis of autoimmune blistering diseases: overview of 10-year experience. An Bras Dermatol. 2014;89:885-889.
- Sabolinski ML, Beutner EH, Krasny S, et al. Substrate specificity of antiepithelial antibodies of pemphigus vulgaris and pemphigus foliaceus sera in immunofluorescence tests on monkey and guinea pig esophagus sections. J Invest Dermatol. 1987;88:545-549.
- Palacios-Álvarez I, Riquelme-McLoughlin C, Curto-Barredo L, et al. Rituximab treatment of pemphigus foliaceus: a retrospective study of 12 patients. J Am Acad Dermatol. 2021;85:484-486.
- Murrell DF, Sprecher E. Rituximab and short-course prednisone as the new gold standard for new-onset pemphigus vulgaris and pemphigus foliaceus. Br J Dermatol. 2017;177:1143-1144.
- Gregoriou S, Efthymiou O, Stefanaki C, et al. Management of pemphigus vulgaris: challenges and solutions. Clin Cosmet Investig Dermatol. 2015;8:521-527.
- Saleh MA. A prospective study comparing patients with early and late relapsing pemphigus treated with rituximab. J Am Acad Dermatol. 2018;79:97-103.
- Castillo CG, Kauffman CA, Miceli MH. Blastomycosis. Infect Dis Clin North Am. 2016;30:247-264.
- Herzum A, Gasparini G, Emanuele C, et al. Atypical and rare forms of cutaneous lupus erythematosus: the importance of the diagnosis for the best management of patients. Dermatology. 2013;1-10.
- Tull TJ, Noy M, Bunker CB, et al. Sebopsoriasis in patients with HIV: a case series of 20 patients. Br J Dermatol. 2016; 173:813-815.
- Balagula Y, Mattei P, Wisco OJ, et al. The great imitator revised: the spectrum of atypical cutaneous manifestations of secondary syphilis. Int J Dermatol. 2014;53:1434-1441.
A 40-year-old Black man presented for evaluation of a thick plaque throughout the scalp (top), scaly plaques on the cheeks (bottom), and a spreading rash on the trunk that had progressed over the last few months. He had no relevant medical history, took no medications, and was in a monogamous relationship with a female partner. He previously saw an outside dermatologist who gave him triamcinolone cream, which was mildly helpful. Physical examination revealed a thick verrucous plaque throughout the scalp extending onto the forehead; thick plaques on the cheeks; and numerous, thinly eroded lesions on the trunk. Biopsies and a laboratory workup were performed.
Cutaneous Protothecosis
To the Editor:
Protothecosis infections are caused by an achlorophyllic algae of the species Prototheca. Prototheca organisms are found mostly in soil and water.1 Human infections are rare and involve 2 species, Prototheca wickerhamii and Prototheca zopfii. The former most commonly is responsible for human infections, though P zopfii results in more serious systemic infections with a poor prognosis. There are various types of Prototheca infection presentations, with a 2007 review of 117 cases reporting that cutaneous infections are most common (66%), followed by systemic infections (19%), and olecranon bursitis (15%).2 Skin lesions most commonly occur on the extremities and face, and they present as vesiculobullous and ulcerative lesions with purulent drainage. The skin lesions also may appear as erythematous plaques or nodules, subcutaneous papules, verrucous or herpetiformis lesions, or pyogenic granuloma–like lesions.3 Protothecosis typically affects immunocompromised individuals, especially those with a history of chronic corticosteroid use, malignancy, diabetes mellitus, AIDS, and/or organ transplant.1 We present a case of cutaneous protothecosis on the dorsal distal extremity of a 94-year-old woman. History of exposure to soil while gardening was elicited from the patient, and no immunosuppressive history was present aside from the patient’s age. This case may prompt workup for malignancy or immunosuppression in this patient subset.
A 94-year-old woman with a medical history of cutaneous squamous cell carcinoma (SCC) presented with a growing lesion on the dorsal surface of the left fourth digit of 2 months’ duration. The patient reported the lesion was painful, and she noted preceding trauma to the area that was suspected to have occurred while gardening. Physical examination revealed an ulcerated, hypertrophic, erythematous nodule on the dorsal surface of the left fourth metacarpophalangeal joint. The differential diagnosis included SCC, inflamed cyst, verruca vulgaris, and orf virus due to the clinical presentation. A shave biopsy was performed, and the lesion subsequently was treated with electrodesiccation and curettage.
Histopathologic evaluation revealed pseudoepitheliomatous hyperplasia with a mixed inflammatory infiltrate including lymphocytes and histiocytes. A morula within the dermis was characteristic of a protothecosis infection (Figure 1). On follow-up visit 6 weeks later, the lesion had grown back to its original size and morphology (Figure 2). At this time, the lesion was again treated with shave removal, followed by electrodesiccation and curettage, and the patient was placed on oral fluconazole 200 mg daily for 1 month. When the lesion did not resolve with fluconazole, she was referred to infectious disease as well as general surgery for surgical removal and debridement of the lesion. Unfortunately, the patient was lost to follow-up.
Protothecosis is an infectious disease comprised of achlorophyllic algae found in soil and water that rarely affects humans. When it does affect humans, cutaneous infections are most common. All human cases in which organisms were identified to species level have been caused by P wickerhamii or P zopfii species.2 Inoculation is suspected to occur through trauma to affected skin, especially when in the context of contaminated water. Our patient reported history of trauma to the hand, with soil from gardening as the potential aquagenic source of the infection.
The clinical presentation of protothecosis ranges from localized cutaneous to disseminated systemic infections, with most reported cases of systemic disease occurring in immunocompromised individuals. The cutaneous lesions of protothecosis vary greatly in clinical appearance including ulcerative nodules (as in our case), papules, plaques, pustules, and vesicles with erosion or crusting.4
Cutaneous protothecosis has the potential to mimic many other skin diseases and lesions, and, given its rarity, it may not be on the radar of dermatologists. Our patient’s lesion was presumed to be a skin cancer and was treated as such because of the history of SCC and clinical presentation. Although excision of individual lesions of protothecosis can be curative, electrodesiccation and curettage does not appear to be an adequate treatment, as the lesion subsequently recurred. It also is possible that this case represents P zopfii infection, as it did not respond to treatment with oral fluconazole, though in vitro studies with fluconazole to both P zopfii and P wickerhamii had variable treatment success.2 Also, the histopathologic findings were most consistent with P wickerhamii, revealing small, round, symmetrical morula, compared to P zopfii, which typically will display oval or cylindrical, asymmetrical, random internal segmentation.5 This case may warrant determination of species, which can be accomplished by a culture on Sabouraud dextrose agar, carbohydrate and alcohol assimilation test, yeast biochemical card, serological typing by immunoblotting, immunofluorescence study using species-specific antibodies, or amplification by polymerase chain reaction for small subunit ribosomal DNA sequences.2,6-8
The natural history of isolated skin disease is an indolent progressive course; however, reports do exist noting spontaneous resolution.4,9 Treatment options for Prototheca infections can be disappointing and consist of both surgical and medical management, or a combination of the 2 approaches. Reports in the literature support the use of antifungals including ketoconazole, voriconazole, itraconazole, fluconazole, and amphotericin B, with the latter displaying the best activity against Prototheca species.2 Tetracycline has been used in combination with oral or topical amphotericin B and was found to be synergistic in vitro and in case reports at successfully treating cutaneous protothecosis infections. It is possible that our patient was not treated with fluconazole long enough for it to become therapeutic, as most reported treatment regimens are weeks to months in length. Conversely, it may have been of benefit to transition the patient to topical amphotericin B and tetracycline, as fluconazole failed in this patient. However, treatment successes and failures are limited to case reports/case series and in vitro studies, with prospective studies lacking. Due to the variability with in vitro susceptibility profiles for Prototheca species, it generally is not recommended to pursue in vitro susceptibility testing in the management of Prototheca skin infections due to the inconsistency demonstrated between in vitro activity and clinical response to therapy.2
- Silva PC, Costa e Silva SB, Lima RB, et al. Cutaneous protothecosis—case report. An Bras Dermatol. 2013;88:183-185.
- Lass-Flörl C, Mayr A. Human protothecosis. Clin Microbiol Rev. 2007;20:230-242.
- Seok JY, Lee Y, Lee H, et al. Human cutaneous protothecosis: report of a case and literature review. Korean J Pathol. 2013;47:575-578.
- Mayorga J, Barba-Gómez JF, Verduzco-Martínez AP, et al. Protothecosis. Clin Dermatol. 2012;30:432-436.
- Walsh SV, Johnson RA, Tahan SR. Protothecosis: an unusual cause of chronic subcutaneous and soft tissue infection. Am J Dermatopathol. 1998;20:379-382.
- Casal MJ, Gutierrez J. Simple new test for rapid differentiation of Prototheca wickerhamii from Prototheca zopfii. J Clin Microbiol. 1983;18:992-993.
- Arnold, P, Ahearn, DG. The systematics of the genus Prototheca with a description of a new species P. filamenta. Mycologia 1972;64:265-275.
- Roesler U, Scholz H, Hensel H. Emended phenotypic characterization of Prototheca zopfii: a proposal for three biotypes and standards for their identification. Int J Syst Evol Microbiol. 2003;53:1195-1199.
- Todd JR, King JW, Oberle A, et al. Protothecosis: report of a case with 20-year follow-up, and review of previously published cases. Med Mycol. 2012;50:673-689.
To the Editor:
Protothecosis infections are caused by an achlorophyllic algae of the species Prototheca. Prototheca organisms are found mostly in soil and water.1 Human infections are rare and involve 2 species, Prototheca wickerhamii and Prototheca zopfii. The former most commonly is responsible for human infections, though P zopfii results in more serious systemic infections with a poor prognosis. There are various types of Prototheca infection presentations, with a 2007 review of 117 cases reporting that cutaneous infections are most common (66%), followed by systemic infections (19%), and olecranon bursitis (15%).2 Skin lesions most commonly occur on the extremities and face, and they present as vesiculobullous and ulcerative lesions with purulent drainage. The skin lesions also may appear as erythematous plaques or nodules, subcutaneous papules, verrucous or herpetiformis lesions, or pyogenic granuloma–like lesions.3 Protothecosis typically affects immunocompromised individuals, especially those with a history of chronic corticosteroid use, malignancy, diabetes mellitus, AIDS, and/or organ transplant.1 We present a case of cutaneous protothecosis on the dorsal distal extremity of a 94-year-old woman. History of exposure to soil while gardening was elicited from the patient, and no immunosuppressive history was present aside from the patient’s age. This case may prompt workup for malignancy or immunosuppression in this patient subset.
A 94-year-old woman with a medical history of cutaneous squamous cell carcinoma (SCC) presented with a growing lesion on the dorsal surface of the left fourth digit of 2 months’ duration. The patient reported the lesion was painful, and she noted preceding trauma to the area that was suspected to have occurred while gardening. Physical examination revealed an ulcerated, hypertrophic, erythematous nodule on the dorsal surface of the left fourth metacarpophalangeal joint. The differential diagnosis included SCC, inflamed cyst, verruca vulgaris, and orf virus due to the clinical presentation. A shave biopsy was performed, and the lesion subsequently was treated with electrodesiccation and curettage.
Histopathologic evaluation revealed pseudoepitheliomatous hyperplasia with a mixed inflammatory infiltrate including lymphocytes and histiocytes. A morula within the dermis was characteristic of a protothecosis infection (Figure 1). On follow-up visit 6 weeks later, the lesion had grown back to its original size and morphology (Figure 2). At this time, the lesion was again treated with shave removal, followed by electrodesiccation and curettage, and the patient was placed on oral fluconazole 200 mg daily for 1 month. When the lesion did not resolve with fluconazole, she was referred to infectious disease as well as general surgery for surgical removal and debridement of the lesion. Unfortunately, the patient was lost to follow-up.
Protothecosis is an infectious disease comprised of achlorophyllic algae found in soil and water that rarely affects humans. When it does affect humans, cutaneous infections are most common. All human cases in which organisms were identified to species level have been caused by P wickerhamii or P zopfii species.2 Inoculation is suspected to occur through trauma to affected skin, especially when in the context of contaminated water. Our patient reported history of trauma to the hand, with soil from gardening as the potential aquagenic source of the infection.
The clinical presentation of protothecosis ranges from localized cutaneous to disseminated systemic infections, with most reported cases of systemic disease occurring in immunocompromised individuals. The cutaneous lesions of protothecosis vary greatly in clinical appearance including ulcerative nodules (as in our case), papules, plaques, pustules, and vesicles with erosion or crusting.4
Cutaneous protothecosis has the potential to mimic many other skin diseases and lesions, and, given its rarity, it may not be on the radar of dermatologists. Our patient’s lesion was presumed to be a skin cancer and was treated as such because of the history of SCC and clinical presentation. Although excision of individual lesions of protothecosis can be curative, electrodesiccation and curettage does not appear to be an adequate treatment, as the lesion subsequently recurred. It also is possible that this case represents P zopfii infection, as it did not respond to treatment with oral fluconazole, though in vitro studies with fluconazole to both P zopfii and P wickerhamii had variable treatment success.2 Also, the histopathologic findings were most consistent with P wickerhamii, revealing small, round, symmetrical morula, compared to P zopfii, which typically will display oval or cylindrical, asymmetrical, random internal segmentation.5 This case may warrant determination of species, which can be accomplished by a culture on Sabouraud dextrose agar, carbohydrate and alcohol assimilation test, yeast biochemical card, serological typing by immunoblotting, immunofluorescence study using species-specific antibodies, or amplification by polymerase chain reaction for small subunit ribosomal DNA sequences.2,6-8
The natural history of isolated skin disease is an indolent progressive course; however, reports do exist noting spontaneous resolution.4,9 Treatment options for Prototheca infections can be disappointing and consist of both surgical and medical management, or a combination of the 2 approaches. Reports in the literature support the use of antifungals including ketoconazole, voriconazole, itraconazole, fluconazole, and amphotericin B, with the latter displaying the best activity against Prototheca species.2 Tetracycline has been used in combination with oral or topical amphotericin B and was found to be synergistic in vitro and in case reports at successfully treating cutaneous protothecosis infections. It is possible that our patient was not treated with fluconazole long enough for it to become therapeutic, as most reported treatment regimens are weeks to months in length. Conversely, it may have been of benefit to transition the patient to topical amphotericin B and tetracycline, as fluconazole failed in this patient. However, treatment successes and failures are limited to case reports/case series and in vitro studies, with prospective studies lacking. Due to the variability with in vitro susceptibility profiles for Prototheca species, it generally is not recommended to pursue in vitro susceptibility testing in the management of Prototheca skin infections due to the inconsistency demonstrated between in vitro activity and clinical response to therapy.2
To the Editor:
Protothecosis infections are caused by an achlorophyllic algae of the species Prototheca. Prototheca organisms are found mostly in soil and water.1 Human infections are rare and involve 2 species, Prototheca wickerhamii and Prototheca zopfii. The former most commonly is responsible for human infections, though P zopfii results in more serious systemic infections with a poor prognosis. There are various types of Prototheca infection presentations, with a 2007 review of 117 cases reporting that cutaneous infections are most common (66%), followed by systemic infections (19%), and olecranon bursitis (15%).2 Skin lesions most commonly occur on the extremities and face, and they present as vesiculobullous and ulcerative lesions with purulent drainage. The skin lesions also may appear as erythematous plaques or nodules, subcutaneous papules, verrucous or herpetiformis lesions, or pyogenic granuloma–like lesions.3 Protothecosis typically affects immunocompromised individuals, especially those with a history of chronic corticosteroid use, malignancy, diabetes mellitus, AIDS, and/or organ transplant.1 We present a case of cutaneous protothecosis on the dorsal distal extremity of a 94-year-old woman. History of exposure to soil while gardening was elicited from the patient, and no immunosuppressive history was present aside from the patient’s age. This case may prompt workup for malignancy or immunosuppression in this patient subset.
A 94-year-old woman with a medical history of cutaneous squamous cell carcinoma (SCC) presented with a growing lesion on the dorsal surface of the left fourth digit of 2 months’ duration. The patient reported the lesion was painful, and she noted preceding trauma to the area that was suspected to have occurred while gardening. Physical examination revealed an ulcerated, hypertrophic, erythematous nodule on the dorsal surface of the left fourth metacarpophalangeal joint. The differential diagnosis included SCC, inflamed cyst, verruca vulgaris, and orf virus due to the clinical presentation. A shave biopsy was performed, and the lesion subsequently was treated with electrodesiccation and curettage.
Histopathologic evaluation revealed pseudoepitheliomatous hyperplasia with a mixed inflammatory infiltrate including lymphocytes and histiocytes. A morula within the dermis was characteristic of a protothecosis infection (Figure 1). On follow-up visit 6 weeks later, the lesion had grown back to its original size and morphology (Figure 2). At this time, the lesion was again treated with shave removal, followed by electrodesiccation and curettage, and the patient was placed on oral fluconazole 200 mg daily for 1 month. When the lesion did not resolve with fluconazole, she was referred to infectious disease as well as general surgery for surgical removal and debridement of the lesion. Unfortunately, the patient was lost to follow-up.
Protothecosis is an infectious disease comprised of achlorophyllic algae found in soil and water that rarely affects humans. When it does affect humans, cutaneous infections are most common. All human cases in which organisms were identified to species level have been caused by P wickerhamii or P zopfii species.2 Inoculation is suspected to occur through trauma to affected skin, especially when in the context of contaminated water. Our patient reported history of trauma to the hand, with soil from gardening as the potential aquagenic source of the infection.
The clinical presentation of protothecosis ranges from localized cutaneous to disseminated systemic infections, with most reported cases of systemic disease occurring in immunocompromised individuals. The cutaneous lesions of protothecosis vary greatly in clinical appearance including ulcerative nodules (as in our case), papules, plaques, pustules, and vesicles with erosion or crusting.4
Cutaneous protothecosis has the potential to mimic many other skin diseases and lesions, and, given its rarity, it may not be on the radar of dermatologists. Our patient’s lesion was presumed to be a skin cancer and was treated as such because of the history of SCC and clinical presentation. Although excision of individual lesions of protothecosis can be curative, electrodesiccation and curettage does not appear to be an adequate treatment, as the lesion subsequently recurred. It also is possible that this case represents P zopfii infection, as it did not respond to treatment with oral fluconazole, though in vitro studies with fluconazole to both P zopfii and P wickerhamii had variable treatment success.2 Also, the histopathologic findings were most consistent with P wickerhamii, revealing small, round, symmetrical morula, compared to P zopfii, which typically will display oval or cylindrical, asymmetrical, random internal segmentation.5 This case may warrant determination of species, which can be accomplished by a culture on Sabouraud dextrose agar, carbohydrate and alcohol assimilation test, yeast biochemical card, serological typing by immunoblotting, immunofluorescence study using species-specific antibodies, or amplification by polymerase chain reaction for small subunit ribosomal DNA sequences.2,6-8
The natural history of isolated skin disease is an indolent progressive course; however, reports do exist noting spontaneous resolution.4,9 Treatment options for Prototheca infections can be disappointing and consist of both surgical and medical management, or a combination of the 2 approaches. Reports in the literature support the use of antifungals including ketoconazole, voriconazole, itraconazole, fluconazole, and amphotericin B, with the latter displaying the best activity against Prototheca species.2 Tetracycline has been used in combination with oral or topical amphotericin B and was found to be synergistic in vitro and in case reports at successfully treating cutaneous protothecosis infections. It is possible that our patient was not treated with fluconazole long enough for it to become therapeutic, as most reported treatment regimens are weeks to months in length. Conversely, it may have been of benefit to transition the patient to topical amphotericin B and tetracycline, as fluconazole failed in this patient. However, treatment successes and failures are limited to case reports/case series and in vitro studies, with prospective studies lacking. Due to the variability with in vitro susceptibility profiles for Prototheca species, it generally is not recommended to pursue in vitro susceptibility testing in the management of Prototheca skin infections due to the inconsistency demonstrated between in vitro activity and clinical response to therapy.2
- Silva PC, Costa e Silva SB, Lima RB, et al. Cutaneous protothecosis—case report. An Bras Dermatol. 2013;88:183-185.
- Lass-Flörl C, Mayr A. Human protothecosis. Clin Microbiol Rev. 2007;20:230-242.
- Seok JY, Lee Y, Lee H, et al. Human cutaneous protothecosis: report of a case and literature review. Korean J Pathol. 2013;47:575-578.
- Mayorga J, Barba-Gómez JF, Verduzco-Martínez AP, et al. Protothecosis. Clin Dermatol. 2012;30:432-436.
- Walsh SV, Johnson RA, Tahan SR. Protothecosis: an unusual cause of chronic subcutaneous and soft tissue infection. Am J Dermatopathol. 1998;20:379-382.
- Casal MJ, Gutierrez J. Simple new test for rapid differentiation of Prototheca wickerhamii from Prototheca zopfii. J Clin Microbiol. 1983;18:992-993.
- Arnold, P, Ahearn, DG. The systematics of the genus Prototheca with a description of a new species P. filamenta. Mycologia 1972;64:265-275.
- Roesler U, Scholz H, Hensel H. Emended phenotypic characterization of Prototheca zopfii: a proposal for three biotypes and standards for their identification. Int J Syst Evol Microbiol. 2003;53:1195-1199.
- Todd JR, King JW, Oberle A, et al. Protothecosis: report of a case with 20-year follow-up, and review of previously published cases. Med Mycol. 2012;50:673-689.
- Silva PC, Costa e Silva SB, Lima RB, et al. Cutaneous protothecosis—case report. An Bras Dermatol. 2013;88:183-185.
- Lass-Flörl C, Mayr A. Human protothecosis. Clin Microbiol Rev. 2007;20:230-242.
- Seok JY, Lee Y, Lee H, et al. Human cutaneous protothecosis: report of a case and literature review. Korean J Pathol. 2013;47:575-578.
- Mayorga J, Barba-Gómez JF, Verduzco-Martínez AP, et al. Protothecosis. Clin Dermatol. 2012;30:432-436.
- Walsh SV, Johnson RA, Tahan SR. Protothecosis: an unusual cause of chronic subcutaneous and soft tissue infection. Am J Dermatopathol. 1998;20:379-382.
- Casal MJ, Gutierrez J. Simple new test for rapid differentiation of Prototheca wickerhamii from Prototheca zopfii. J Clin Microbiol. 1983;18:992-993.
- Arnold, P, Ahearn, DG. The systematics of the genus Prototheca with a description of a new species P. filamenta. Mycologia 1972;64:265-275.
- Roesler U, Scholz H, Hensel H. Emended phenotypic characterization of Prototheca zopfii: a proposal for three biotypes and standards for their identification. Int J Syst Evol Microbiol. 2003;53:1195-1199.
- Todd JR, King JW, Oberle A, et al. Protothecosis: report of a case with 20-year follow-up, and review of previously published cases. Med Mycol. 2012;50:673-689.
Practice Points
- Cutaneous protothecosis is a rare skin infection most commonly reported in immunocompromised individuals with recent exposure to contaminated soil or water. Cutaneous protothecosis has the potential to mimic many other skin diseases and lesions, including eczema; nonmelanoma skin cancer; or bacterial, viral, and fungal skin infections.
- A skin biopsy is essential for diagnosis, and histopathology is characteristic with soccer ball–appearing morula noted in a mixed inflammatory infiltrate.