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.

Top Sections
Product Review
Coding
Dermpath Diagnosis
For Residents
Photo Challenge
Tips
ct
Main menu
CUTIS Main Menu
Explore menu
CUTIS Explore Menu
Proclivity ID
18823001
Unpublish
Negative Keywords
ammunition
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
Negative Keywords Excluded Elements
div[contains(@class, 'alert ad-blocker')]
section[contains(@class, 'nav-hidden')]
section[contains(@class, 'nav-hidden active')
Altmetric
DSM Affiliated
Display in offset block
Disqus Exclude
Best Practices
CE/CME
Education Center
Medical Education Library
Enable Disqus
Display Author and Disclosure Link
Publication Type
Clinical
Slot System
Featured Buckets
Disable Sticky Ads
Disable Ad Block Mitigation
Featured Buckets Admin
Show Ads on this Publication's Homepage
Consolidated Pub
Show Article Page Numbers on TOC
Expire Announcement Bar
Thu, 08/01/2024 - 08:10
Use larger logo size
Off
publication_blueconic_enabled
Off
Show More Destinations Menu
Disable Adhesion on Publication
Off
Restore Menu Label on Mobile Navigation
Disable Facebook Pixel from Publication
Exclude this publication from publication selection on articles and quiz
Challenge Center
Disable Inline Native ads
survey writer start date
Thu, 08/01/2024 - 08:10
Current Issue
Title
Cutis
Description

A peer-reviewed, indexed journal for dermatologists with original research, image quizzes, cases and reviews, and columns.

Current Issue Reference

Navigating Hair Loss in Medical School: Experiences of 2 Young Black Women

Article Type
Changed
Thu, 12/21/2023 - 10:38
Display Headline
Navigating Hair Loss in Medical School: Experiences of 2 Young Black Women

As medical students, we often assume we are exempt from the diagnoses we learn about. During the first 2 years of medical school, we learn about alopecia as a condition that may be associated with stress, hormonal imbalances, nutrient deficiencies, and aging. However, our curricula do not explore the subtypes, psychosocial impact, or even the overwhelming number of Black women who are disproportionately affected by alopecia. For Black women, hair is a colossal part of their cultural identity, learning from a young age how to nurture and style natural coils. It becomes devastating when women begin to lose them. 

The diagnosis of alopecia subtypes in Black women has been explored in the literature; however, understanding the unique experiences of young Black women is an important part of patient care, as alopecia often is destructive to the patient’s self-image. Therefore, it is important to shed light on these experiences so others feel empowered and supported in their journeys. Herein, we share the experiences of 2 authors (J.D. and C.A.V.O.)—both young Black women—who navigated unexpected hair loss in medical school.

Jewell’s Story

During my first year of medical school, I noticed my hair was shedding more than usual, and my ponytail was not as thick as it once was. I also had an area in my crown that was abnormally thin. My parents suggested that it was a consequence of stress, but I knew something was not right. With only 1 Black dermatologist within 2 hours of Nashville, Tennessee, I remember worrying about seeing a dermatologist who did not understand Black hair. I still scheduled an appointment, but I remember debating if I should straighten my hair or wear my naturally curly Afro. The first dermatologist I saw diagnosed me with seborrheic dermatitis—without even examining my scalp. She told me that I had a “full head of hair” and that I had nothing to worry about. I was unconvinced. Weeks later, I met with another dermatologist who took the time to listen to my concerns. After a scalp biopsy and laboratory work, she diagnosed me with telogen effluvium and androgenetic alopecia. Months later, I had the opportunity to visit the Black dermatologist, and she diagnosed me with central centrifugal cicatricial alopecia. I am grateful for the earlier dermatologists I saw, but I finally feel at ease with my diagnosis and treatment plan after being seen by the latter.

Chidubem’s Story 

From a young age, I was conditioned to think my hair was thick, unmanageable, and a nuisance. I grew accustomed to people yanking on my hair, and my gentle whispers of “this hurts” and “the braid is too tight” being ignored. That continued into adulthood. While studying for the US Medical Licensing Examination, I noticed a burning sensation on my scalp. I decided to ignore it. However, as the days progressed, the slight burning sensation turned into intense burning and itching. I still ignored it. Not only did I lack the funds for a dermatology appointment, but my licensing examination was approaching, and it was more important than anything related to my hair. After the examination, I eventually made an appointment with my primary care physician, who attributed my symptoms to the stressors of medical school. “I think you are having migraines,” she told me. So, I continued to ignore my symptoms. A year passed, and a hair braider pointed out that I had 2 well-defined bald patches on my scalp. I remember feeling angry and confused as to how I missed those findings. I could no longer ignore it—it bothered me less when no one else knew about it. I quickly made a dermatology appointment. Although I opted out of a biopsy, we decided to treat my hair loss empirically, and I have experienced drastic improvement.

Final Thoughts

We are 2 Black women living more than 500 miles away from each other at different medical institutions, yet we share the same experience, which many other women unfortunately face alone. It is not uncommon for us to feel unheard, dismissed, or misdiagnosed. We write this for the Black woman sorting through the feelings of confusion and shock as she traces the hairless spot on her scalp. We write this for the medical student ignoring their symptoms until after their examination. We even write this for any nondermatologists uncomfortable with diagnosing and treating textured hair. To improve patient satisfaction and overall health outcomes, physicians must approach patients with both knowledge and cultural competency. Most importantly, dermatologists (and other physicians) should be appropriately trained in not only the structural differences of textured hair but also the unique practices and beliefs among Black women in relation to their hair.

Acknowledgments—Jewell Dinkins is the inaugural recipient of the Janssen–Skin of Color Research Fellowship at Howard University (Washington, DC), and Chidubem A.V. Okeke is the inaugural recipient of the Women’s Dermatologic Society–La Roche-Posay dermatology fellowship at Howard University.

Article PDF
Author and Disclosure Information

Jewell Dinkins and Chidubem A.V. Okeke are from the Howard University College of Medicine, Washington, DC. Jewell Dinkins also is from Meharry Medical College, Nashville, Tennessee. Drs. Byrd and Frey are from the Department of Dermatology, Howard University, Washington, DC.

Jewell Dinkins and Chidubem A.V. Okeke report no conflict of interest. Dr. Byrd is a consultant for Senté Inc and Sonoma Biotherapeutics and is on the advisory board for Novartis. Dr. Frey is a consultant for and has received consultancy fees from Avita Medical, Benev, Ferndale Pharma, Galderma Laboratories, L’Oreal, Nutrafol, Nutraceutical Wellness Inc, Procter & Gamble, Regeneron, and Sun Pharma.

Correspondence: Jewell Dinkins, MS, Janssen–Skin of Color Research Fellow, Meharry Medical College, 1005 Dr. D.B. Todd Jr Blvd, Nashville, TN 37208 (jdinkins20@email.mmc.edu).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E30-E31
Sections
Author and Disclosure Information

Jewell Dinkins and Chidubem A.V. Okeke are from the Howard University College of Medicine, Washington, DC. Jewell Dinkins also is from Meharry Medical College, Nashville, Tennessee. Drs. Byrd and Frey are from the Department of Dermatology, Howard University, Washington, DC.

Jewell Dinkins and Chidubem A.V. Okeke report no conflict of interest. Dr. Byrd is a consultant for Senté Inc and Sonoma Biotherapeutics and is on the advisory board for Novartis. Dr. Frey is a consultant for and has received consultancy fees from Avita Medical, Benev, Ferndale Pharma, Galderma Laboratories, L’Oreal, Nutrafol, Nutraceutical Wellness Inc, Procter & Gamble, Regeneron, and Sun Pharma.

Correspondence: Jewell Dinkins, MS, Janssen–Skin of Color Research Fellow, Meharry Medical College, 1005 Dr. D.B. Todd Jr Blvd, Nashville, TN 37208 (jdinkins20@email.mmc.edu).

Author and Disclosure Information

Jewell Dinkins and Chidubem A.V. Okeke are from the Howard University College of Medicine, Washington, DC. Jewell Dinkins also is from Meharry Medical College, Nashville, Tennessee. Drs. Byrd and Frey are from the Department of Dermatology, Howard University, Washington, DC.

Jewell Dinkins and Chidubem A.V. Okeke report no conflict of interest. Dr. Byrd is a consultant for Senté Inc and Sonoma Biotherapeutics and is on the advisory board for Novartis. Dr. Frey is a consultant for and has received consultancy fees from Avita Medical, Benev, Ferndale Pharma, Galderma Laboratories, L’Oreal, Nutrafol, Nutraceutical Wellness Inc, Procter & Gamble, Regeneron, and Sun Pharma.

Correspondence: Jewell Dinkins, MS, Janssen–Skin of Color Research Fellow, Meharry Medical College, 1005 Dr. D.B. Todd Jr Blvd, Nashville, TN 37208 (jdinkins20@email.mmc.edu).

Article PDF
Article PDF

As medical students, we often assume we are exempt from the diagnoses we learn about. During the first 2 years of medical school, we learn about alopecia as a condition that may be associated with stress, hormonal imbalances, nutrient deficiencies, and aging. However, our curricula do not explore the subtypes, psychosocial impact, or even the overwhelming number of Black women who are disproportionately affected by alopecia. For Black women, hair is a colossal part of their cultural identity, learning from a young age how to nurture and style natural coils. It becomes devastating when women begin to lose them. 

The diagnosis of alopecia subtypes in Black women has been explored in the literature; however, understanding the unique experiences of young Black women is an important part of patient care, as alopecia often is destructive to the patient’s self-image. Therefore, it is important to shed light on these experiences so others feel empowered and supported in their journeys. Herein, we share the experiences of 2 authors (J.D. and C.A.V.O.)—both young Black women—who navigated unexpected hair loss in medical school.

Jewell’s Story

During my first year of medical school, I noticed my hair was shedding more than usual, and my ponytail was not as thick as it once was. I also had an area in my crown that was abnormally thin. My parents suggested that it was a consequence of stress, but I knew something was not right. With only 1 Black dermatologist within 2 hours of Nashville, Tennessee, I remember worrying about seeing a dermatologist who did not understand Black hair. I still scheduled an appointment, but I remember debating if I should straighten my hair or wear my naturally curly Afro. The first dermatologist I saw diagnosed me with seborrheic dermatitis—without even examining my scalp. She told me that I had a “full head of hair” and that I had nothing to worry about. I was unconvinced. Weeks later, I met with another dermatologist who took the time to listen to my concerns. After a scalp biopsy and laboratory work, she diagnosed me with telogen effluvium and androgenetic alopecia. Months later, I had the opportunity to visit the Black dermatologist, and she diagnosed me with central centrifugal cicatricial alopecia. I am grateful for the earlier dermatologists I saw, but I finally feel at ease with my diagnosis and treatment plan after being seen by the latter.

Chidubem’s Story 

From a young age, I was conditioned to think my hair was thick, unmanageable, and a nuisance. I grew accustomed to people yanking on my hair, and my gentle whispers of “this hurts” and “the braid is too tight” being ignored. That continued into adulthood. While studying for the US Medical Licensing Examination, I noticed a burning sensation on my scalp. I decided to ignore it. However, as the days progressed, the slight burning sensation turned into intense burning and itching. I still ignored it. Not only did I lack the funds for a dermatology appointment, but my licensing examination was approaching, and it was more important than anything related to my hair. After the examination, I eventually made an appointment with my primary care physician, who attributed my symptoms to the stressors of medical school. “I think you are having migraines,” she told me. So, I continued to ignore my symptoms. A year passed, and a hair braider pointed out that I had 2 well-defined bald patches on my scalp. I remember feeling angry and confused as to how I missed those findings. I could no longer ignore it—it bothered me less when no one else knew about it. I quickly made a dermatology appointment. Although I opted out of a biopsy, we decided to treat my hair loss empirically, and I have experienced drastic improvement.

Final Thoughts

We are 2 Black women living more than 500 miles away from each other at different medical institutions, yet we share the same experience, which many other women unfortunately face alone. It is not uncommon for us to feel unheard, dismissed, or misdiagnosed. We write this for the Black woman sorting through the feelings of confusion and shock as she traces the hairless spot on her scalp. We write this for the medical student ignoring their symptoms until after their examination. We even write this for any nondermatologists uncomfortable with diagnosing and treating textured hair. To improve patient satisfaction and overall health outcomes, physicians must approach patients with both knowledge and cultural competency. Most importantly, dermatologists (and other physicians) should be appropriately trained in not only the structural differences of textured hair but also the unique practices and beliefs among Black women in relation to their hair.

Acknowledgments—Jewell Dinkins is the inaugural recipient of the Janssen–Skin of Color Research Fellowship at Howard University (Washington, DC), and Chidubem A.V. Okeke is the inaugural recipient of the Women’s Dermatologic Society–La Roche-Posay dermatology fellowship at Howard University.

As medical students, we often assume we are exempt from the diagnoses we learn about. During the first 2 years of medical school, we learn about alopecia as a condition that may be associated with stress, hormonal imbalances, nutrient deficiencies, and aging. However, our curricula do not explore the subtypes, psychosocial impact, or even the overwhelming number of Black women who are disproportionately affected by alopecia. For Black women, hair is a colossal part of their cultural identity, learning from a young age how to nurture and style natural coils. It becomes devastating when women begin to lose them. 

The diagnosis of alopecia subtypes in Black women has been explored in the literature; however, understanding the unique experiences of young Black women is an important part of patient care, as alopecia often is destructive to the patient’s self-image. Therefore, it is important to shed light on these experiences so others feel empowered and supported in their journeys. Herein, we share the experiences of 2 authors (J.D. and C.A.V.O.)—both young Black women—who navigated unexpected hair loss in medical school.

Jewell’s Story

During my first year of medical school, I noticed my hair was shedding more than usual, and my ponytail was not as thick as it once was. I also had an area in my crown that was abnormally thin. My parents suggested that it was a consequence of stress, but I knew something was not right. With only 1 Black dermatologist within 2 hours of Nashville, Tennessee, I remember worrying about seeing a dermatologist who did not understand Black hair. I still scheduled an appointment, but I remember debating if I should straighten my hair or wear my naturally curly Afro. The first dermatologist I saw diagnosed me with seborrheic dermatitis—without even examining my scalp. She told me that I had a “full head of hair” and that I had nothing to worry about. I was unconvinced. Weeks later, I met with another dermatologist who took the time to listen to my concerns. After a scalp biopsy and laboratory work, she diagnosed me with telogen effluvium and androgenetic alopecia. Months later, I had the opportunity to visit the Black dermatologist, and she diagnosed me with central centrifugal cicatricial alopecia. I am grateful for the earlier dermatologists I saw, but I finally feel at ease with my diagnosis and treatment plan after being seen by the latter.

Chidubem’s Story 

From a young age, I was conditioned to think my hair was thick, unmanageable, and a nuisance. I grew accustomed to people yanking on my hair, and my gentle whispers of “this hurts” and “the braid is too tight” being ignored. That continued into adulthood. While studying for the US Medical Licensing Examination, I noticed a burning sensation on my scalp. I decided to ignore it. However, as the days progressed, the slight burning sensation turned into intense burning and itching. I still ignored it. Not only did I lack the funds for a dermatology appointment, but my licensing examination was approaching, and it was more important than anything related to my hair. After the examination, I eventually made an appointment with my primary care physician, who attributed my symptoms to the stressors of medical school. “I think you are having migraines,” she told me. So, I continued to ignore my symptoms. A year passed, and a hair braider pointed out that I had 2 well-defined bald patches on my scalp. I remember feeling angry and confused as to how I missed those findings. I could no longer ignore it—it bothered me less when no one else knew about it. I quickly made a dermatology appointment. Although I opted out of a biopsy, we decided to treat my hair loss empirically, and I have experienced drastic improvement.

Final Thoughts

We are 2 Black women living more than 500 miles away from each other at different medical institutions, yet we share the same experience, which many other women unfortunately face alone. It is not uncommon for us to feel unheard, dismissed, or misdiagnosed. We write this for the Black woman sorting through the feelings of confusion and shock as she traces the hairless spot on her scalp. We write this for the medical student ignoring their symptoms until after their examination. We even write this for any nondermatologists uncomfortable with diagnosing and treating textured hair. To improve patient satisfaction and overall health outcomes, physicians must approach patients with both knowledge and cultural competency. Most importantly, dermatologists (and other physicians) should be appropriately trained in not only the structural differences of textured hair but also the unique practices and beliefs among Black women in relation to their hair.

Acknowledgments—Jewell Dinkins is the inaugural recipient of the Janssen–Skin of Color Research Fellowship at Howard University (Washington, DC), and Chidubem A.V. Okeke is the inaugural recipient of the Women’s Dermatologic Society–La Roche-Posay dermatology fellowship at Howard University.

Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E30-E31
Page Number
E30-E31
Publications
Publications
Topics
Article Type
Display Headline
Navigating Hair Loss in Medical School: Experiences of 2 Young Black Women
Display Headline
Navigating Hair Loss in Medical School: Experiences of 2 Young Black Women
Sections
Inside the Article

Practice Points

  • Hair loss is a common dermatologic concern among Black women and can represent a diagnostic challenge to dermatologists who may not be familiar with textured hair.
  • Dermatologists should practice cultural sensitivity and provide relevant recommendations to Black patients dealing with hair loss.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Thalidomide Analogue Drug Eruption Along the Lines of Blaschko

Article Type
Changed
Thu, 12/21/2023 - 10:31
Display Headline
Thalidomide Analogue Drug Eruption Along the Lines of Blaschko

To the Editor:

Lenalidomide is a thalidomide analogue used to treat various hematologic malignancies, including non-Hodgkin lymphoma, myelodysplastic syndrome, and multiple myeloma (MM).1 Lenalidomide is referred to as a degrader therapeutic because it induces targeted protein degradation of disease-relevant proteins (eg, Ikaros family zinc finger protein 1 [IKZF1], Ikaros family zinc finger protein 3 [IKZF3], and casein kinase I isoform-α [CK1α]) as its primary mechanism of action.1,2 Although cutaneous adverse events are relatively common among thalidomide analogues, the morphologic and histopathologic descriptions of these drug eruptions have not been fully elucidated.3,4 We report a novel pityriasiform drug eruption followed by a clinical eruption suggestive of blaschkitis in a patient with MM who was being treated with lenalidomide.

A 76-year-old man presented to the dermatology clinic with a progressive, mildly pruritic eruption on the chest and axillae of 1 year’s duration. He had a medical history of chronic hepatitis B, malignant carcinoid tumor of the colon, prostate cancer, and MM. The eruption emerged 1 to 2 weeks after the patient started oral lenalidomide 10 mg/d and oral dexamethasone40 mg/wk following autologous stem cell transplantation for MM. The patient had not received any other therapy for MM.

Physical examination revealed multiple erythematous, hyperpigmented, scaly papules and plaques on the lateral chest and within the axillae (Figure 1). A skin biopsy from the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate with scattered eosinophils, neutrophils, and extravasated erythrocytes. The overlying epidermis showed spongiosis with parakeratosis in addition to lymphocytic exocytosis (Figure 2). No fungal organisms were highlighted on periodic acid–Schiff staining. After this evaluation, we recommended that the patient discontinue lenalidomide and start taking a topical over-the-counter corticosteroid for 2 weeks. Over time, he noted marked improvement in the eruption and associated pruritus.

Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.
FIGURE 1. Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.

After a drug holiday of 2 months, the patient resumed a maintenance dosage of oral lenalidomide 10 mg/d. Four or 5 days after restarting lenalidomide, a pruritic eruption appeared that involved the axillae and the left lower abdomen, circling around to the left lower back. The axillary eruption resolved with a topical over-the-counter corticosteroid; the abdominal eruption persisted.

A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes.
FIGURE 2. A and B, A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes. The overlying epidermis was spongiotic with parakeratosis and lymphocytic exocytosis (H&E, original magnifications ×100 and ×200).

At the 3-month follow-up visit, physical examination revealed erythematous macules and papules that coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline (Figure 3).

At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossi
FIGURE 3. A and B, At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline.

We recommended that the patient continue treatment through this eruption; he was instructed to apply a corticosteroid cream and resume lenalidomide at the maintenance dosage. A month later, he reported that the eruption and associated pruritus resolved with the corticosteroid cream and resumption of the maintenance dose of lenalidomide. The patient noted no further spread of the eruption.

Cutaneous adverse events are common following lenalidomide. In prior trials, the overall incidence of any-grade rash following lenalidomide exposure was 22% to 33%.5 A meta-analysis of 10 trials determined the overall incidence of all-grade and high-grade cutaneous adverse events after exposure to lenalidomide was 27.2% and 3.6%, respectively.6 Our case represents a pityriasiform eruption due to lenalidomide followed by a secondary eruption suggestive of blaschkitis.

 

 

The rash due to lenalidomide has been described as morbilliform, urticarial, dermatitic, acneform, and undefined.7 Lenalidomide-induced rash typically develops during the first month of therapy, similar to our patient’s presentation. It has even been observed in the first week of therapy.8 Severe reactions such as Stevens-Johnson syndrome and toxic epidermal necrolysis have been reported.5,6 Risk factors associated with rash secondary to lenalidomide include advanced age (≥70 years), presence of Bence-Jones protein-type MM in urine, and no prior chemotherapy.8 Our patient had 2 of these risk factors: advanced age and no prior chemotherapy for MM. The exact pathogenesis by which lenalidomide leads to a pityriasiform eruption, as in our patient, or to a rash in general is unclear. Studies have hypothesized that a lenalidomide-induced rash could be attributable to a delayed hypersensitivity type IV reaction or to a reaction related to the molecular mechanism of action of the drug.9

At the molecular level, the antimyeloma effects of lenalidomide include promoting degradation of transcription factors IKZF1 and IKZF3, which subsequently increases production of IL-2.1,2,9 Recombinant IL-2 has been associated with an increased incidence of rash in other cancers.9 Overexpression of programmed death 1(PD-1) and its ligand (PD-L1) has been demonstrated in MM; lenalidomide has been shown to downregulate both PD-1 and PD-L1. Patients receiving PD-1 and PD-L1 inhibitors commonly have developed rash.9 However, the association between lenalidomide and its downregulation of PD-1 and PD-L1 leading to rash has not been fully elucidated. Given the multiple malignancies in our patient—MM, prostate cancer, malignant carcinoid tumor—an underlying paraneoplastic phenomenon may be possible. Additionally, because our patient initially received dexamethasone along with lenalidomide, the manifestation of the initial pityriasiform rash may have been less severe due to the steroid use. Although our patient underwent a 2-month drug holiday following the initial pityriasiform eruption, most lenalidomide-induced rashes do not necessitate discontinuation of the drug.5,7

Our patient’s secondary drug eruption was clinically suggestive of lenalidomide-induced blaschkitis. A report of a German patient with plasmacytoma described a unilateral papular exanthem that developed 4 months after lenalidomide was initiated.10 The papular exanthem following the lines of Blaschko lines extended from that patient’s posterior left foot to the calf and on to the thigh and flank,10 which was more extensive than our patient’s eruption. Blaschkitis in this patient resolved with a corticosteroid cream and UV light therapy10; lenalidomide was not discontinued, similar to our patient.

The pathogenesis of our patient’s secondary eruption that preferentially involved the lines of Blaschko is unclear. After the initial pityriasiform eruption, the secondary eruption was blaschkitis. Distinguishing dermatomes from the lines of Blaschko, which are thought to represent pathways of epidermal cell migration and proliferation during embryologic development, is important. Genodermatoses such as incontinentia pigmenti and hypomelanosis of Ito involve the lines of Blaschko11; other disorders in the differential diagnosis of linear configurations include linear lichen planus, linear cutaneous lupus erythematosus, linear morphea, and lichen striatus.11 Notably, drug-induced blaschkitis is rare.

Cutaneous adverse reactions from thalidomide analogues are relatively common. Our case of lenalidomide-associated blaschkitis that developed following an initial pityriasiform drug eruption in a patient with MM highlights that dermatologists need to collaborate with the oncologist regarding the severity of drug eruptions to determine if the patient should continue treatment through the cutaneous eruptions or discontinue a vital medication.

References
  1. Jan M, Sperling AS, Ebert BL. Cancer therapies based on targeted protein degradation—lessons learned with lenalidomide. Nat Rev Clin Oncol. 2021;18:401-417. doi:10.1038/s41571-021-00479-z
  2. Shah UA, Mailankody S. Emerging immunotherapies in multiple myeloma. BMJ. 2020;370:3176. doi:10.1136/BMJ.M3176
  3. Richardson PG, Blood E, Mitsiades CS, et al. A randomized phase 2 study of lenalidomide therapy for patients with relapsed or relapsed and refractory multiple myeloma. Blood. 2006;108:3458-3464. doi:10.1182/BLOOD-2006-04-015909
  4. Benboubker L, Dimopoulos MA, Dispenzieri A, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med. 2014;371:906-917. doi:10.1056/NEJMOA1402551
  5. Tinsley SM, Kurtin SE, Ridgeway JA. Practical management of lenalidomide-related rash. Clin Lymphoma Myeloma Leuk. 2015;15(suppl):S64-S69. doi:10.1016/J.CLML.2015.02.008
  6. Nardone B, Wu S, Garden BC, et al. Risk of rash associated with lenalidomide in cancer patients: a systematic review of the literature and meta-analysis. Clin Lymphoma Myeloma Leuk. 2013;13:424-429. doi:10.1016/J.CLML.2013.03.006
  7. Sviggum HP, Davis MDP, Rajkumar SV, et al. Dermatologic adverse effects of lenalidomide therapy for amyloidosis and multiple myeloma. Arch Dermatol. 2006;142:1298-1302. doi:10.1001/ARCHDERM.142.10.1298
  8. Sugi T, Nishigami Y, Saigo H, et al. Analysis of risk factors for lenalidomide-associated skin rash in patients with multiple myeloma. Leuk Lymphoma. 2021;62:1405-1410. doi:10.1080/10428194.2021.1876867
  9. Barley K, He W, Agarwal S, et al. Outcomes and management of lenalidomide-associated rash in patients with multiple myeloma. Leuk Lymphoma. 2016;57:2510-2515. doi:10.3109/10428194.2016.1151507
  10. Grape J, Frosch P. Papular drug eruption along the lines of Blaschko caused by lenalidomide [in German]. Hautarzt. 2011;62:618-620. doi:10.1007/S00105-010-2121-6
  11. Bolognia JL, Orlow SJ, Glick SA. Lines of Blaschko. J Am Acad Dermatol. 1994;31(2 pt 1):157-190. doi:10.1016/S0190-9622(94)70143-1
Article PDF
Author and Disclosure Information

From the Tulane University School of Medicine, New Orleans, Louisiana. Drs. Jafari and Williams are from the Department of Dermatology, and Dr. Vick is from the Department of Dermatopathology.

The authors report no conflict of interest.

Correspondence: Alexander J. Jafari, MD, MPH, Department of Dermatology, Tulane University School of Medicine, 1430 Tulane Ave, #8036, New Orleans, LA 70112 (ajafari@tulane.edu).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E27-E29
Sections
Author and Disclosure Information

From the Tulane University School of Medicine, New Orleans, Louisiana. Drs. Jafari and Williams are from the Department of Dermatology, and Dr. Vick is from the Department of Dermatopathology.

The authors report no conflict of interest.

Correspondence: Alexander J. Jafari, MD, MPH, Department of Dermatology, Tulane University School of Medicine, 1430 Tulane Ave, #8036, New Orleans, LA 70112 (ajafari@tulane.edu).

Author and Disclosure Information

From the Tulane University School of Medicine, New Orleans, Louisiana. Drs. Jafari and Williams are from the Department of Dermatology, and Dr. Vick is from the Department of Dermatopathology.

The authors report no conflict of interest.

Correspondence: Alexander J. Jafari, MD, MPH, Department of Dermatology, Tulane University School of Medicine, 1430 Tulane Ave, #8036, New Orleans, LA 70112 (ajafari@tulane.edu).

Article PDF
Article PDF

To the Editor:

Lenalidomide is a thalidomide analogue used to treat various hematologic malignancies, including non-Hodgkin lymphoma, myelodysplastic syndrome, and multiple myeloma (MM).1 Lenalidomide is referred to as a degrader therapeutic because it induces targeted protein degradation of disease-relevant proteins (eg, Ikaros family zinc finger protein 1 [IKZF1], Ikaros family zinc finger protein 3 [IKZF3], and casein kinase I isoform-α [CK1α]) as its primary mechanism of action.1,2 Although cutaneous adverse events are relatively common among thalidomide analogues, the morphologic and histopathologic descriptions of these drug eruptions have not been fully elucidated.3,4 We report a novel pityriasiform drug eruption followed by a clinical eruption suggestive of blaschkitis in a patient with MM who was being treated with lenalidomide.

A 76-year-old man presented to the dermatology clinic with a progressive, mildly pruritic eruption on the chest and axillae of 1 year’s duration. He had a medical history of chronic hepatitis B, malignant carcinoid tumor of the colon, prostate cancer, and MM. The eruption emerged 1 to 2 weeks after the patient started oral lenalidomide 10 mg/d and oral dexamethasone40 mg/wk following autologous stem cell transplantation for MM. The patient had not received any other therapy for MM.

Physical examination revealed multiple erythematous, hyperpigmented, scaly papules and plaques on the lateral chest and within the axillae (Figure 1). A skin biopsy from the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate with scattered eosinophils, neutrophils, and extravasated erythrocytes. The overlying epidermis showed spongiosis with parakeratosis in addition to lymphocytic exocytosis (Figure 2). No fungal organisms were highlighted on periodic acid–Schiff staining. After this evaluation, we recommended that the patient discontinue lenalidomide and start taking a topical over-the-counter corticosteroid for 2 weeks. Over time, he noted marked improvement in the eruption and associated pruritus.

Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.
FIGURE 1. Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.

After a drug holiday of 2 months, the patient resumed a maintenance dosage of oral lenalidomide 10 mg/d. Four or 5 days after restarting lenalidomide, a pruritic eruption appeared that involved the axillae and the left lower abdomen, circling around to the left lower back. The axillary eruption resolved with a topical over-the-counter corticosteroid; the abdominal eruption persisted.

A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes.
FIGURE 2. A and B, A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes. The overlying epidermis was spongiotic with parakeratosis and lymphocytic exocytosis (H&E, original magnifications ×100 and ×200).

At the 3-month follow-up visit, physical examination revealed erythematous macules and papules that coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline (Figure 3).

At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossi
FIGURE 3. A and B, At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline.

We recommended that the patient continue treatment through this eruption; he was instructed to apply a corticosteroid cream and resume lenalidomide at the maintenance dosage. A month later, he reported that the eruption and associated pruritus resolved with the corticosteroid cream and resumption of the maintenance dose of lenalidomide. The patient noted no further spread of the eruption.

Cutaneous adverse events are common following lenalidomide. In prior trials, the overall incidence of any-grade rash following lenalidomide exposure was 22% to 33%.5 A meta-analysis of 10 trials determined the overall incidence of all-grade and high-grade cutaneous adverse events after exposure to lenalidomide was 27.2% and 3.6%, respectively.6 Our case represents a pityriasiform eruption due to lenalidomide followed by a secondary eruption suggestive of blaschkitis.

 

 

The rash due to lenalidomide has been described as morbilliform, urticarial, dermatitic, acneform, and undefined.7 Lenalidomide-induced rash typically develops during the first month of therapy, similar to our patient’s presentation. It has even been observed in the first week of therapy.8 Severe reactions such as Stevens-Johnson syndrome and toxic epidermal necrolysis have been reported.5,6 Risk factors associated with rash secondary to lenalidomide include advanced age (≥70 years), presence of Bence-Jones protein-type MM in urine, and no prior chemotherapy.8 Our patient had 2 of these risk factors: advanced age and no prior chemotherapy for MM. The exact pathogenesis by which lenalidomide leads to a pityriasiform eruption, as in our patient, or to a rash in general is unclear. Studies have hypothesized that a lenalidomide-induced rash could be attributable to a delayed hypersensitivity type IV reaction or to a reaction related to the molecular mechanism of action of the drug.9

At the molecular level, the antimyeloma effects of lenalidomide include promoting degradation of transcription factors IKZF1 and IKZF3, which subsequently increases production of IL-2.1,2,9 Recombinant IL-2 has been associated with an increased incidence of rash in other cancers.9 Overexpression of programmed death 1(PD-1) and its ligand (PD-L1) has been demonstrated in MM; lenalidomide has been shown to downregulate both PD-1 and PD-L1. Patients receiving PD-1 and PD-L1 inhibitors commonly have developed rash.9 However, the association between lenalidomide and its downregulation of PD-1 and PD-L1 leading to rash has not been fully elucidated. Given the multiple malignancies in our patient—MM, prostate cancer, malignant carcinoid tumor—an underlying paraneoplastic phenomenon may be possible. Additionally, because our patient initially received dexamethasone along with lenalidomide, the manifestation of the initial pityriasiform rash may have been less severe due to the steroid use. Although our patient underwent a 2-month drug holiday following the initial pityriasiform eruption, most lenalidomide-induced rashes do not necessitate discontinuation of the drug.5,7

Our patient’s secondary drug eruption was clinically suggestive of lenalidomide-induced blaschkitis. A report of a German patient with plasmacytoma described a unilateral papular exanthem that developed 4 months after lenalidomide was initiated.10 The papular exanthem following the lines of Blaschko lines extended from that patient’s posterior left foot to the calf and on to the thigh and flank,10 which was more extensive than our patient’s eruption. Blaschkitis in this patient resolved with a corticosteroid cream and UV light therapy10; lenalidomide was not discontinued, similar to our patient.

The pathogenesis of our patient’s secondary eruption that preferentially involved the lines of Blaschko is unclear. After the initial pityriasiform eruption, the secondary eruption was blaschkitis. Distinguishing dermatomes from the lines of Blaschko, which are thought to represent pathways of epidermal cell migration and proliferation during embryologic development, is important. Genodermatoses such as incontinentia pigmenti and hypomelanosis of Ito involve the lines of Blaschko11; other disorders in the differential diagnosis of linear configurations include linear lichen planus, linear cutaneous lupus erythematosus, linear morphea, and lichen striatus.11 Notably, drug-induced blaschkitis is rare.

Cutaneous adverse reactions from thalidomide analogues are relatively common. Our case of lenalidomide-associated blaschkitis that developed following an initial pityriasiform drug eruption in a patient with MM highlights that dermatologists need to collaborate with the oncologist regarding the severity of drug eruptions to determine if the patient should continue treatment through the cutaneous eruptions or discontinue a vital medication.

To the Editor:

Lenalidomide is a thalidomide analogue used to treat various hematologic malignancies, including non-Hodgkin lymphoma, myelodysplastic syndrome, and multiple myeloma (MM).1 Lenalidomide is referred to as a degrader therapeutic because it induces targeted protein degradation of disease-relevant proteins (eg, Ikaros family zinc finger protein 1 [IKZF1], Ikaros family zinc finger protein 3 [IKZF3], and casein kinase I isoform-α [CK1α]) as its primary mechanism of action.1,2 Although cutaneous adverse events are relatively common among thalidomide analogues, the morphologic and histopathologic descriptions of these drug eruptions have not been fully elucidated.3,4 We report a novel pityriasiform drug eruption followed by a clinical eruption suggestive of blaschkitis in a patient with MM who was being treated with lenalidomide.

A 76-year-old man presented to the dermatology clinic with a progressive, mildly pruritic eruption on the chest and axillae of 1 year’s duration. He had a medical history of chronic hepatitis B, malignant carcinoid tumor of the colon, prostate cancer, and MM. The eruption emerged 1 to 2 weeks after the patient started oral lenalidomide 10 mg/d and oral dexamethasone40 mg/wk following autologous stem cell transplantation for MM. The patient had not received any other therapy for MM.

Physical examination revealed multiple erythematous, hyperpigmented, scaly papules and plaques on the lateral chest and within the axillae (Figure 1). A skin biopsy from the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate with scattered eosinophils, neutrophils, and extravasated erythrocytes. The overlying epidermis showed spongiosis with parakeratosis in addition to lymphocytic exocytosis (Figure 2). No fungal organisms were highlighted on periodic acid–Schiff staining. After this evaluation, we recommended that the patient discontinue lenalidomide and start taking a topical over-the-counter corticosteroid for 2 weeks. Over time, he noted marked improvement in the eruption and associated pruritus.

Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.
FIGURE 1. Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.

After a drug holiday of 2 months, the patient resumed a maintenance dosage of oral lenalidomide 10 mg/d. Four or 5 days after restarting lenalidomide, a pruritic eruption appeared that involved the axillae and the left lower abdomen, circling around to the left lower back. The axillary eruption resolved with a topical over-the-counter corticosteroid; the abdominal eruption persisted.

A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes.
FIGURE 2. A and B, A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes. The overlying epidermis was spongiotic with parakeratosis and lymphocytic exocytosis (H&E, original magnifications ×100 and ×200).

At the 3-month follow-up visit, physical examination revealed erythematous macules and papules that coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline (Figure 3).

At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossi
FIGURE 3. A and B, At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline.

We recommended that the patient continue treatment through this eruption; he was instructed to apply a corticosteroid cream and resume lenalidomide at the maintenance dosage. A month later, he reported that the eruption and associated pruritus resolved with the corticosteroid cream and resumption of the maintenance dose of lenalidomide. The patient noted no further spread of the eruption.

Cutaneous adverse events are common following lenalidomide. In prior trials, the overall incidence of any-grade rash following lenalidomide exposure was 22% to 33%.5 A meta-analysis of 10 trials determined the overall incidence of all-grade and high-grade cutaneous adverse events after exposure to lenalidomide was 27.2% and 3.6%, respectively.6 Our case represents a pityriasiform eruption due to lenalidomide followed by a secondary eruption suggestive of blaschkitis.

 

 

The rash due to lenalidomide has been described as morbilliform, urticarial, dermatitic, acneform, and undefined.7 Lenalidomide-induced rash typically develops during the first month of therapy, similar to our patient’s presentation. It has even been observed in the first week of therapy.8 Severe reactions such as Stevens-Johnson syndrome and toxic epidermal necrolysis have been reported.5,6 Risk factors associated with rash secondary to lenalidomide include advanced age (≥70 years), presence of Bence-Jones protein-type MM in urine, and no prior chemotherapy.8 Our patient had 2 of these risk factors: advanced age and no prior chemotherapy for MM. The exact pathogenesis by which lenalidomide leads to a pityriasiform eruption, as in our patient, or to a rash in general is unclear. Studies have hypothesized that a lenalidomide-induced rash could be attributable to a delayed hypersensitivity type IV reaction or to a reaction related to the molecular mechanism of action of the drug.9

At the molecular level, the antimyeloma effects of lenalidomide include promoting degradation of transcription factors IKZF1 and IKZF3, which subsequently increases production of IL-2.1,2,9 Recombinant IL-2 has been associated with an increased incidence of rash in other cancers.9 Overexpression of programmed death 1(PD-1) and its ligand (PD-L1) has been demonstrated in MM; lenalidomide has been shown to downregulate both PD-1 and PD-L1. Patients receiving PD-1 and PD-L1 inhibitors commonly have developed rash.9 However, the association between lenalidomide and its downregulation of PD-1 and PD-L1 leading to rash has not been fully elucidated. Given the multiple malignancies in our patient—MM, prostate cancer, malignant carcinoid tumor—an underlying paraneoplastic phenomenon may be possible. Additionally, because our patient initially received dexamethasone along with lenalidomide, the manifestation of the initial pityriasiform rash may have been less severe due to the steroid use. Although our patient underwent a 2-month drug holiday following the initial pityriasiform eruption, most lenalidomide-induced rashes do not necessitate discontinuation of the drug.5,7

Our patient’s secondary drug eruption was clinically suggestive of lenalidomide-induced blaschkitis. A report of a German patient with plasmacytoma described a unilateral papular exanthem that developed 4 months after lenalidomide was initiated.10 The papular exanthem following the lines of Blaschko lines extended from that patient’s posterior left foot to the calf and on to the thigh and flank,10 which was more extensive than our patient’s eruption. Blaschkitis in this patient resolved with a corticosteroid cream and UV light therapy10; lenalidomide was not discontinued, similar to our patient.

The pathogenesis of our patient’s secondary eruption that preferentially involved the lines of Blaschko is unclear. After the initial pityriasiform eruption, the secondary eruption was blaschkitis. Distinguishing dermatomes from the lines of Blaschko, which are thought to represent pathways of epidermal cell migration and proliferation during embryologic development, is important. Genodermatoses such as incontinentia pigmenti and hypomelanosis of Ito involve the lines of Blaschko11; other disorders in the differential diagnosis of linear configurations include linear lichen planus, linear cutaneous lupus erythematosus, linear morphea, and lichen striatus.11 Notably, drug-induced blaschkitis is rare.

Cutaneous adverse reactions from thalidomide analogues are relatively common. Our case of lenalidomide-associated blaschkitis that developed following an initial pityriasiform drug eruption in a patient with MM highlights that dermatologists need to collaborate with the oncologist regarding the severity of drug eruptions to determine if the patient should continue treatment through the cutaneous eruptions or discontinue a vital medication.

References
  1. Jan M, Sperling AS, Ebert BL. Cancer therapies based on targeted protein degradation—lessons learned with lenalidomide. Nat Rev Clin Oncol. 2021;18:401-417. doi:10.1038/s41571-021-00479-z
  2. Shah UA, Mailankody S. Emerging immunotherapies in multiple myeloma. BMJ. 2020;370:3176. doi:10.1136/BMJ.M3176
  3. Richardson PG, Blood E, Mitsiades CS, et al. A randomized phase 2 study of lenalidomide therapy for patients with relapsed or relapsed and refractory multiple myeloma. Blood. 2006;108:3458-3464. doi:10.1182/BLOOD-2006-04-015909
  4. Benboubker L, Dimopoulos MA, Dispenzieri A, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med. 2014;371:906-917. doi:10.1056/NEJMOA1402551
  5. Tinsley SM, Kurtin SE, Ridgeway JA. Practical management of lenalidomide-related rash. Clin Lymphoma Myeloma Leuk. 2015;15(suppl):S64-S69. doi:10.1016/J.CLML.2015.02.008
  6. Nardone B, Wu S, Garden BC, et al. Risk of rash associated with lenalidomide in cancer patients: a systematic review of the literature and meta-analysis. Clin Lymphoma Myeloma Leuk. 2013;13:424-429. doi:10.1016/J.CLML.2013.03.006
  7. Sviggum HP, Davis MDP, Rajkumar SV, et al. Dermatologic adverse effects of lenalidomide therapy for amyloidosis and multiple myeloma. Arch Dermatol. 2006;142:1298-1302. doi:10.1001/ARCHDERM.142.10.1298
  8. Sugi T, Nishigami Y, Saigo H, et al. Analysis of risk factors for lenalidomide-associated skin rash in patients with multiple myeloma. Leuk Lymphoma. 2021;62:1405-1410. doi:10.1080/10428194.2021.1876867
  9. Barley K, He W, Agarwal S, et al. Outcomes and management of lenalidomide-associated rash in patients with multiple myeloma. Leuk Lymphoma. 2016;57:2510-2515. doi:10.3109/10428194.2016.1151507
  10. Grape J, Frosch P. Papular drug eruption along the lines of Blaschko caused by lenalidomide [in German]. Hautarzt. 2011;62:618-620. doi:10.1007/S00105-010-2121-6
  11. Bolognia JL, Orlow SJ, Glick SA. Lines of Blaschko. J Am Acad Dermatol. 1994;31(2 pt 1):157-190. doi:10.1016/S0190-9622(94)70143-1
References
  1. Jan M, Sperling AS, Ebert BL. Cancer therapies based on targeted protein degradation—lessons learned with lenalidomide. Nat Rev Clin Oncol. 2021;18:401-417. doi:10.1038/s41571-021-00479-z
  2. Shah UA, Mailankody S. Emerging immunotherapies in multiple myeloma. BMJ. 2020;370:3176. doi:10.1136/BMJ.M3176
  3. Richardson PG, Blood E, Mitsiades CS, et al. A randomized phase 2 study of lenalidomide therapy for patients with relapsed or relapsed and refractory multiple myeloma. Blood. 2006;108:3458-3464. doi:10.1182/BLOOD-2006-04-015909
  4. Benboubker L, Dimopoulos MA, Dispenzieri A, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med. 2014;371:906-917. doi:10.1056/NEJMOA1402551
  5. Tinsley SM, Kurtin SE, Ridgeway JA. Practical management of lenalidomide-related rash. Clin Lymphoma Myeloma Leuk. 2015;15(suppl):S64-S69. doi:10.1016/J.CLML.2015.02.008
  6. Nardone B, Wu S, Garden BC, et al. Risk of rash associated with lenalidomide in cancer patients: a systematic review of the literature and meta-analysis. Clin Lymphoma Myeloma Leuk. 2013;13:424-429. doi:10.1016/J.CLML.2013.03.006
  7. Sviggum HP, Davis MDP, Rajkumar SV, et al. Dermatologic adverse effects of lenalidomide therapy for amyloidosis and multiple myeloma. Arch Dermatol. 2006;142:1298-1302. doi:10.1001/ARCHDERM.142.10.1298
  8. Sugi T, Nishigami Y, Saigo H, et al. Analysis of risk factors for lenalidomide-associated skin rash in patients with multiple myeloma. Leuk Lymphoma. 2021;62:1405-1410. doi:10.1080/10428194.2021.1876867
  9. Barley K, He W, Agarwal S, et al. Outcomes and management of lenalidomide-associated rash in patients with multiple myeloma. Leuk Lymphoma. 2016;57:2510-2515. doi:10.3109/10428194.2016.1151507
  10. Grape J, Frosch P. Papular drug eruption along the lines of Blaschko caused by lenalidomide [in German]. Hautarzt. 2011;62:618-620. doi:10.1007/S00105-010-2121-6
  11. Bolognia JL, Orlow SJ, Glick SA. Lines of Blaschko. J Am Acad Dermatol. 1994;31(2 pt 1):157-190. doi:10.1016/S0190-9622(94)70143-1
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E27-E29
Page Number
E27-E29
Publications
Publications
Topics
Article Type
Display Headline
Thalidomide Analogue Drug Eruption Along the Lines of Blaschko
Display Headline
Thalidomide Analogue Drug Eruption Along the Lines of Blaschko
Sections
Inside the Article

Practice Points

  • Dermatologists should be aware of the variety of cutaneous adverse events that can arise from the use of immunotherapeutic agents for hematologic malignancies.
  • Some cutaneous reactions to immunotherapeutic medications, such as pityriasiform eruption and blaschkitis, generally are benign and may not necessitate halting an important therapy.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

The Role of Toluidine Blue in Mohs Micrographic Surgery: A Systematic Review

Article Type
Changed
Thu, 12/21/2023 - 08:12
Display Headline
The Role of Toluidine Blue in Mohs Micrographic Surgery: A Systematic Review

Toluidine blue (TB), a dye with metachromatic staining properties, was developed in 1856 by William Henry Perkin.1 Metachromasia is a perceptible change in the color of staining of living tissue due to the electrochemical properties of the tissue. Tissues that contain high concentrations of ionized sulfate and phosphate groups (high concentrations of free electronegative groups) form polymeric aggregates of the basic dye solution that alter the absorbed wavelengths of light.2 The function of this characteristic is to use a single dye to highlight different structures in tissue based on their relative chemical differences.3

Toluidine blue primarily was used within the dye industry until the 1960s, when it was first used in vital staining of the oral mucosa.2 Because of the tissue absorption potential, this technique was used to detect the location of oral malignancies.4 Since then, TB has progressively been used for staining fresh frozen sections in Mohs micrographic surgery (MMS). In a 2003 survey study (N=310), 16.8% of surgeons performing MMS reported using TB in their laboratory.5 We sought to systematically review the published literature describing the uses of TB in the setting of fresh frozen sections and MMS.

Methods

We conducted a systematic search of the PubMed and Cochrane databases for articles published before December 1, 2019, to identify any relevant studies in English. Electronic searches were performed using the terms toluidine blue and Mohs or Mohs micrographic surgery. We manually checked the bibliographies of the identified articles to further identify eligible studies.

Eligibility Criteria—The inclusion criteria were articles that (1) considered TB in the context of MMS, (2) were published in peer-reviewed journals, (3) were published in English, and (4) were available as full text. Systematic reviews were excluded.

Data Extraction and Outcomes—All relevant information regarding the study characteristics, including design, level of evidence, methodologic quality of evidence, pathology examined, and outcome measures, were collected by 2 independent reviewers (T.L. and A.D.) using a predetermined data sheet. The same 2 reviewers were used for all steps of the review process, data were independently obtained, and any discrepancy was introduced for a third opinion (D.H.) and agreed upon by the majority.

Quality Assessment—The level of evidence was evaluated based on the criteria of the Oxford Centre for Evidence-Based Medicine. Two reviewers (T.L. and A.D.) graded each article included in the review.

PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.
FIGURE 1. PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.

Results

A total of 25 articles were reviewed. After the titles and abstracts were screened for relevance, 12 articles remained (Figure 1). Of these, 1 compared basal cell carcinoma (BCC) and squamous cell carcinoma (SCC), 4 were related to BCC, 3 were related to SCC, 1 was related to microcystic adnexal carcinoma (MAC), 1 was related to primary cutaneous adenoid cystic carcinoma (PCACC), and 2 were related to technical aspects of the staining process (Table 1).

Summary of Articles Published on Toluidine Blue in Mohs Micrographic Surgery

 

 

A majority of the articles included in this review were qualitative and observational in nature, describing the staining characteristics of TB. Study characteristics are summarized in Table 1.

Comment

Basal Cell Carcinoma—Toluidine blue staining characteristics help to identify BCC nests by differentiating them from hair follicles in frozen sections. The metachromatic characteristic of TB stains the inner root sheath deep blue and highlights the surrounding stromal mucin of BCC a magenta color.18,19 In hematoxylin and eosin (H&E) stains, these 2 distinct structures can be differentiated by cleft formation around tumor nests, mitotic figures, and the lack of a fibrous sheath present in BCC tumors.20 The advantages and limitations of TB staining of BCC are presented in Table 2.

Advantages and Limitations of Toluidine Blue Staining in BCC and SCC

Humphreys et al6 suggested a noticeable difference between H&E and TB in the staining of cellular and stromal components. The nuclear detail of tumor cells was subjectively sharper and clearer with TB staining. The staining of stromal components may provide the most assistance in locating BCC islands. Mucopolysaccharide staining may be absent in H&E but stain a deep magenta with TB. Although the presence of mucopolysaccharides does not specifically indicate a tumor, it may prompt further attention and provide an indicator for sparse and infiltrative tumor cells.6 The metachromatic stromal change may indicate a narrow tumor-free margin where additional deeper sections often reveal tumor that may warrant additional resection margin in more aggressive malignancies. In particular, sclerosing/morpheaform BCCs have been shown to induce glycosaminoglycan synthesis and are highlighted more readily with TB than with H&E when compared to surrounding tissue.21 This differentiation in staining has remained a popular reason to routinely incorporate TB into the staining of infiltrative and morpheaform variants of BCC. Additionally, stromal mast cells are believed to be more abundant in the stroma of BCC and are more readily visualized in tissue specimens stained with TB, appearing as bright purple metachromatic granules. These granules are larger than normal and are increased in number.6

The margin behavior of BCC stained with TB was further characterized by Goldberg et al,8 who coined the term setting sun sign, which may be present in sequential sections of a disappearing nodule of a BCC tumor. Stroma, inflammatory infiltrate, and mast cells produce a magenta glow surrounding BCC tumors that is reminiscent of a setting sun (Figure 2). Invasive BCC is considered variable in this presentation, primarily because of zones of cell-free fluid and edema or the second area of inflammatory cells. This unique sign may benefit the inspecting Mohs surgeon by providing a clue to an underlying process that may have residual BCC tumors. The setting sun sign also may assist in identifying exact surgical margins.8

Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.
FIGURE 2. A and B, Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.

The nasal surface has a predilection for BCC.22 The skin of the nose has numerous look-alike structures to consider for complete tumor removal and avoidance of unnecessary removal. One challenge is distinguishing follicular basaloid proliferations (FBP) from BCC, a scenario that is more common on the nose.22 When TB staining was used, the sensitivity for detecting FBP reached 100% in 34 cases reviewed by Donaldson and Weber.10 None of the cases examined showed TB metachromasia surrounding FBP, thus indicating that TB can dependably identify this benign entity. Conversely, 5% (N=279) of BCCs confirmed on H&E did not exhibit surrounding TB metachromasia. This finding is concerning regarding the specificity of TB staining for BCC, but the authors of this study suggested the possibility that these exceptions were benign “simulants” (ie, trichoepithelioma) of BCC.10

The use of TB also has been shown to be statistically beneficial in Mohs training. In a single-center, single-fellow experiment, the sensitivity and specificity of using TB for BCC were extrapolated.9 Using TB as an adjunct in deep sections showed superior sensitivity to H&E alone in identifying BCC, increasing sensitivity from 96.3% to 99.7%. In a cohort of 352 BCC excisions and frozen sections, only 1 BCC was not completely excised. If H&E only had been performed, the fellow would have missed 13 residual BCC tumors.9

Bennett and Taher7 described a case in which hyaluronic acid (HA) from a filler injection was confused with the HA surrounding BCC tumor nests. They found that when TB is used as an adjunct, the HA filler is easier to differentiate from the HA surrounding the BCC tumor nests. In frozen sections stained with TB, the HA filler appeared as an amorphous, metachromatic, reddish-purple, whereas the HA surrounding the BCC tumor nests appeared as a well-defined red. These findings were less obvious in the same sections stained with H&E alone.7

 

 

Squamous Cell Carcinoma—In early investigations, the utility of TB in identifying SCC in frozen sections was thought to be limited. The description by Humphreys and colleagues6 of staining characteristics in SCC suggested that the nuclear detail that H&E provides is more easily recognized. The deep aqua nuclear staining produced with TB was considered more difficult to observe than the cytoplasmic eosinophilia of pyknotic and keratinizing cells in H&E.6

Toluidine blue may be beneficial in providing unique staining characteristics to further detail tumors that are difficult to interpret, such as spindle cell SCC and perineural invasion of aggressive SCC. In H&E, squamous cells of spindle cell SCC (scSCC) blend into the background of inflammatory cells and can be perceptibly difficult to locate. A small cohort of 3 Mohs surgeons who routinely use H&E were surveyed on their ability to detect a proven scSCC in H&E or TB by photograph.12 All 3 were able to detect the scSCC in the TB photographs, but only 2 of 3 were able to detect it in H&E photographs. All 3 surgeons agreed that TB was preferable to H&E for this tumor type. These findings suggested that TB may be superior and preferred over H&E for visualizing tumor cells of scSCC.12 The TB staining characteristics of perineural invasion of aggressive SCC have been referred to as the perineural corona sign because of the bright magenta stain that forms around affected nerves.13 Drosou et al13 suggested that TB may enhance the diagnostic accuracy for perineural SCC.

Rare Tumors—The adjunctive use of TB with H&E has been examined in rare tumors. Published reports have highlighted its use in MMS for treating MAC and PCACC. Toluidine blue exhibits staining advantages for these tumors. It may render isolated nests and perineural invasion of MAC more easily visible on frozen section.15

Although PCACC is rare, the recurrence rate is high.23 Toluidine blue has been used with MMS to ensure complete removal and higher cure rates. The metachromatic nature of TB is advantageous in staining the HA present in these tumors. Those who have reported the use of TB for PCACC prefer it to H&E for frozen sections.14

Technical Aspects—The staining time for TB-treated slides is reduced compared to H&E staining; staining can be efficiently done in frozen sections in less than 2.5 minutes using the method shown in Table 3.17 In comparison, typical H&E staining takes 9 minutes, and older TB techniques take 7 minutes.6

Rapid Toluidine Blue Staining Protocol

Conclusion

Toluidine blue may play an important and helpful role in the successful diagnosis and treatment of particular cutaneous tumors by providing additional diagnostic information. Although surgeons performing MMS will continue using the staining protocols with which they are most comfortable, adjunctive use of TB over time may provide an additional benefit at low risk for disrupting practice efficiency or workflow. Many Mohs surgeons are accustomed to using this stain, even preferring to interpret only TB-stained slides for cutaneous malignancy. Most published studies on this topic have been observational in nature, and additional controlled trials may be warranted to determine the effects on outcomes in real-world practice.

References
  1. Culling CF, Allison TR. Cellular Pathology Technique. 4th ed. Butterworths; 1985.
  2. Bergeron JA, Singer M. Metachromasy: an experimental and theoretical reevaluation. J Biophys Biochem Cytol. 1958;4:433-457. doi:10.1083/jcb.4.4.433
  3. Epstein JB, Scully C, Spinelli J. Toluidine blue and Lugol’s iodine application in the assessment of oral malignant disease and lesions at risk of malignancy. J Oral Pathol Med. 1992;21:160-163. doi:10.1111/j.1600-0714.1992.tb00094.x
  4. Warnakulasuriya KA, Johnson NW. Sensitivity and specificity of OraScan (R) toluidine blue mouthrinse in the detection of oral cancer and precancer. J Oral Pathol Med. 1996;25:97-103. doi:10.1111/j.1600-0714.1996.tb00201.x
  5. Silapunt S, Peterson SR, Alcalay J, et al. Mohs tissue mapping and processing: a survey study. Dermatol Surg. 2003;29:1109-1112; discussion 1112.
  6. Humphreys TR, Nemeth A, McCrevey S, et al. A pilot study comparing toluidine blue and hematoxylin and eosin staining of basal cell and squamous cell carcinoma during Mohs surgery. Dermatol Surg. 1996;22:693-697. doi:10.1111/j.1524-4725.1996.tb00619.x
  7. Bennett R, Taher M. Restylane persistent for 23 months found during Mohs micrographic surgery: a source of confusion with hyaluronic acid surrounding basal cell carcinoma. Dermatol Surg. 2005;31:1366-1369. doi:10.1111/j.1524-4725.2005.31223
  8. Goldberg LH, Wang SQ, Kimyai-Asadi A. The setting sun sign: visualizing the margins of a basal cell carcinoma on serial frozen sections stained with toluidine blue. Dermatol Surg. 2007;33:761-763. doi:10.1111/j.1524-4725.2007.33158.x
  9. Tehrani H, May K, Morris A, et al. Does the dual use of toluidine blue and hematoxylin and eosin staining improve basal cell carcinoma detection by Mohs surgery trainees? Dermatol Surg. 2013;39:995-1000. doi:10.1111/dsu.12180
  10. Donaldson MR, Weber LA. Toluidine blue supports differentiation of folliculocentric basaloid proliferation from basal cell carcinoma on frozen sections in a small single-practice cohort. Dermatol Surg. 2017;43:1303-1306. doi:10.1097/DSS.0000000000001107
  11. Styperek AR, Goldberg LH, Goldschmidt LE, et al. Toluidine blue and hematoxylin and eosin stains are comparable in evaluating squamous cell carcinoma during Mohs. Dermatol Surg. 2016;42:1279-1284. doi:10.1097/DSS.0000000000000872
  12. Trieu D, Drosou A, Goldberg LH, et al. Detecting spindle cell squamous cell carcinomas with toluidine blue on frozen sections. Dermatol Surg. 2014;40:1259-1260. doi:10.1097/DSS.0000000000000147
  13. Drosou A, Trieu D, Goldberg LH, et al. The perineural corona sign: enhancing detection of perineural squamous cell carcinoma during Mohs micrographic surgery with toluidine blue stain. J Am Acad Dermatol. 2014;71:826-827. doi:10.1016/j.jaad.2014.04.076
  14. Chesser RS, Bertler DE, Fitzpatrick JE, et al. Primary cutaneous adenoid cystic carcinoma treated with Mohs micrographic surgery toluidine blue technique. J Dermatol Surg Oncol. 1992;18:175-176. doi:10.1111/j.1524-4725.1992.tb02794.x
  15. Wang SQ, Goldberg LH, Nemeth A. The merits of adding toluidine blue-stained slides in Mohs surgery in the treatment of a microcystic adnexal carcinoma. J Am Acad Dermatol. 2007;56:1067-1069. doi:10.1016/j.jaad.2007.01.008
  16. Chen CL, Wilson S, Afzalneia R, et al. Topical aluminum chloride and Monsel’s solution block toluidine blue staining in Mohs frozen sections: mechanism and solution. Dermatol Surg. 2019;45:1019-1025. doi:10.1097/DSS.0000000000001761
  17. Todd MM, Lee JW, Marks VJ. Rapid toluidine blue stain for Mohs’ micrographic surgery. Dermatol Surg. 2005;31:244-245. doi:10.1111/j.1524-4725.2005.31053
  18. Picoto AM, Picoto A. Technical procedures for Mohs fresh tissue surgery. J Derm Surg Oncol. 1986;12:134-138. doi:10.1111/j.1524-4725.1986.tb01442.x
  19. Sperling LC, Winton GB. The transverse anatomy of androgenic alopecia. J Derm Surg Oncol. 1990;16:1127-1133. doi:10.1111/j.1524 -4725.1990.tb00024.x
  20. Smith-Zagone MJ, Schwartz MR. Frozen section of skin specimens. Arch Pathol Lab Med. 2005;129:1536-1543. doi:10.5858/2005-129-1536-FSOSS
  21. Moy RL, Potter TS, Uitto J. Increased glycosaminoglycans production in sclerosing basal cell carcinoma–derived fibroblasts and stimulation of normal skin fibroblast glycosaminoglycans production by a cytokine-derived from sclerosing basal cell carcinoma. Dermatol Surg. 2000;26:1029-1036. doi:10.1046/j.1524-4725.2000.0260111029.x
  22. Leshin B, White WL. Folliculocentric basaloid proliferation. The bulge (der Wulst) revisited. Arch Dermatol. 1990;126:900-906. doi:10.1001/archderm.126.7.900
  23. Seab JA, Graham JH. Primary cutaneous adenoid cystic carcinoma.J Am Acad Dermatol. 1987;17:113-118. doi:10.1016/s0190 -9622(87)70182-0
Article PDF
Author and Disclosure Information

Dr. Long is from the Health Corporation of America and Virginia College of Osteopathic Medicine, Blacksburg. Dr. Dunn is in private practice, Tampa, Florida. Drs. Hill and Akin are from the Department of Dermatology, Texas Tech University Health Sciences Center, Lubbock. Dr. Akin also is from Midland Dermatology and Skin Cancer Center, Texas. Dr. Goldberg is from DermSurgery Associates, Houston, Texas.

The authors report no conflicts of interest.

Correspondence: Tyler Long, DO, HCA LewisGale Hospital Montgomery Medical Education, 700 S Main St, Blacksburg, VA 24060 (longjtyler@yahoo.com).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E6-E11
Sections
Author and Disclosure Information

Dr. Long is from the Health Corporation of America and Virginia College of Osteopathic Medicine, Blacksburg. Dr. Dunn is in private practice, Tampa, Florida. Drs. Hill and Akin are from the Department of Dermatology, Texas Tech University Health Sciences Center, Lubbock. Dr. Akin also is from Midland Dermatology and Skin Cancer Center, Texas. Dr. Goldberg is from DermSurgery Associates, Houston, Texas.

The authors report no conflicts of interest.

Correspondence: Tyler Long, DO, HCA LewisGale Hospital Montgomery Medical Education, 700 S Main St, Blacksburg, VA 24060 (longjtyler@yahoo.com).

Author and Disclosure Information

Dr. Long is from the Health Corporation of America and Virginia College of Osteopathic Medicine, Blacksburg. Dr. Dunn is in private practice, Tampa, Florida. Drs. Hill and Akin are from the Department of Dermatology, Texas Tech University Health Sciences Center, Lubbock. Dr. Akin also is from Midland Dermatology and Skin Cancer Center, Texas. Dr. Goldberg is from DermSurgery Associates, Houston, Texas.

The authors report no conflicts of interest.

Correspondence: Tyler Long, DO, HCA LewisGale Hospital Montgomery Medical Education, 700 S Main St, Blacksburg, VA 24060 (longjtyler@yahoo.com).

Article PDF
Article PDF

Toluidine blue (TB), a dye with metachromatic staining properties, was developed in 1856 by William Henry Perkin.1 Metachromasia is a perceptible change in the color of staining of living tissue due to the electrochemical properties of the tissue. Tissues that contain high concentrations of ionized sulfate and phosphate groups (high concentrations of free electronegative groups) form polymeric aggregates of the basic dye solution that alter the absorbed wavelengths of light.2 The function of this characteristic is to use a single dye to highlight different structures in tissue based on their relative chemical differences.3

Toluidine blue primarily was used within the dye industry until the 1960s, when it was first used in vital staining of the oral mucosa.2 Because of the tissue absorption potential, this technique was used to detect the location of oral malignancies.4 Since then, TB has progressively been used for staining fresh frozen sections in Mohs micrographic surgery (MMS). In a 2003 survey study (N=310), 16.8% of surgeons performing MMS reported using TB in their laboratory.5 We sought to systematically review the published literature describing the uses of TB in the setting of fresh frozen sections and MMS.

Methods

We conducted a systematic search of the PubMed and Cochrane databases for articles published before December 1, 2019, to identify any relevant studies in English. Electronic searches were performed using the terms toluidine blue and Mohs or Mohs micrographic surgery. We manually checked the bibliographies of the identified articles to further identify eligible studies.

Eligibility Criteria—The inclusion criteria were articles that (1) considered TB in the context of MMS, (2) were published in peer-reviewed journals, (3) were published in English, and (4) were available as full text. Systematic reviews were excluded.

Data Extraction and Outcomes—All relevant information regarding the study characteristics, including design, level of evidence, methodologic quality of evidence, pathology examined, and outcome measures, were collected by 2 independent reviewers (T.L. and A.D.) using a predetermined data sheet. The same 2 reviewers were used for all steps of the review process, data were independently obtained, and any discrepancy was introduced for a third opinion (D.H.) and agreed upon by the majority.

Quality Assessment—The level of evidence was evaluated based on the criteria of the Oxford Centre for Evidence-Based Medicine. Two reviewers (T.L. and A.D.) graded each article included in the review.

PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.
FIGURE 1. PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.

Results

A total of 25 articles were reviewed. After the titles and abstracts were screened for relevance, 12 articles remained (Figure 1). Of these, 1 compared basal cell carcinoma (BCC) and squamous cell carcinoma (SCC), 4 were related to BCC, 3 were related to SCC, 1 was related to microcystic adnexal carcinoma (MAC), 1 was related to primary cutaneous adenoid cystic carcinoma (PCACC), and 2 were related to technical aspects of the staining process (Table 1).

Summary of Articles Published on Toluidine Blue in Mohs Micrographic Surgery

 

 

A majority of the articles included in this review were qualitative and observational in nature, describing the staining characteristics of TB. Study characteristics are summarized in Table 1.

Comment

Basal Cell Carcinoma—Toluidine blue staining characteristics help to identify BCC nests by differentiating them from hair follicles in frozen sections. The metachromatic characteristic of TB stains the inner root sheath deep blue and highlights the surrounding stromal mucin of BCC a magenta color.18,19 In hematoxylin and eosin (H&E) stains, these 2 distinct structures can be differentiated by cleft formation around tumor nests, mitotic figures, and the lack of a fibrous sheath present in BCC tumors.20 The advantages and limitations of TB staining of BCC are presented in Table 2.

Advantages and Limitations of Toluidine Blue Staining in BCC and SCC

Humphreys et al6 suggested a noticeable difference between H&E and TB in the staining of cellular and stromal components. The nuclear detail of tumor cells was subjectively sharper and clearer with TB staining. The staining of stromal components may provide the most assistance in locating BCC islands. Mucopolysaccharide staining may be absent in H&E but stain a deep magenta with TB. Although the presence of mucopolysaccharides does not specifically indicate a tumor, it may prompt further attention and provide an indicator for sparse and infiltrative tumor cells.6 The metachromatic stromal change may indicate a narrow tumor-free margin where additional deeper sections often reveal tumor that may warrant additional resection margin in more aggressive malignancies. In particular, sclerosing/morpheaform BCCs have been shown to induce glycosaminoglycan synthesis and are highlighted more readily with TB than with H&E when compared to surrounding tissue.21 This differentiation in staining has remained a popular reason to routinely incorporate TB into the staining of infiltrative and morpheaform variants of BCC. Additionally, stromal mast cells are believed to be more abundant in the stroma of BCC and are more readily visualized in tissue specimens stained with TB, appearing as bright purple metachromatic granules. These granules are larger than normal and are increased in number.6

The margin behavior of BCC stained with TB was further characterized by Goldberg et al,8 who coined the term setting sun sign, which may be present in sequential sections of a disappearing nodule of a BCC tumor. Stroma, inflammatory infiltrate, and mast cells produce a magenta glow surrounding BCC tumors that is reminiscent of a setting sun (Figure 2). Invasive BCC is considered variable in this presentation, primarily because of zones of cell-free fluid and edema or the second area of inflammatory cells. This unique sign may benefit the inspecting Mohs surgeon by providing a clue to an underlying process that may have residual BCC tumors. The setting sun sign also may assist in identifying exact surgical margins.8

Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.
FIGURE 2. A and B, Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.

The nasal surface has a predilection for BCC.22 The skin of the nose has numerous look-alike structures to consider for complete tumor removal and avoidance of unnecessary removal. One challenge is distinguishing follicular basaloid proliferations (FBP) from BCC, a scenario that is more common on the nose.22 When TB staining was used, the sensitivity for detecting FBP reached 100% in 34 cases reviewed by Donaldson and Weber.10 None of the cases examined showed TB metachromasia surrounding FBP, thus indicating that TB can dependably identify this benign entity. Conversely, 5% (N=279) of BCCs confirmed on H&E did not exhibit surrounding TB metachromasia. This finding is concerning regarding the specificity of TB staining for BCC, but the authors of this study suggested the possibility that these exceptions were benign “simulants” (ie, trichoepithelioma) of BCC.10

The use of TB also has been shown to be statistically beneficial in Mohs training. In a single-center, single-fellow experiment, the sensitivity and specificity of using TB for BCC were extrapolated.9 Using TB as an adjunct in deep sections showed superior sensitivity to H&E alone in identifying BCC, increasing sensitivity from 96.3% to 99.7%. In a cohort of 352 BCC excisions and frozen sections, only 1 BCC was not completely excised. If H&E only had been performed, the fellow would have missed 13 residual BCC tumors.9

Bennett and Taher7 described a case in which hyaluronic acid (HA) from a filler injection was confused with the HA surrounding BCC tumor nests. They found that when TB is used as an adjunct, the HA filler is easier to differentiate from the HA surrounding the BCC tumor nests. In frozen sections stained with TB, the HA filler appeared as an amorphous, metachromatic, reddish-purple, whereas the HA surrounding the BCC tumor nests appeared as a well-defined red. These findings were less obvious in the same sections stained with H&E alone.7

 

 

Squamous Cell Carcinoma—In early investigations, the utility of TB in identifying SCC in frozen sections was thought to be limited. The description by Humphreys and colleagues6 of staining characteristics in SCC suggested that the nuclear detail that H&E provides is more easily recognized. The deep aqua nuclear staining produced with TB was considered more difficult to observe than the cytoplasmic eosinophilia of pyknotic and keratinizing cells in H&E.6

Toluidine blue may be beneficial in providing unique staining characteristics to further detail tumors that are difficult to interpret, such as spindle cell SCC and perineural invasion of aggressive SCC. In H&E, squamous cells of spindle cell SCC (scSCC) blend into the background of inflammatory cells and can be perceptibly difficult to locate. A small cohort of 3 Mohs surgeons who routinely use H&E were surveyed on their ability to detect a proven scSCC in H&E or TB by photograph.12 All 3 were able to detect the scSCC in the TB photographs, but only 2 of 3 were able to detect it in H&E photographs. All 3 surgeons agreed that TB was preferable to H&E for this tumor type. These findings suggested that TB may be superior and preferred over H&E for visualizing tumor cells of scSCC.12 The TB staining characteristics of perineural invasion of aggressive SCC have been referred to as the perineural corona sign because of the bright magenta stain that forms around affected nerves.13 Drosou et al13 suggested that TB may enhance the diagnostic accuracy for perineural SCC.

Rare Tumors—The adjunctive use of TB with H&E has been examined in rare tumors. Published reports have highlighted its use in MMS for treating MAC and PCACC. Toluidine blue exhibits staining advantages for these tumors. It may render isolated nests and perineural invasion of MAC more easily visible on frozen section.15

Although PCACC is rare, the recurrence rate is high.23 Toluidine blue has been used with MMS to ensure complete removal and higher cure rates. The metachromatic nature of TB is advantageous in staining the HA present in these tumors. Those who have reported the use of TB for PCACC prefer it to H&E for frozen sections.14

Technical Aspects—The staining time for TB-treated slides is reduced compared to H&E staining; staining can be efficiently done in frozen sections in less than 2.5 minutes using the method shown in Table 3.17 In comparison, typical H&E staining takes 9 minutes, and older TB techniques take 7 minutes.6

Rapid Toluidine Blue Staining Protocol

Conclusion

Toluidine blue may play an important and helpful role in the successful diagnosis and treatment of particular cutaneous tumors by providing additional diagnostic information. Although surgeons performing MMS will continue using the staining protocols with which they are most comfortable, adjunctive use of TB over time may provide an additional benefit at low risk for disrupting practice efficiency or workflow. Many Mohs surgeons are accustomed to using this stain, even preferring to interpret only TB-stained slides for cutaneous malignancy. Most published studies on this topic have been observational in nature, and additional controlled trials may be warranted to determine the effects on outcomes in real-world practice.

Toluidine blue (TB), a dye with metachromatic staining properties, was developed in 1856 by William Henry Perkin.1 Metachromasia is a perceptible change in the color of staining of living tissue due to the electrochemical properties of the tissue. Tissues that contain high concentrations of ionized sulfate and phosphate groups (high concentrations of free electronegative groups) form polymeric aggregates of the basic dye solution that alter the absorbed wavelengths of light.2 The function of this characteristic is to use a single dye to highlight different structures in tissue based on their relative chemical differences.3

Toluidine blue primarily was used within the dye industry until the 1960s, when it was first used in vital staining of the oral mucosa.2 Because of the tissue absorption potential, this technique was used to detect the location of oral malignancies.4 Since then, TB has progressively been used for staining fresh frozen sections in Mohs micrographic surgery (MMS). In a 2003 survey study (N=310), 16.8% of surgeons performing MMS reported using TB in their laboratory.5 We sought to systematically review the published literature describing the uses of TB in the setting of fresh frozen sections and MMS.

Methods

We conducted a systematic search of the PubMed and Cochrane databases for articles published before December 1, 2019, to identify any relevant studies in English. Electronic searches were performed using the terms toluidine blue and Mohs or Mohs micrographic surgery. We manually checked the bibliographies of the identified articles to further identify eligible studies.

Eligibility Criteria—The inclusion criteria were articles that (1) considered TB in the context of MMS, (2) were published in peer-reviewed journals, (3) were published in English, and (4) were available as full text. Systematic reviews were excluded.

Data Extraction and Outcomes—All relevant information regarding the study characteristics, including design, level of evidence, methodologic quality of evidence, pathology examined, and outcome measures, were collected by 2 independent reviewers (T.L. and A.D.) using a predetermined data sheet. The same 2 reviewers were used for all steps of the review process, data were independently obtained, and any discrepancy was introduced for a third opinion (D.H.) and agreed upon by the majority.

Quality Assessment—The level of evidence was evaluated based on the criteria of the Oxford Centre for Evidence-Based Medicine. Two reviewers (T.L. and A.D.) graded each article included in the review.

PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.
FIGURE 1. PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.

Results

A total of 25 articles were reviewed. After the titles and abstracts were screened for relevance, 12 articles remained (Figure 1). Of these, 1 compared basal cell carcinoma (BCC) and squamous cell carcinoma (SCC), 4 were related to BCC, 3 were related to SCC, 1 was related to microcystic adnexal carcinoma (MAC), 1 was related to primary cutaneous adenoid cystic carcinoma (PCACC), and 2 were related to technical aspects of the staining process (Table 1).

Summary of Articles Published on Toluidine Blue in Mohs Micrographic Surgery

 

 

A majority of the articles included in this review were qualitative and observational in nature, describing the staining characteristics of TB. Study characteristics are summarized in Table 1.

Comment

Basal Cell Carcinoma—Toluidine blue staining characteristics help to identify BCC nests by differentiating them from hair follicles in frozen sections. The metachromatic characteristic of TB stains the inner root sheath deep blue and highlights the surrounding stromal mucin of BCC a magenta color.18,19 In hematoxylin and eosin (H&E) stains, these 2 distinct structures can be differentiated by cleft formation around tumor nests, mitotic figures, and the lack of a fibrous sheath present in BCC tumors.20 The advantages and limitations of TB staining of BCC are presented in Table 2.

Advantages and Limitations of Toluidine Blue Staining in BCC and SCC

Humphreys et al6 suggested a noticeable difference between H&E and TB in the staining of cellular and stromal components. The nuclear detail of tumor cells was subjectively sharper and clearer with TB staining. The staining of stromal components may provide the most assistance in locating BCC islands. Mucopolysaccharide staining may be absent in H&E but stain a deep magenta with TB. Although the presence of mucopolysaccharides does not specifically indicate a tumor, it may prompt further attention and provide an indicator for sparse and infiltrative tumor cells.6 The metachromatic stromal change may indicate a narrow tumor-free margin where additional deeper sections often reveal tumor that may warrant additional resection margin in more aggressive malignancies. In particular, sclerosing/morpheaform BCCs have been shown to induce glycosaminoglycan synthesis and are highlighted more readily with TB than with H&E when compared to surrounding tissue.21 This differentiation in staining has remained a popular reason to routinely incorporate TB into the staining of infiltrative and morpheaform variants of BCC. Additionally, stromal mast cells are believed to be more abundant in the stroma of BCC and are more readily visualized in tissue specimens stained with TB, appearing as bright purple metachromatic granules. These granules are larger than normal and are increased in number.6

The margin behavior of BCC stained with TB was further characterized by Goldberg et al,8 who coined the term setting sun sign, which may be present in sequential sections of a disappearing nodule of a BCC tumor. Stroma, inflammatory infiltrate, and mast cells produce a magenta glow surrounding BCC tumors that is reminiscent of a setting sun (Figure 2). Invasive BCC is considered variable in this presentation, primarily because of zones of cell-free fluid and edema or the second area of inflammatory cells. This unique sign may benefit the inspecting Mohs surgeon by providing a clue to an underlying process that may have residual BCC tumors. The setting sun sign also may assist in identifying exact surgical margins.8

Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.
FIGURE 2. A and B, Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.

The nasal surface has a predilection for BCC.22 The skin of the nose has numerous look-alike structures to consider for complete tumor removal and avoidance of unnecessary removal. One challenge is distinguishing follicular basaloid proliferations (FBP) from BCC, a scenario that is more common on the nose.22 When TB staining was used, the sensitivity for detecting FBP reached 100% in 34 cases reviewed by Donaldson and Weber.10 None of the cases examined showed TB metachromasia surrounding FBP, thus indicating that TB can dependably identify this benign entity. Conversely, 5% (N=279) of BCCs confirmed on H&E did not exhibit surrounding TB metachromasia. This finding is concerning regarding the specificity of TB staining for BCC, but the authors of this study suggested the possibility that these exceptions were benign “simulants” (ie, trichoepithelioma) of BCC.10

The use of TB also has been shown to be statistically beneficial in Mohs training. In a single-center, single-fellow experiment, the sensitivity and specificity of using TB for BCC were extrapolated.9 Using TB as an adjunct in deep sections showed superior sensitivity to H&E alone in identifying BCC, increasing sensitivity from 96.3% to 99.7%. In a cohort of 352 BCC excisions and frozen sections, only 1 BCC was not completely excised. If H&E only had been performed, the fellow would have missed 13 residual BCC tumors.9

Bennett and Taher7 described a case in which hyaluronic acid (HA) from a filler injection was confused with the HA surrounding BCC tumor nests. They found that when TB is used as an adjunct, the HA filler is easier to differentiate from the HA surrounding the BCC tumor nests. In frozen sections stained with TB, the HA filler appeared as an amorphous, metachromatic, reddish-purple, whereas the HA surrounding the BCC tumor nests appeared as a well-defined red. These findings were less obvious in the same sections stained with H&E alone.7

 

 

Squamous Cell Carcinoma—In early investigations, the utility of TB in identifying SCC in frozen sections was thought to be limited. The description by Humphreys and colleagues6 of staining characteristics in SCC suggested that the nuclear detail that H&E provides is more easily recognized. The deep aqua nuclear staining produced with TB was considered more difficult to observe than the cytoplasmic eosinophilia of pyknotic and keratinizing cells in H&E.6

Toluidine blue may be beneficial in providing unique staining characteristics to further detail tumors that are difficult to interpret, such as spindle cell SCC and perineural invasion of aggressive SCC. In H&E, squamous cells of spindle cell SCC (scSCC) blend into the background of inflammatory cells and can be perceptibly difficult to locate. A small cohort of 3 Mohs surgeons who routinely use H&E were surveyed on their ability to detect a proven scSCC in H&E or TB by photograph.12 All 3 were able to detect the scSCC in the TB photographs, but only 2 of 3 were able to detect it in H&E photographs. All 3 surgeons agreed that TB was preferable to H&E for this tumor type. These findings suggested that TB may be superior and preferred over H&E for visualizing tumor cells of scSCC.12 The TB staining characteristics of perineural invasion of aggressive SCC have been referred to as the perineural corona sign because of the bright magenta stain that forms around affected nerves.13 Drosou et al13 suggested that TB may enhance the diagnostic accuracy for perineural SCC.

Rare Tumors—The adjunctive use of TB with H&E has been examined in rare tumors. Published reports have highlighted its use in MMS for treating MAC and PCACC. Toluidine blue exhibits staining advantages for these tumors. It may render isolated nests and perineural invasion of MAC more easily visible on frozen section.15

Although PCACC is rare, the recurrence rate is high.23 Toluidine blue has been used with MMS to ensure complete removal and higher cure rates. The metachromatic nature of TB is advantageous in staining the HA present in these tumors. Those who have reported the use of TB for PCACC prefer it to H&E for frozen sections.14

Technical Aspects—The staining time for TB-treated slides is reduced compared to H&E staining; staining can be efficiently done in frozen sections in less than 2.5 minutes using the method shown in Table 3.17 In comparison, typical H&E staining takes 9 minutes, and older TB techniques take 7 minutes.6

Rapid Toluidine Blue Staining Protocol

Conclusion

Toluidine blue may play an important and helpful role in the successful diagnosis and treatment of particular cutaneous tumors by providing additional diagnostic information. Although surgeons performing MMS will continue using the staining protocols with which they are most comfortable, adjunctive use of TB over time may provide an additional benefit at low risk for disrupting practice efficiency or workflow. Many Mohs surgeons are accustomed to using this stain, even preferring to interpret only TB-stained slides for cutaneous malignancy. Most published studies on this topic have been observational in nature, and additional controlled trials may be warranted to determine the effects on outcomes in real-world practice.

References
  1. Culling CF, Allison TR. Cellular Pathology Technique. 4th ed. Butterworths; 1985.
  2. Bergeron JA, Singer M. Metachromasy: an experimental and theoretical reevaluation. J Biophys Biochem Cytol. 1958;4:433-457. doi:10.1083/jcb.4.4.433
  3. Epstein JB, Scully C, Spinelli J. Toluidine blue and Lugol’s iodine application in the assessment of oral malignant disease and lesions at risk of malignancy. J Oral Pathol Med. 1992;21:160-163. doi:10.1111/j.1600-0714.1992.tb00094.x
  4. Warnakulasuriya KA, Johnson NW. Sensitivity and specificity of OraScan (R) toluidine blue mouthrinse in the detection of oral cancer and precancer. J Oral Pathol Med. 1996;25:97-103. doi:10.1111/j.1600-0714.1996.tb00201.x
  5. Silapunt S, Peterson SR, Alcalay J, et al. Mohs tissue mapping and processing: a survey study. Dermatol Surg. 2003;29:1109-1112; discussion 1112.
  6. Humphreys TR, Nemeth A, McCrevey S, et al. A pilot study comparing toluidine blue and hematoxylin and eosin staining of basal cell and squamous cell carcinoma during Mohs surgery. Dermatol Surg. 1996;22:693-697. doi:10.1111/j.1524-4725.1996.tb00619.x
  7. Bennett R, Taher M. Restylane persistent for 23 months found during Mohs micrographic surgery: a source of confusion with hyaluronic acid surrounding basal cell carcinoma. Dermatol Surg. 2005;31:1366-1369. doi:10.1111/j.1524-4725.2005.31223
  8. Goldberg LH, Wang SQ, Kimyai-Asadi A. The setting sun sign: visualizing the margins of a basal cell carcinoma on serial frozen sections stained with toluidine blue. Dermatol Surg. 2007;33:761-763. doi:10.1111/j.1524-4725.2007.33158.x
  9. Tehrani H, May K, Morris A, et al. Does the dual use of toluidine blue and hematoxylin and eosin staining improve basal cell carcinoma detection by Mohs surgery trainees? Dermatol Surg. 2013;39:995-1000. doi:10.1111/dsu.12180
  10. Donaldson MR, Weber LA. Toluidine blue supports differentiation of folliculocentric basaloid proliferation from basal cell carcinoma on frozen sections in a small single-practice cohort. Dermatol Surg. 2017;43:1303-1306. doi:10.1097/DSS.0000000000001107
  11. Styperek AR, Goldberg LH, Goldschmidt LE, et al. Toluidine blue and hematoxylin and eosin stains are comparable in evaluating squamous cell carcinoma during Mohs. Dermatol Surg. 2016;42:1279-1284. doi:10.1097/DSS.0000000000000872
  12. Trieu D, Drosou A, Goldberg LH, et al. Detecting spindle cell squamous cell carcinomas with toluidine blue on frozen sections. Dermatol Surg. 2014;40:1259-1260. doi:10.1097/DSS.0000000000000147
  13. Drosou A, Trieu D, Goldberg LH, et al. The perineural corona sign: enhancing detection of perineural squamous cell carcinoma during Mohs micrographic surgery with toluidine blue stain. J Am Acad Dermatol. 2014;71:826-827. doi:10.1016/j.jaad.2014.04.076
  14. Chesser RS, Bertler DE, Fitzpatrick JE, et al. Primary cutaneous adenoid cystic carcinoma treated with Mohs micrographic surgery toluidine blue technique. J Dermatol Surg Oncol. 1992;18:175-176. doi:10.1111/j.1524-4725.1992.tb02794.x
  15. Wang SQ, Goldberg LH, Nemeth A. The merits of adding toluidine blue-stained slides in Mohs surgery in the treatment of a microcystic adnexal carcinoma. J Am Acad Dermatol. 2007;56:1067-1069. doi:10.1016/j.jaad.2007.01.008
  16. Chen CL, Wilson S, Afzalneia R, et al. Topical aluminum chloride and Monsel’s solution block toluidine blue staining in Mohs frozen sections: mechanism and solution. Dermatol Surg. 2019;45:1019-1025. doi:10.1097/DSS.0000000000001761
  17. Todd MM, Lee JW, Marks VJ. Rapid toluidine blue stain for Mohs’ micrographic surgery. Dermatol Surg. 2005;31:244-245. doi:10.1111/j.1524-4725.2005.31053
  18. Picoto AM, Picoto A. Technical procedures for Mohs fresh tissue surgery. J Derm Surg Oncol. 1986;12:134-138. doi:10.1111/j.1524-4725.1986.tb01442.x
  19. Sperling LC, Winton GB. The transverse anatomy of androgenic alopecia. J Derm Surg Oncol. 1990;16:1127-1133. doi:10.1111/j.1524 -4725.1990.tb00024.x
  20. Smith-Zagone MJ, Schwartz MR. Frozen section of skin specimens. Arch Pathol Lab Med. 2005;129:1536-1543. doi:10.5858/2005-129-1536-FSOSS
  21. Moy RL, Potter TS, Uitto J. Increased glycosaminoglycans production in sclerosing basal cell carcinoma–derived fibroblasts and stimulation of normal skin fibroblast glycosaminoglycans production by a cytokine-derived from sclerosing basal cell carcinoma. Dermatol Surg. 2000;26:1029-1036. doi:10.1046/j.1524-4725.2000.0260111029.x
  22. Leshin B, White WL. Folliculocentric basaloid proliferation. The bulge (der Wulst) revisited. Arch Dermatol. 1990;126:900-906. doi:10.1001/archderm.126.7.900
  23. Seab JA, Graham JH. Primary cutaneous adenoid cystic carcinoma.J Am Acad Dermatol. 1987;17:113-118. doi:10.1016/s0190 -9622(87)70182-0
References
  1. Culling CF, Allison TR. Cellular Pathology Technique. 4th ed. Butterworths; 1985.
  2. Bergeron JA, Singer M. Metachromasy: an experimental and theoretical reevaluation. J Biophys Biochem Cytol. 1958;4:433-457. doi:10.1083/jcb.4.4.433
  3. Epstein JB, Scully C, Spinelli J. Toluidine blue and Lugol’s iodine application in the assessment of oral malignant disease and lesions at risk of malignancy. J Oral Pathol Med. 1992;21:160-163. doi:10.1111/j.1600-0714.1992.tb00094.x
  4. Warnakulasuriya KA, Johnson NW. Sensitivity and specificity of OraScan (R) toluidine blue mouthrinse in the detection of oral cancer and precancer. J Oral Pathol Med. 1996;25:97-103. doi:10.1111/j.1600-0714.1996.tb00201.x
  5. Silapunt S, Peterson SR, Alcalay J, et al. Mohs tissue mapping and processing: a survey study. Dermatol Surg. 2003;29:1109-1112; discussion 1112.
  6. Humphreys TR, Nemeth A, McCrevey S, et al. A pilot study comparing toluidine blue and hematoxylin and eosin staining of basal cell and squamous cell carcinoma during Mohs surgery. Dermatol Surg. 1996;22:693-697. doi:10.1111/j.1524-4725.1996.tb00619.x
  7. Bennett R, Taher M. Restylane persistent for 23 months found during Mohs micrographic surgery: a source of confusion with hyaluronic acid surrounding basal cell carcinoma. Dermatol Surg. 2005;31:1366-1369. doi:10.1111/j.1524-4725.2005.31223
  8. Goldberg LH, Wang SQ, Kimyai-Asadi A. The setting sun sign: visualizing the margins of a basal cell carcinoma on serial frozen sections stained with toluidine blue. Dermatol Surg. 2007;33:761-763. doi:10.1111/j.1524-4725.2007.33158.x
  9. Tehrani H, May K, Morris A, et al. Does the dual use of toluidine blue and hematoxylin and eosin staining improve basal cell carcinoma detection by Mohs surgery trainees? Dermatol Surg. 2013;39:995-1000. doi:10.1111/dsu.12180
  10. Donaldson MR, Weber LA. Toluidine blue supports differentiation of folliculocentric basaloid proliferation from basal cell carcinoma on frozen sections in a small single-practice cohort. Dermatol Surg. 2017;43:1303-1306. doi:10.1097/DSS.0000000000001107
  11. Styperek AR, Goldberg LH, Goldschmidt LE, et al. Toluidine blue and hematoxylin and eosin stains are comparable in evaluating squamous cell carcinoma during Mohs. Dermatol Surg. 2016;42:1279-1284. doi:10.1097/DSS.0000000000000872
  12. Trieu D, Drosou A, Goldberg LH, et al. Detecting spindle cell squamous cell carcinomas with toluidine blue on frozen sections. Dermatol Surg. 2014;40:1259-1260. doi:10.1097/DSS.0000000000000147
  13. Drosou A, Trieu D, Goldberg LH, et al. The perineural corona sign: enhancing detection of perineural squamous cell carcinoma during Mohs micrographic surgery with toluidine blue stain. J Am Acad Dermatol. 2014;71:826-827. doi:10.1016/j.jaad.2014.04.076
  14. Chesser RS, Bertler DE, Fitzpatrick JE, et al. Primary cutaneous adenoid cystic carcinoma treated with Mohs micrographic surgery toluidine blue technique. J Dermatol Surg Oncol. 1992;18:175-176. doi:10.1111/j.1524-4725.1992.tb02794.x
  15. Wang SQ, Goldberg LH, Nemeth A. The merits of adding toluidine blue-stained slides in Mohs surgery in the treatment of a microcystic adnexal carcinoma. J Am Acad Dermatol. 2007;56:1067-1069. doi:10.1016/j.jaad.2007.01.008
  16. Chen CL, Wilson S, Afzalneia R, et al. Topical aluminum chloride and Monsel’s solution block toluidine blue staining in Mohs frozen sections: mechanism and solution. Dermatol Surg. 2019;45:1019-1025. doi:10.1097/DSS.0000000000001761
  17. Todd MM, Lee JW, Marks VJ. Rapid toluidine blue stain for Mohs’ micrographic surgery. Dermatol Surg. 2005;31:244-245. doi:10.1111/j.1524-4725.2005.31053
  18. Picoto AM, Picoto A. Technical procedures for Mohs fresh tissue surgery. J Derm Surg Oncol. 1986;12:134-138. doi:10.1111/j.1524-4725.1986.tb01442.x
  19. Sperling LC, Winton GB. The transverse anatomy of androgenic alopecia. J Derm Surg Oncol. 1990;16:1127-1133. doi:10.1111/j.1524 -4725.1990.tb00024.x
  20. Smith-Zagone MJ, Schwartz MR. Frozen section of skin specimens. Arch Pathol Lab Med. 2005;129:1536-1543. doi:10.5858/2005-129-1536-FSOSS
  21. Moy RL, Potter TS, Uitto J. Increased glycosaminoglycans production in sclerosing basal cell carcinoma–derived fibroblasts and stimulation of normal skin fibroblast glycosaminoglycans production by a cytokine-derived from sclerosing basal cell carcinoma. Dermatol Surg. 2000;26:1029-1036. doi:10.1046/j.1524-4725.2000.0260111029.x
  22. Leshin B, White WL. Folliculocentric basaloid proliferation. The bulge (der Wulst) revisited. Arch Dermatol. 1990;126:900-906. doi:10.1001/archderm.126.7.900
  23. Seab JA, Graham JH. Primary cutaneous adenoid cystic carcinoma.J Am Acad Dermatol. 1987;17:113-118. doi:10.1016/s0190 -9622(87)70182-0
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E6-E11
Page Number
E6-E11
Publications
Publications
Topics
Article Type
Display Headline
The Role of Toluidine Blue in Mohs Micrographic Surgery: A Systematic Review
Display Headline
The Role of Toluidine Blue in Mohs Micrographic Surgery: A Systematic Review
Sections
Inside the Article

Practice Points

  • Toluidine blue (TB) staining can be integrated into Mohs micrographic surgery (MMS) for enhanced diagnosis of cutaneous tumors. Its metachromatic properties can aid in differentiating tumor cells from surrounding tissues, especially in basal cell carcinomas and squamous cell carcinomas.
  • It is important to develop expertise in interpreting TB-stained sections, as it may offer clearer visualization of nuclear details and stromal components, potentially leading to more accurate diagnosis and effective tumor margin identification.
  • Toluidine blue staining can be incorporated into routine MMS practice considering its quick staining process and low disruption to workflow. This can potentially improve diagnostic efficiency without significantly lengthening surgery time.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Reactive Angioendotheliomatosis Following Ad26.COV2.S Vaccination

Article Type
Changed
Thu, 12/21/2023 - 07:50
Display Headline
Reactive Angioendotheliomatosis Following Ad26.COV2.S Vaccination

To the Editor:

Reactive angioendotheliomatosis (RAE) is a rare self-limited cutaneous vascular proliferation of endothelial cells within blood vessels that manifests clinically as infiltrated red-blue patches and plaques with purpura that can progress to occlude vascular lumina. The etiology of RAE is mostly idiopathic; however, the disorder typically occurs in association with a range of systemic diseases, including infection, cryoglobulinemia, leukemia, antiphospholipid syndrome, peripheral vascular disease, and arteriovenous fistula. Histopathologic examination of these lesions shows marked proliferation of endothelial cells, including occlusion of the lumen of blood vessels over wide areas.

After ruling out malignancy, treatment of RAE focuses on targeting the underlying cause or disease, if any is present; 75% of reported cases occur in association with systemic disease.1 Onset can occur at any age without predilection for sex. Reactive angioendotheliomatosis commonly manifests on the extremities but may occur on the head and neck in rare instances.2

The rarity of the condition and its poorly defined clinical characteristics make it difficult to develop a treatment plan. There are no standardized treatment guidelines for the reactive form of angiomatosis. We report a case of RAE that developed 2 weeks after vaccination with the Ad26.COV2.S vaccine (Johnson & Johnson Innovative Medicine [formerly Janssen Pharmaceutical Companies of Johnson & Johnson]) that improved following 2 weeks of treatment with a topical corticosteroid and an oral antihistamine.

A 58-year-old man presented to an outpatient dermatology clinic with pruritus and occasional paresthesia associated with a rash over the left arm of 1 month’s duration. The patient suspected that the rash may have formed secondary to the bite of oak mites on the arms and chest while he was carrying milled wood. Further inquiry into the patient’s history revealed that he received the Ad26.COV2.S vaccine 2 weeks prior to the appearance of the rash. He denied mechanical trauma. His medical history included hypercholesterolemia and a mild COVID-19 infection 8 months prior to the appearance of the rash that did not require hospitalization. He denied fever or chills during the 2 weeks following vaccination. The pruritus was minimally relieved for short periods with over-the-counter calamine lotion. The patient’s medication regimen included daily pravastatin and loratadine at the time of the initial visit. He used acetaminophen as needed for knee pain.

A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.
FIGURE 1. A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.

Physical examination revealed palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula (Figure 1A), left anterolateral shoulder, left lateral volar forearm, and thenar eminence of the left hand (Figure 1B). Notably, the entire right arm, conjunctivae, tongue, lips, and bilateral fingernails were clear. Three 4-mm punch biopsies were performed at the initial presentation: 1 perilesional biopsy for direct immunofluorescence testing and 2 lesional biopsies for routine histologic evaluation. An extensive serologic workup failed to reveal abnormalities. An activated partial thromboplastin time, dilute Russell viper venom time, serum protein electrophoresis, and levels of rheumatoid factor and angiotensin-converting enzyme were within reference range. Anticardiolipin antibodies IgA, IgM, and IgG were negative. A cryoglobulin test was negative.

Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).
FIGURE 2. Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).

Histopathology revealed a proliferation of irregularly shaped vascular spaces with plump endothelium in the papillary dermis (Figure 2). Scattered leukocyte common antigen-positive lymphocytes were noted within lesions. The epidermis appeared normal, without evidence of spongiosis or alteration of the stratum corneum. Immunohistochemical studies of the perilesional skin biopsy revealed positivity for CD31 and D2-40 (Figure 3). Specimens were negative for CD20 and human herpesvirus 8. Direct immunofluorescence of the perilesional biopsy was negative.

Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).
FIGURE 3. Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).

A diagnosis of RAE was made based on clinical and histologic findings. Treatment with triamcinolone ointment 0.1% twice daily and oral cetirizine 10 mg twice daily was initiated. Re-evaluation 2 weeks later revealed notable improvement in the affected areas, including decreased edema, improvement of the purpura, and absence of pruritus. The patient noted no further spread or blister formation while the active areas were being treated with the topical steroid. The treatment regimen was modified to triamcinolone ointment 0.1% once daily, and cetirizine was discontinued. At 3-month follow-up, active areas had completely resolved (Figure 4) and triamcinolone was discontinued. To date, the patient has not had recurrence of symptoms and remains healthy.

At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.
FIGURE 4. A and B, At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.

 

 

Gottron and Nikolowski3 reported the first case of RAE in an adult patient who presented with purpuric patches secondary to skin infarction. Current definitions use the umbrella term cutaneous reactive angiomatosis to cover 3 major subtypes: reactive angioendotheliomatosis, diffuse dermal angioendotheliomatosis, and acroangiodermatitis (pseudo-Kaposi sarcoma [KS]). The manifestation of these subgroups is clinically similar, and they must be differentiated through histologic evaluation.4

Reactive angioendotheliomatosis has an unknown pathogenesis and is poorly defined clinically. The exact pathophysiology is unknown but likely is linked to vaso-occlusion and hypoxia.1 A PubMed search of articles indexed for MEDLINE, as well as a review of Science Direct, Google Scholar, and Cochrane Library, using the terms reactive angioendotheliomatosis, COVID, vaccine, Ad26.COV2.S, and RAE in any combination revealed no prior cases of RAE in association with Ad26.COV2.S vaccination.

By the late 1980s, systemic angioendotheliomatosis was segregated into 2 distinct entities: malignant and reactive.4 The differential diagnosis of malignant systemic angioendotheliomatosis includes KS and angiosarcoma; nonmalignant causes are the variants of cutaneous reactive angiomatosis. It is important to rule out KS because of its malignant and deceptive nature. It is unknown if KS originates in blood vessels or lymphatic endothelial cells; however, evidence is strongly in favor of blood vessel origin using CD31 and CD34 endothelial markers.5 CD34 positivity is more reliable than CD31 in diagnosing KS, but the absence of both markers does not offer enough evidence to rule out KS on its own.6

In our patient, histopathology revealed cells positive for CD31 and D2-40; the latter is a lymphatic endothelial cell marker that stains the endothelium of lymphatic channels but not blood vessels.7 Positive D2-40 can be indicative of KS and non-KS lesions, each with a distinct staining pattern. D2-40 staining on non-KS lesions is confined to lymphatic vessels, as it was in our patient; in contrast, spindle-shaped cells also will be stained in KS lesions.8

Another cell marker, CD20, is a B cell–specific protein that can be measured to help diagnose malignant diseases such as B-cell lymphoma and leukemia. Human herpesvirus 8 (also known as KS-associated herpesvirus) is the infectious cause of KS and traditionally has been detected using methods such as the polymerase chain reaction.9,10

Most cases of RAE are idiopathic and occur in association with systemic disease, which was not the case in our patient. We speculated that his reaction was most likely triggered by vascular transfection of endothelial cells secondary to Ad26.COV2.S vaccination. Alternatively, vaccination may have caused vascular occlusion, though the lack of cyanosis, nail changes, and route of inoculant make this less likely.

All approved COVID-19 vaccines are designed solely for intramuscular injection. In comparison to other types of tissue, muscles have superior vascularity, allowing for enhanced mobilization of compounds, which results in faster systemic circulation.11 Alternative methods of injection, including intravascular, subcutaneous, and intradermal, may lead to decreased efficacy or adverse events, or both.

 

 

Prior cases of RAE have been treated with laser therapy, topical or systemic corticosteroids, excisional removal, or topical β-blockers, such as timolol.12β-Blocking agents act on β-adrenergic receptors on endothelial cells to inhibit angiogenesis by reducing release of blood vessel growth-signaling molecules and triggering apoptosis. In this patient, topical steroids and oral antihistamines were sufficient treatment.

Vaccine-related adverse events have been reported but remain rare. The benefits of Ad26.COV2.S vaccination for protection against COVID-19 outweigh the extremely low risk for adverse events.13 For that reason, the Centers for Disease Control and Prevention recommends a booster for individuals who are eligible to maximize protection. Intramuscular injection of Ad26.COV2.S resulted in a lower incidence of moderate to severe COVID-19 cases in all age groups vs the placebo group. Hypersensitivity adverse events were reported in 0.4% of Ad26.COV2.S-vaccinated patients vs 0.4% of patients who received a placebo; the more common reactions were nonanaphylactic.13

There have been 12 reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, which sparked nationwide controversy over the safety of the Ad26.COV2.S vaccine.14 After further investigation into those reports, the US Food and Drug Administration and the Centers for Disease Control and Prevention concluded that the benefits of the Ad26.COV2.S vaccine outweigh the low risk for associated thrombosis.15

Although adverse reactions are rare, it is important that health care providers take proper safety measures before and while administering any COVID-19 vaccine. Patients should be screened for contraindications to the COVID-19 vaccine to mitigate adverse effects seen in the small percentage of patients who may need to take alternative precautions.

The broad tissue tropism and high transmissibility of SARS-CoV-2 are the main contributors to its infection having reached pandemic scale. The spike (S) protein on SARS-CoV-2 binds to ACE2, the most thoroughly studied SARS-CoV-2 receptor, which is found in a range of tissues, including arterial endothelial cells, leading to its transfection. Several studies have proposed that expression of the S protein causes endothelial dysfunction through cytokine release, activation of complement, and ultimately microvascular occlusion.16

Recent developments in the use of viral-like particles, such as vesicular stomatitis virus, may mitigate future cases of RAE that are associated with endothelial cell transfection. Vesicular stomatitis virus is a popular model virus for research applications due to its glycoprotein and matrix protein contributing to its broad tropism. Recent efforts to alter these proteins have successfully limited the broad tropism of vesicular stomatitis virus.17

The SARS-CoV-2 virus must be handled in a Biosafety Level 3 laboratory. Conversely, pseudoviruses can be handled in lower containment facilities due to their safe and efficacious nature, offering an avenue to expedite vaccine development against many viral outbreaks, including SARS-CoV-2.18

 

 

An increasing number of cutaneous manifestations have been associated with COVID-19 infection and vaccination. Eruptive pseudoangiomatosis, a rare self-limiting exanthem, has been reported in association with ­COVID-19 vaccination.19 Eruptive pseudoangiomatosis manifests as erythematous blanchable papules that resemble angiomas, typically in a widespread distribution. Eruptive pseudoangiomatosis has striking similarities to RAE histologically; both manifest as dilated dermal blood vessels with plump endothelial cells.

Our case is unique because of the vasculitic palpable nature of the lesions, which were localized to the left arm. Eruptive pseudoangiomatosis formation after COVID-19 infection or SARS-CoV-2 vaccination may suggest alteration of ACE2 by binding of S protein.20 Such alteration of the ACE2 pathway would lead to inflammation of angiotensin II, causing proliferation of endothelial cells in the formation of angiomalike lesions. This hypothesis suggests a paraviral eruption secondary to an immunologic reaction, not a classical virtual eruption from direct contact of the virus on blood vessels. Although EPA and RAE are harmless and self-limiting, these reports will spread awareness of the increasing number of skin manifestations related to COVID-19 and SARS-CoV-2 virus vaccination.

Acknowledgment—Thoughtful insights and comments on this manuscript were provided by Christine J. Ko, MD (New Haven, Connecticut); Christine L. Egan, MD (Glen Mills, Pennsylvania); Howard A. Bueller, MD (Delray Beach, Florida); and Juan Pablo Robles, PhD (Juriquilla, Mexico).

References
  1. McMenamin ME, Fletcher CDM. Reactive angioendotheliomatosis: a study of 15 cases demonstrating a wide clinicopathologic spectrum. Am J Surg Pathol. 2002;26:686-697. doi:10.1097/00000478-200206000-00001
  2. Khan S, Pujani M, Jetley S, et al. Angiomatosis: a rare vascular proliferation of head and neck region. J Cutan Aesthet Surg. 2015;8:108-110. doi:10.4103/0974-2077.158448
  3. Gottron HA, Nikolowski W. Extrarenal Lohlein focal nephritis of the skin in endocarditis. Arch Klin Exp Dermatol. 1958;207:156-176.
  4. Cooper PH. Angioendotheliomatosis: two separate diseases. J Cutan Pathol. 1988;15:259. doi:10.1111/j.1600-0560.1988.tb00556.x
  5. Cancian L, Hansen A, Boshoff C. Cellular origin of Kaposi’s sarcoma and Kaposi’s sarcoma-associated herpesvirus-induced cell reprogramming. Trends Cell Biol. Sep 2013;23:421-32. doi:10.1016/j.tcb.2013.04.001
  6. Russell Jones R, Orchard G, Zelger B, et al. Immunostaining for CD31 and CD34 in Kaposi sarcoma. J Clin Pathol. 1995;48:1011-1016. doi:10.1136/jcp.48.11.1011
  7. Kahn HJ, Bailey D, Marks A. Monoclonal antibody D2-40, a new marker of lymphatic endothelium, reacts with Kaposi’s sarcoma and a subset of angiosarcomas. Mod Pathol. 2002;15:434-440. doi:10.1038/modpathol.3880543
  8. Genedy RM, Hamza AM, Abdel Latef AA, et al. Sensitivity and specificity of D2-40 in differentiating Kaposi sarcoma from its mimickers. J Egyptian Womens Dermatolog Soc. 2021;18:67-74. doi:10.4103/jewd.jewd_61_20
  9. Mesri EA, Cesarman E, Boshoff C. Kaposi’s sarcoma and its associated herpesvirus. Nat Rev Cancer. 2010;10:707-719. doi:10.1038/nrc2888
  10. Patel RM, Goldblum JR, Hsi ED. Immunohistochemical detection of human herpes virus-8 latent nuclear antigen-1 is useful in the diagnosis of Kaposi sarcoma. Mod Pathol. 2004;17:456-460. doi:10.1038/modpathol.3800061
  11. Zuckerman JN. The importance of injecting vaccines into muscle. Different patients need different needle sizes. BMJ. 2000;321:1237-1238. doi:10.1136/bmj.321.7271.1237
  12. Bhatia R, Hazarika N, Chandrasekaran D, et al. Treatment of posttraumatic reactive angioendotheliomatosis with topical timolol maleate. JAMA Dermatol. 2021;157:1002-1004. doi:10.1001/jamadermatol.2021.1770
  13. Sadoff J, Gray G, Vandebosch A, et al; ENSEMBLE Study Group. Safety and efficacy of single-dose Ad26.COV2.S vaccine against Covid-19. N Engl J Med. 2021;384:2187-2201. doi:10.1056/NEJMoa2101544
  14. See I, Su JR, Lale A, et al. US case reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, March 2 to April 21, 2021. JAMA. 2021;325:2448-2456. doi:10.1001/jama.2021.7517
  15. Berry CT, Eliliwi M, Gallagher S, et al. Cutaneous small vessel vasculitis following single-dose Janssen Ad26.COV2.S vaccination. JAAD Case Rep. 2021;15:11-14. doi:10.1016/j.jdcr.2021.07.002
  16. Flaumenhaft R, Enjyoji K, Schmaier AA. Vasculopathy in COVID-19. Blood. 2022;140:222-235. doi:10.1182/blood.2021012250
  17. Hastie E, Cataldi M, Marriott I, et al. Understanding and altering cell tropism of vesicular stomatitis virus. Virus Res. 2013;176:16-32. doi:10.1016/j.virusres.2013.06.003
  18. Xiong H-L, Wu Y-T, Cao J-L, et al. Robust neutralization assay based on SARS-CoV-2 S-protein-bearing vesicular stomatitis virus (VSV) pseudovirus and ACE2-overexpressing BHK21 cells. Emerg Microbes Infect. 2020;9:2105-2113. doi:10.1080/22221751.2020.1815589
  19. Mohta A, Jain SK, Mehta RD, et al. Development of eruptive pseudoangiomatosis following COVID-19 immunization – apropos of 5 cases. J Eur Acad Dermatol Venereol. 2021;35:e722-e725. doi:10.1111/jdv.17499
  20. Angeli F, Spanevello A, Reboldi G, et al. SARS-CoV-2 vaccines: lights and shadows. Eur J Intern Med. 2021;88:1-8. doi:10.1016/j.ejim.2021.04.019
Article PDF
Author and Disclosure Information

Collin Faulkner is from the State University of New York at Buffalo. Dr. Jabbour is from the State University of New York Upstate Medical University, Syracuse. Dr. Kanik is from CBLPath, Rye Brook, New York. Dr. Schoeneck is from FamilyCare Medical Group, Camillus, New York. Dr. Tangoren is from I. A. Tangoren, MD, PLLC, Dermatology & Dermatologic Surgery, Syracuse.

The authors report no conflict of interest.

Correspondence: Austin J. Jabbour, MD, 850 Republican St, Seattle WA 98109 (austin.jabbour@gmail.com).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E20-E23
Sections
Author and Disclosure Information

Collin Faulkner is from the State University of New York at Buffalo. Dr. Jabbour is from the State University of New York Upstate Medical University, Syracuse. Dr. Kanik is from CBLPath, Rye Brook, New York. Dr. Schoeneck is from FamilyCare Medical Group, Camillus, New York. Dr. Tangoren is from I. A. Tangoren, MD, PLLC, Dermatology & Dermatologic Surgery, Syracuse.

The authors report no conflict of interest.

Correspondence: Austin J. Jabbour, MD, 850 Republican St, Seattle WA 98109 (austin.jabbour@gmail.com).

Author and Disclosure Information

Collin Faulkner is from the State University of New York at Buffalo. Dr. Jabbour is from the State University of New York Upstate Medical University, Syracuse. Dr. Kanik is from CBLPath, Rye Brook, New York. Dr. Schoeneck is from FamilyCare Medical Group, Camillus, New York. Dr. Tangoren is from I. A. Tangoren, MD, PLLC, Dermatology & Dermatologic Surgery, Syracuse.

The authors report no conflict of interest.

Correspondence: Austin J. Jabbour, MD, 850 Republican St, Seattle WA 98109 (austin.jabbour@gmail.com).

Article PDF
Article PDF

To the Editor:

Reactive angioendotheliomatosis (RAE) is a rare self-limited cutaneous vascular proliferation of endothelial cells within blood vessels that manifests clinically as infiltrated red-blue patches and plaques with purpura that can progress to occlude vascular lumina. The etiology of RAE is mostly idiopathic; however, the disorder typically occurs in association with a range of systemic diseases, including infection, cryoglobulinemia, leukemia, antiphospholipid syndrome, peripheral vascular disease, and arteriovenous fistula. Histopathologic examination of these lesions shows marked proliferation of endothelial cells, including occlusion of the lumen of blood vessels over wide areas.

After ruling out malignancy, treatment of RAE focuses on targeting the underlying cause or disease, if any is present; 75% of reported cases occur in association with systemic disease.1 Onset can occur at any age without predilection for sex. Reactive angioendotheliomatosis commonly manifests on the extremities but may occur on the head and neck in rare instances.2

The rarity of the condition and its poorly defined clinical characteristics make it difficult to develop a treatment plan. There are no standardized treatment guidelines for the reactive form of angiomatosis. We report a case of RAE that developed 2 weeks after vaccination with the Ad26.COV2.S vaccine (Johnson & Johnson Innovative Medicine [formerly Janssen Pharmaceutical Companies of Johnson & Johnson]) that improved following 2 weeks of treatment with a topical corticosteroid and an oral antihistamine.

A 58-year-old man presented to an outpatient dermatology clinic with pruritus and occasional paresthesia associated with a rash over the left arm of 1 month’s duration. The patient suspected that the rash may have formed secondary to the bite of oak mites on the arms and chest while he was carrying milled wood. Further inquiry into the patient’s history revealed that he received the Ad26.COV2.S vaccine 2 weeks prior to the appearance of the rash. He denied mechanical trauma. His medical history included hypercholesterolemia and a mild COVID-19 infection 8 months prior to the appearance of the rash that did not require hospitalization. He denied fever or chills during the 2 weeks following vaccination. The pruritus was minimally relieved for short periods with over-the-counter calamine lotion. The patient’s medication regimen included daily pravastatin and loratadine at the time of the initial visit. He used acetaminophen as needed for knee pain.

A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.
FIGURE 1. A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.

Physical examination revealed palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula (Figure 1A), left anterolateral shoulder, left lateral volar forearm, and thenar eminence of the left hand (Figure 1B). Notably, the entire right arm, conjunctivae, tongue, lips, and bilateral fingernails were clear. Three 4-mm punch biopsies were performed at the initial presentation: 1 perilesional biopsy for direct immunofluorescence testing and 2 lesional biopsies for routine histologic evaluation. An extensive serologic workup failed to reveal abnormalities. An activated partial thromboplastin time, dilute Russell viper venom time, serum protein electrophoresis, and levels of rheumatoid factor and angiotensin-converting enzyme were within reference range. Anticardiolipin antibodies IgA, IgM, and IgG were negative. A cryoglobulin test was negative.

Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).
FIGURE 2. Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).

Histopathology revealed a proliferation of irregularly shaped vascular spaces with plump endothelium in the papillary dermis (Figure 2). Scattered leukocyte common antigen-positive lymphocytes were noted within lesions. The epidermis appeared normal, without evidence of spongiosis or alteration of the stratum corneum. Immunohistochemical studies of the perilesional skin biopsy revealed positivity for CD31 and D2-40 (Figure 3). Specimens were negative for CD20 and human herpesvirus 8. Direct immunofluorescence of the perilesional biopsy was negative.

Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).
FIGURE 3. Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).

A diagnosis of RAE was made based on clinical and histologic findings. Treatment with triamcinolone ointment 0.1% twice daily and oral cetirizine 10 mg twice daily was initiated. Re-evaluation 2 weeks later revealed notable improvement in the affected areas, including decreased edema, improvement of the purpura, and absence of pruritus. The patient noted no further spread or blister formation while the active areas were being treated with the topical steroid. The treatment regimen was modified to triamcinolone ointment 0.1% once daily, and cetirizine was discontinued. At 3-month follow-up, active areas had completely resolved (Figure 4) and triamcinolone was discontinued. To date, the patient has not had recurrence of symptoms and remains healthy.

At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.
FIGURE 4. A and B, At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.

 

 

Gottron and Nikolowski3 reported the first case of RAE in an adult patient who presented with purpuric patches secondary to skin infarction. Current definitions use the umbrella term cutaneous reactive angiomatosis to cover 3 major subtypes: reactive angioendotheliomatosis, diffuse dermal angioendotheliomatosis, and acroangiodermatitis (pseudo-Kaposi sarcoma [KS]). The manifestation of these subgroups is clinically similar, and they must be differentiated through histologic evaluation.4

Reactive angioendotheliomatosis has an unknown pathogenesis and is poorly defined clinically. The exact pathophysiology is unknown but likely is linked to vaso-occlusion and hypoxia.1 A PubMed search of articles indexed for MEDLINE, as well as a review of Science Direct, Google Scholar, and Cochrane Library, using the terms reactive angioendotheliomatosis, COVID, vaccine, Ad26.COV2.S, and RAE in any combination revealed no prior cases of RAE in association with Ad26.COV2.S vaccination.

By the late 1980s, systemic angioendotheliomatosis was segregated into 2 distinct entities: malignant and reactive.4 The differential diagnosis of malignant systemic angioendotheliomatosis includes KS and angiosarcoma; nonmalignant causes are the variants of cutaneous reactive angiomatosis. It is important to rule out KS because of its malignant and deceptive nature. It is unknown if KS originates in blood vessels or lymphatic endothelial cells; however, evidence is strongly in favor of blood vessel origin using CD31 and CD34 endothelial markers.5 CD34 positivity is more reliable than CD31 in diagnosing KS, but the absence of both markers does not offer enough evidence to rule out KS on its own.6

In our patient, histopathology revealed cells positive for CD31 and D2-40; the latter is a lymphatic endothelial cell marker that stains the endothelium of lymphatic channels but not blood vessels.7 Positive D2-40 can be indicative of KS and non-KS lesions, each with a distinct staining pattern. D2-40 staining on non-KS lesions is confined to lymphatic vessels, as it was in our patient; in contrast, spindle-shaped cells also will be stained in KS lesions.8

Another cell marker, CD20, is a B cell–specific protein that can be measured to help diagnose malignant diseases such as B-cell lymphoma and leukemia. Human herpesvirus 8 (also known as KS-associated herpesvirus) is the infectious cause of KS and traditionally has been detected using methods such as the polymerase chain reaction.9,10

Most cases of RAE are idiopathic and occur in association with systemic disease, which was not the case in our patient. We speculated that his reaction was most likely triggered by vascular transfection of endothelial cells secondary to Ad26.COV2.S vaccination. Alternatively, vaccination may have caused vascular occlusion, though the lack of cyanosis, nail changes, and route of inoculant make this less likely.

All approved COVID-19 vaccines are designed solely for intramuscular injection. In comparison to other types of tissue, muscles have superior vascularity, allowing for enhanced mobilization of compounds, which results in faster systemic circulation.11 Alternative methods of injection, including intravascular, subcutaneous, and intradermal, may lead to decreased efficacy or adverse events, or both.

 

 

Prior cases of RAE have been treated with laser therapy, topical or systemic corticosteroids, excisional removal, or topical β-blockers, such as timolol.12β-Blocking agents act on β-adrenergic receptors on endothelial cells to inhibit angiogenesis by reducing release of blood vessel growth-signaling molecules and triggering apoptosis. In this patient, topical steroids and oral antihistamines were sufficient treatment.

Vaccine-related adverse events have been reported but remain rare. The benefits of Ad26.COV2.S vaccination for protection against COVID-19 outweigh the extremely low risk for adverse events.13 For that reason, the Centers for Disease Control and Prevention recommends a booster for individuals who are eligible to maximize protection. Intramuscular injection of Ad26.COV2.S resulted in a lower incidence of moderate to severe COVID-19 cases in all age groups vs the placebo group. Hypersensitivity adverse events were reported in 0.4% of Ad26.COV2.S-vaccinated patients vs 0.4% of patients who received a placebo; the more common reactions were nonanaphylactic.13

There have been 12 reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, which sparked nationwide controversy over the safety of the Ad26.COV2.S vaccine.14 After further investigation into those reports, the US Food and Drug Administration and the Centers for Disease Control and Prevention concluded that the benefits of the Ad26.COV2.S vaccine outweigh the low risk for associated thrombosis.15

Although adverse reactions are rare, it is important that health care providers take proper safety measures before and while administering any COVID-19 vaccine. Patients should be screened for contraindications to the COVID-19 vaccine to mitigate adverse effects seen in the small percentage of patients who may need to take alternative precautions.

The broad tissue tropism and high transmissibility of SARS-CoV-2 are the main contributors to its infection having reached pandemic scale. The spike (S) protein on SARS-CoV-2 binds to ACE2, the most thoroughly studied SARS-CoV-2 receptor, which is found in a range of tissues, including arterial endothelial cells, leading to its transfection. Several studies have proposed that expression of the S protein causes endothelial dysfunction through cytokine release, activation of complement, and ultimately microvascular occlusion.16

Recent developments in the use of viral-like particles, such as vesicular stomatitis virus, may mitigate future cases of RAE that are associated with endothelial cell transfection. Vesicular stomatitis virus is a popular model virus for research applications due to its glycoprotein and matrix protein contributing to its broad tropism. Recent efforts to alter these proteins have successfully limited the broad tropism of vesicular stomatitis virus.17

The SARS-CoV-2 virus must be handled in a Biosafety Level 3 laboratory. Conversely, pseudoviruses can be handled in lower containment facilities due to their safe and efficacious nature, offering an avenue to expedite vaccine development against many viral outbreaks, including SARS-CoV-2.18

 

 

An increasing number of cutaneous manifestations have been associated with COVID-19 infection and vaccination. Eruptive pseudoangiomatosis, a rare self-limiting exanthem, has been reported in association with ­COVID-19 vaccination.19 Eruptive pseudoangiomatosis manifests as erythematous blanchable papules that resemble angiomas, typically in a widespread distribution. Eruptive pseudoangiomatosis has striking similarities to RAE histologically; both manifest as dilated dermal blood vessels with plump endothelial cells.

Our case is unique because of the vasculitic palpable nature of the lesions, which were localized to the left arm. Eruptive pseudoangiomatosis formation after COVID-19 infection or SARS-CoV-2 vaccination may suggest alteration of ACE2 by binding of S protein.20 Such alteration of the ACE2 pathway would lead to inflammation of angiotensin II, causing proliferation of endothelial cells in the formation of angiomalike lesions. This hypothesis suggests a paraviral eruption secondary to an immunologic reaction, not a classical virtual eruption from direct contact of the virus on blood vessels. Although EPA and RAE are harmless and self-limiting, these reports will spread awareness of the increasing number of skin manifestations related to COVID-19 and SARS-CoV-2 virus vaccination.

Acknowledgment—Thoughtful insights and comments on this manuscript were provided by Christine J. Ko, MD (New Haven, Connecticut); Christine L. Egan, MD (Glen Mills, Pennsylvania); Howard A. Bueller, MD (Delray Beach, Florida); and Juan Pablo Robles, PhD (Juriquilla, Mexico).

To the Editor:

Reactive angioendotheliomatosis (RAE) is a rare self-limited cutaneous vascular proliferation of endothelial cells within blood vessels that manifests clinically as infiltrated red-blue patches and plaques with purpura that can progress to occlude vascular lumina. The etiology of RAE is mostly idiopathic; however, the disorder typically occurs in association with a range of systemic diseases, including infection, cryoglobulinemia, leukemia, antiphospholipid syndrome, peripheral vascular disease, and arteriovenous fistula. Histopathologic examination of these lesions shows marked proliferation of endothelial cells, including occlusion of the lumen of blood vessels over wide areas.

After ruling out malignancy, treatment of RAE focuses on targeting the underlying cause or disease, if any is present; 75% of reported cases occur in association with systemic disease.1 Onset can occur at any age without predilection for sex. Reactive angioendotheliomatosis commonly manifests on the extremities but may occur on the head and neck in rare instances.2

The rarity of the condition and its poorly defined clinical characteristics make it difficult to develop a treatment plan. There are no standardized treatment guidelines for the reactive form of angiomatosis. We report a case of RAE that developed 2 weeks after vaccination with the Ad26.COV2.S vaccine (Johnson & Johnson Innovative Medicine [formerly Janssen Pharmaceutical Companies of Johnson & Johnson]) that improved following 2 weeks of treatment with a topical corticosteroid and an oral antihistamine.

A 58-year-old man presented to an outpatient dermatology clinic with pruritus and occasional paresthesia associated with a rash over the left arm of 1 month’s duration. The patient suspected that the rash may have formed secondary to the bite of oak mites on the arms and chest while he was carrying milled wood. Further inquiry into the patient’s history revealed that he received the Ad26.COV2.S vaccine 2 weeks prior to the appearance of the rash. He denied mechanical trauma. His medical history included hypercholesterolemia and a mild COVID-19 infection 8 months prior to the appearance of the rash that did not require hospitalization. He denied fever or chills during the 2 weeks following vaccination. The pruritus was minimally relieved for short periods with over-the-counter calamine lotion. The patient’s medication regimen included daily pravastatin and loratadine at the time of the initial visit. He used acetaminophen as needed for knee pain.

A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.
FIGURE 1. A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.

Physical examination revealed palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula (Figure 1A), left anterolateral shoulder, left lateral volar forearm, and thenar eminence of the left hand (Figure 1B). Notably, the entire right arm, conjunctivae, tongue, lips, and bilateral fingernails were clear. Three 4-mm punch biopsies were performed at the initial presentation: 1 perilesional biopsy for direct immunofluorescence testing and 2 lesional biopsies for routine histologic evaluation. An extensive serologic workup failed to reveal abnormalities. An activated partial thromboplastin time, dilute Russell viper venom time, serum protein electrophoresis, and levels of rheumatoid factor and angiotensin-converting enzyme were within reference range. Anticardiolipin antibodies IgA, IgM, and IgG were negative. A cryoglobulin test was negative.

Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).
FIGURE 2. Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).

Histopathology revealed a proliferation of irregularly shaped vascular spaces with plump endothelium in the papillary dermis (Figure 2). Scattered leukocyte common antigen-positive lymphocytes were noted within lesions. The epidermis appeared normal, without evidence of spongiosis or alteration of the stratum corneum. Immunohistochemical studies of the perilesional skin biopsy revealed positivity for CD31 and D2-40 (Figure 3). Specimens were negative for CD20 and human herpesvirus 8. Direct immunofluorescence of the perilesional biopsy was negative.

Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).
FIGURE 3. Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).

A diagnosis of RAE was made based on clinical and histologic findings. Treatment with triamcinolone ointment 0.1% twice daily and oral cetirizine 10 mg twice daily was initiated. Re-evaluation 2 weeks later revealed notable improvement in the affected areas, including decreased edema, improvement of the purpura, and absence of pruritus. The patient noted no further spread or blister formation while the active areas were being treated with the topical steroid. The treatment regimen was modified to triamcinolone ointment 0.1% once daily, and cetirizine was discontinued. At 3-month follow-up, active areas had completely resolved (Figure 4) and triamcinolone was discontinued. To date, the patient has not had recurrence of symptoms and remains healthy.

At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.
FIGURE 4. A and B, At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.

 

 

Gottron and Nikolowski3 reported the first case of RAE in an adult patient who presented with purpuric patches secondary to skin infarction. Current definitions use the umbrella term cutaneous reactive angiomatosis to cover 3 major subtypes: reactive angioendotheliomatosis, diffuse dermal angioendotheliomatosis, and acroangiodermatitis (pseudo-Kaposi sarcoma [KS]). The manifestation of these subgroups is clinically similar, and they must be differentiated through histologic evaluation.4

Reactive angioendotheliomatosis has an unknown pathogenesis and is poorly defined clinically. The exact pathophysiology is unknown but likely is linked to vaso-occlusion and hypoxia.1 A PubMed search of articles indexed for MEDLINE, as well as a review of Science Direct, Google Scholar, and Cochrane Library, using the terms reactive angioendotheliomatosis, COVID, vaccine, Ad26.COV2.S, and RAE in any combination revealed no prior cases of RAE in association with Ad26.COV2.S vaccination.

By the late 1980s, systemic angioendotheliomatosis was segregated into 2 distinct entities: malignant and reactive.4 The differential diagnosis of malignant systemic angioendotheliomatosis includes KS and angiosarcoma; nonmalignant causes are the variants of cutaneous reactive angiomatosis. It is important to rule out KS because of its malignant and deceptive nature. It is unknown if KS originates in blood vessels or lymphatic endothelial cells; however, evidence is strongly in favor of blood vessel origin using CD31 and CD34 endothelial markers.5 CD34 positivity is more reliable than CD31 in diagnosing KS, but the absence of both markers does not offer enough evidence to rule out KS on its own.6

In our patient, histopathology revealed cells positive for CD31 and D2-40; the latter is a lymphatic endothelial cell marker that stains the endothelium of lymphatic channels but not blood vessels.7 Positive D2-40 can be indicative of KS and non-KS lesions, each with a distinct staining pattern. D2-40 staining on non-KS lesions is confined to lymphatic vessels, as it was in our patient; in contrast, spindle-shaped cells also will be stained in KS lesions.8

Another cell marker, CD20, is a B cell–specific protein that can be measured to help diagnose malignant diseases such as B-cell lymphoma and leukemia. Human herpesvirus 8 (also known as KS-associated herpesvirus) is the infectious cause of KS and traditionally has been detected using methods such as the polymerase chain reaction.9,10

Most cases of RAE are idiopathic and occur in association with systemic disease, which was not the case in our patient. We speculated that his reaction was most likely triggered by vascular transfection of endothelial cells secondary to Ad26.COV2.S vaccination. Alternatively, vaccination may have caused vascular occlusion, though the lack of cyanosis, nail changes, and route of inoculant make this less likely.

All approved COVID-19 vaccines are designed solely for intramuscular injection. In comparison to other types of tissue, muscles have superior vascularity, allowing for enhanced mobilization of compounds, which results in faster systemic circulation.11 Alternative methods of injection, including intravascular, subcutaneous, and intradermal, may lead to decreased efficacy or adverse events, or both.

 

 

Prior cases of RAE have been treated with laser therapy, topical or systemic corticosteroids, excisional removal, or topical β-blockers, such as timolol.12β-Blocking agents act on β-adrenergic receptors on endothelial cells to inhibit angiogenesis by reducing release of blood vessel growth-signaling molecules and triggering apoptosis. In this patient, topical steroids and oral antihistamines were sufficient treatment.

Vaccine-related adverse events have been reported but remain rare. The benefits of Ad26.COV2.S vaccination for protection against COVID-19 outweigh the extremely low risk for adverse events.13 For that reason, the Centers for Disease Control and Prevention recommends a booster for individuals who are eligible to maximize protection. Intramuscular injection of Ad26.COV2.S resulted in a lower incidence of moderate to severe COVID-19 cases in all age groups vs the placebo group. Hypersensitivity adverse events were reported in 0.4% of Ad26.COV2.S-vaccinated patients vs 0.4% of patients who received a placebo; the more common reactions were nonanaphylactic.13

There have been 12 reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, which sparked nationwide controversy over the safety of the Ad26.COV2.S vaccine.14 After further investigation into those reports, the US Food and Drug Administration and the Centers for Disease Control and Prevention concluded that the benefits of the Ad26.COV2.S vaccine outweigh the low risk for associated thrombosis.15

Although adverse reactions are rare, it is important that health care providers take proper safety measures before and while administering any COVID-19 vaccine. Patients should be screened for contraindications to the COVID-19 vaccine to mitigate adverse effects seen in the small percentage of patients who may need to take alternative precautions.

The broad tissue tropism and high transmissibility of SARS-CoV-2 are the main contributors to its infection having reached pandemic scale. The spike (S) protein on SARS-CoV-2 binds to ACE2, the most thoroughly studied SARS-CoV-2 receptor, which is found in a range of tissues, including arterial endothelial cells, leading to its transfection. Several studies have proposed that expression of the S protein causes endothelial dysfunction through cytokine release, activation of complement, and ultimately microvascular occlusion.16

Recent developments in the use of viral-like particles, such as vesicular stomatitis virus, may mitigate future cases of RAE that are associated with endothelial cell transfection. Vesicular stomatitis virus is a popular model virus for research applications due to its glycoprotein and matrix protein contributing to its broad tropism. Recent efforts to alter these proteins have successfully limited the broad tropism of vesicular stomatitis virus.17

The SARS-CoV-2 virus must be handled in a Biosafety Level 3 laboratory. Conversely, pseudoviruses can be handled in lower containment facilities due to their safe and efficacious nature, offering an avenue to expedite vaccine development against many viral outbreaks, including SARS-CoV-2.18

 

 

An increasing number of cutaneous manifestations have been associated with COVID-19 infection and vaccination. Eruptive pseudoangiomatosis, a rare self-limiting exanthem, has been reported in association with ­COVID-19 vaccination.19 Eruptive pseudoangiomatosis manifests as erythematous blanchable papules that resemble angiomas, typically in a widespread distribution. Eruptive pseudoangiomatosis has striking similarities to RAE histologically; both manifest as dilated dermal blood vessels with plump endothelial cells.

Our case is unique because of the vasculitic palpable nature of the lesions, which were localized to the left arm. Eruptive pseudoangiomatosis formation after COVID-19 infection or SARS-CoV-2 vaccination may suggest alteration of ACE2 by binding of S protein.20 Such alteration of the ACE2 pathway would lead to inflammation of angiotensin II, causing proliferation of endothelial cells in the formation of angiomalike lesions. This hypothesis suggests a paraviral eruption secondary to an immunologic reaction, not a classical virtual eruption from direct contact of the virus on blood vessels. Although EPA and RAE are harmless and self-limiting, these reports will spread awareness of the increasing number of skin manifestations related to COVID-19 and SARS-CoV-2 virus vaccination.

Acknowledgment—Thoughtful insights and comments on this manuscript were provided by Christine J. Ko, MD (New Haven, Connecticut); Christine L. Egan, MD (Glen Mills, Pennsylvania); Howard A. Bueller, MD (Delray Beach, Florida); and Juan Pablo Robles, PhD (Juriquilla, Mexico).

References
  1. McMenamin ME, Fletcher CDM. Reactive angioendotheliomatosis: a study of 15 cases demonstrating a wide clinicopathologic spectrum. Am J Surg Pathol. 2002;26:686-697. doi:10.1097/00000478-200206000-00001
  2. Khan S, Pujani M, Jetley S, et al. Angiomatosis: a rare vascular proliferation of head and neck region. J Cutan Aesthet Surg. 2015;8:108-110. doi:10.4103/0974-2077.158448
  3. Gottron HA, Nikolowski W. Extrarenal Lohlein focal nephritis of the skin in endocarditis. Arch Klin Exp Dermatol. 1958;207:156-176.
  4. Cooper PH. Angioendotheliomatosis: two separate diseases. J Cutan Pathol. 1988;15:259. doi:10.1111/j.1600-0560.1988.tb00556.x
  5. Cancian L, Hansen A, Boshoff C. Cellular origin of Kaposi’s sarcoma and Kaposi’s sarcoma-associated herpesvirus-induced cell reprogramming. Trends Cell Biol. Sep 2013;23:421-32. doi:10.1016/j.tcb.2013.04.001
  6. Russell Jones R, Orchard G, Zelger B, et al. Immunostaining for CD31 and CD34 in Kaposi sarcoma. J Clin Pathol. 1995;48:1011-1016. doi:10.1136/jcp.48.11.1011
  7. Kahn HJ, Bailey D, Marks A. Monoclonal antibody D2-40, a new marker of lymphatic endothelium, reacts with Kaposi’s sarcoma and a subset of angiosarcomas. Mod Pathol. 2002;15:434-440. doi:10.1038/modpathol.3880543
  8. Genedy RM, Hamza AM, Abdel Latef AA, et al. Sensitivity and specificity of D2-40 in differentiating Kaposi sarcoma from its mimickers. J Egyptian Womens Dermatolog Soc. 2021;18:67-74. doi:10.4103/jewd.jewd_61_20
  9. Mesri EA, Cesarman E, Boshoff C. Kaposi’s sarcoma and its associated herpesvirus. Nat Rev Cancer. 2010;10:707-719. doi:10.1038/nrc2888
  10. Patel RM, Goldblum JR, Hsi ED. Immunohistochemical detection of human herpes virus-8 latent nuclear antigen-1 is useful in the diagnosis of Kaposi sarcoma. Mod Pathol. 2004;17:456-460. doi:10.1038/modpathol.3800061
  11. Zuckerman JN. The importance of injecting vaccines into muscle. Different patients need different needle sizes. BMJ. 2000;321:1237-1238. doi:10.1136/bmj.321.7271.1237
  12. Bhatia R, Hazarika N, Chandrasekaran D, et al. Treatment of posttraumatic reactive angioendotheliomatosis with topical timolol maleate. JAMA Dermatol. 2021;157:1002-1004. doi:10.1001/jamadermatol.2021.1770
  13. Sadoff J, Gray G, Vandebosch A, et al; ENSEMBLE Study Group. Safety and efficacy of single-dose Ad26.COV2.S vaccine against Covid-19. N Engl J Med. 2021;384:2187-2201. doi:10.1056/NEJMoa2101544
  14. See I, Su JR, Lale A, et al. US case reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, March 2 to April 21, 2021. JAMA. 2021;325:2448-2456. doi:10.1001/jama.2021.7517
  15. Berry CT, Eliliwi M, Gallagher S, et al. Cutaneous small vessel vasculitis following single-dose Janssen Ad26.COV2.S vaccination. JAAD Case Rep. 2021;15:11-14. doi:10.1016/j.jdcr.2021.07.002
  16. Flaumenhaft R, Enjyoji K, Schmaier AA. Vasculopathy in COVID-19. Blood. 2022;140:222-235. doi:10.1182/blood.2021012250
  17. Hastie E, Cataldi M, Marriott I, et al. Understanding and altering cell tropism of vesicular stomatitis virus. Virus Res. 2013;176:16-32. doi:10.1016/j.virusres.2013.06.003
  18. Xiong H-L, Wu Y-T, Cao J-L, et al. Robust neutralization assay based on SARS-CoV-2 S-protein-bearing vesicular stomatitis virus (VSV) pseudovirus and ACE2-overexpressing BHK21 cells. Emerg Microbes Infect. 2020;9:2105-2113. doi:10.1080/22221751.2020.1815589
  19. Mohta A, Jain SK, Mehta RD, et al. Development of eruptive pseudoangiomatosis following COVID-19 immunization – apropos of 5 cases. J Eur Acad Dermatol Venereol. 2021;35:e722-e725. doi:10.1111/jdv.17499
  20. Angeli F, Spanevello A, Reboldi G, et al. SARS-CoV-2 vaccines: lights and shadows. Eur J Intern Med. 2021;88:1-8. doi:10.1016/j.ejim.2021.04.019
References
  1. McMenamin ME, Fletcher CDM. Reactive angioendotheliomatosis: a study of 15 cases demonstrating a wide clinicopathologic spectrum. Am J Surg Pathol. 2002;26:686-697. doi:10.1097/00000478-200206000-00001
  2. Khan S, Pujani M, Jetley S, et al. Angiomatosis: a rare vascular proliferation of head and neck region. J Cutan Aesthet Surg. 2015;8:108-110. doi:10.4103/0974-2077.158448
  3. Gottron HA, Nikolowski W. Extrarenal Lohlein focal nephritis of the skin in endocarditis. Arch Klin Exp Dermatol. 1958;207:156-176.
  4. Cooper PH. Angioendotheliomatosis: two separate diseases. J Cutan Pathol. 1988;15:259. doi:10.1111/j.1600-0560.1988.tb00556.x
  5. Cancian L, Hansen A, Boshoff C. Cellular origin of Kaposi’s sarcoma and Kaposi’s sarcoma-associated herpesvirus-induced cell reprogramming. Trends Cell Biol. Sep 2013;23:421-32. doi:10.1016/j.tcb.2013.04.001
  6. Russell Jones R, Orchard G, Zelger B, et al. Immunostaining for CD31 and CD34 in Kaposi sarcoma. J Clin Pathol. 1995;48:1011-1016. doi:10.1136/jcp.48.11.1011
  7. Kahn HJ, Bailey D, Marks A. Monoclonal antibody D2-40, a new marker of lymphatic endothelium, reacts with Kaposi’s sarcoma and a subset of angiosarcomas. Mod Pathol. 2002;15:434-440. doi:10.1038/modpathol.3880543
  8. Genedy RM, Hamza AM, Abdel Latef AA, et al. Sensitivity and specificity of D2-40 in differentiating Kaposi sarcoma from its mimickers. J Egyptian Womens Dermatolog Soc. 2021;18:67-74. doi:10.4103/jewd.jewd_61_20
  9. Mesri EA, Cesarman E, Boshoff C. Kaposi’s sarcoma and its associated herpesvirus. Nat Rev Cancer. 2010;10:707-719. doi:10.1038/nrc2888
  10. Patel RM, Goldblum JR, Hsi ED. Immunohistochemical detection of human herpes virus-8 latent nuclear antigen-1 is useful in the diagnosis of Kaposi sarcoma. Mod Pathol. 2004;17:456-460. doi:10.1038/modpathol.3800061
  11. Zuckerman JN. The importance of injecting vaccines into muscle. Different patients need different needle sizes. BMJ. 2000;321:1237-1238. doi:10.1136/bmj.321.7271.1237
  12. Bhatia R, Hazarika N, Chandrasekaran D, et al. Treatment of posttraumatic reactive angioendotheliomatosis with topical timolol maleate. JAMA Dermatol. 2021;157:1002-1004. doi:10.1001/jamadermatol.2021.1770
  13. Sadoff J, Gray G, Vandebosch A, et al; ENSEMBLE Study Group. Safety and efficacy of single-dose Ad26.COV2.S vaccine against Covid-19. N Engl J Med. 2021;384:2187-2201. doi:10.1056/NEJMoa2101544
  14. See I, Su JR, Lale A, et al. US case reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, March 2 to April 21, 2021. JAMA. 2021;325:2448-2456. doi:10.1001/jama.2021.7517
  15. Berry CT, Eliliwi M, Gallagher S, et al. Cutaneous small vessel vasculitis following single-dose Janssen Ad26.COV2.S vaccination. JAAD Case Rep. 2021;15:11-14. doi:10.1016/j.jdcr.2021.07.002
  16. Flaumenhaft R, Enjyoji K, Schmaier AA. Vasculopathy in COVID-19. Blood. 2022;140:222-235. doi:10.1182/blood.2021012250
  17. Hastie E, Cataldi M, Marriott I, et al. Understanding and altering cell tropism of vesicular stomatitis virus. Virus Res. 2013;176:16-32. doi:10.1016/j.virusres.2013.06.003
  18. Xiong H-L, Wu Y-T, Cao J-L, et al. Robust neutralization assay based on SARS-CoV-2 S-protein-bearing vesicular stomatitis virus (VSV) pseudovirus and ACE2-overexpressing BHK21 cells. Emerg Microbes Infect. 2020;9:2105-2113. doi:10.1080/22221751.2020.1815589
  19. Mohta A, Jain SK, Mehta RD, et al. Development of eruptive pseudoangiomatosis following COVID-19 immunization – apropos of 5 cases. J Eur Acad Dermatol Venereol. 2021;35:e722-e725. doi:10.1111/jdv.17499
  20. Angeli F, Spanevello A, Reboldi G, et al. SARS-CoV-2 vaccines: lights and shadows. Eur J Intern Med. 2021;88:1-8. doi:10.1016/j.ejim.2021.04.019
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E20-E23
Page Number
E20-E23
Publications
Publications
Topics
Article Type
Display Headline
Reactive Angioendotheliomatosis Following Ad26.COV2.S Vaccination
Display Headline
Reactive Angioendotheliomatosis Following Ad26.COV2.S Vaccination
Sections
Inside the Article

Practice points

  • Reactive angioendotheliomatosis (RAE) is a rare benign vascular proliferation of endothelial cells lining blood vessels that clinically appears similar to Kaposi sarcoma and must be differentiated by microscopic evaluation.
  • An increasing number of reports link SARS-CoV-2 viral infection or vaccination against this virus with various cutaneous manifestations. Our case offers a link between RAE and Ad26.COV2.S vaccination.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Bilateral Burning Palmoplantar Lesions

Article Type
Changed
Wed, 02/28/2024 - 07:30
Display Headline
Bilateral Burning Palmoplantar Lesions

The Diagnosis: Lichen Sclerosus

Histopathology revealed a thin epidermis with homogenization of the upper dermal collagen. By contrast, the lower dermis was sclerotic with patchy chronic dermal infiltrate (Figure). Ultimately, the patient’s clinical presentation and histopathologic findings led to a diagnosis of lichen sclerosus (LS).

Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).
A and B, Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).

Lichen sclerosus is a rare chronic inflammatory skin condition that typically is characterized by porcelainwhite atrophic plaques on the skin, most often involving the external female genitalia including the vulva and perianal area.1 It is thought to be underdiagnosed and underreported.2 Extragenital manifestations may occur, though some cases are characterized by concomitant genital involvement.3,4 Our patient presented with palmoplantar distribution of plaques without genitalia involvement. Approximately 6% to 10% of patients with extragenital LS do not have genital involvement at the time of diagnosis.3,5 Furthermore, LS involving the palms and soles is exceedingly rare.2 Although extragenital LS may be asymptomatic, patients can experience debilitating pruritus; bullae with hemorrhage and erosion; plaque thickening with repeated excoriations; and painful fissuring, especially if lesions are in areas that are susceptible to friction or tension.3,6 New lesions on previously unaffected skin also may develop secondary to trauma through the Koebner phenomenon.1,6

Histologically, LS is characterized by epidermal hyperkeratosis accompanied by follicular plugging, epidermal atrophy with flattened rete ridges, vacuolization of the basal epidermis, marked edema in the superficial dermis (in early lesions) or homogenized collagen in the upper dermis (in established lesions), and a lymphohistiocytic infiltrate beneath the homogenized collagen. Although the pathogenesis of LS is unclear, purported etiologic factors from studies in genital disease include immune dysfunction, genetic predisposition, infection, and trauma.6 Lichen sclerosus is associated strongly with autoimmune diseases including alopecia areata, vitiligo, autoimmune thyroiditis, diabetes mellitus, and pernicious anemia, indicating its potential multifactorial etiology and linkage to T-lymphocyte dysfunction.1 Early LS lesions often appear as flat-topped and slightly scaly, hypopigmented, white or mildly erythematous, polygonal papules that coalesce to form larger plaques with peripheral erythema. With time, the inflammation subsides, and lesions become porcelain-white with varying degrees of palpable sclerosis, resembling thin paperlike wrinkles indicative of epidermal atrophy.6

The differential diagnosis of LS includes lichen planus (LP), morphea, discoid lupus erythematosus (DLE), and vitiligo.3 Lesions of LP commonly are described as flat-topped, polygonal, pink-purple papules localized mostly along the volar wrists, shins, presacral area, and hands.7 Lichen planus is considered to be more pruritic3 than LS and can be further distinguished by biopsy through identifying a well-formed granular layer and numerous cytoid bodies. Unlike LS, LP is not characterized by basement membrane thickening or epidermal atrophy.8

Skin lesions seen in morphea may resemble the classic atrophic white lesions of extragenital LS; however, it is unclear if the appearance of LS-like lesions with morphea is a simultaneous occurrence of 2 separate disorders or the development of clinical findings resembling LS in lesions of morphea.6 Furthermore, morphea involves deep inflammation and sclerosis of the dermis that may extend into subcutaneous fat without follicular plugging of the epidermis.3,9 In contrast, LS primarily affects the epidermis and dermis with the presence of epidermal follicular plugging.6

Lesions seen in DLE are characterized as well-defined, annular, erythematous patches and plaques followed by follicular hyperkeratosis with adherent scaling. Upon removal of the scale, follicle-sized keratotic spikes (carpet tacks) are present.10 Scaling of lesions and the carpet tack sign were absent in our patient. In addition, DLE typically reveals surrounding pigmentation and scarring over plaques,3 which were not observed in our patient.

Vitiligo commonly is associated with extragenital LS. As with LS, vitiligo can be explained by mechanisms of immune checkpoint inhibitor–induced cytotoxicity as well as perforin and granzyme-B expression.11 Although vitiligo resembles the late hypopigmented lesions of extragenital LS, there are no plaques or surface changes, and a larger, more generalized area of the skin typically is involved.3

References
  1. Chamli A, Souissi A. Lichen sclerosus. StatPearls [Internet]. StatPearls Publishing; 2022. http://www.ncbi.nlm.nih.gov/books/NBK538246/
  2. Gaddis KJ, Huang J, Haun PL. An atrophic and spiny eruption of the palms. JAMA Dermatol. 2018;154:1344-1345. doi:10.1001 /jamadermatol.2018.1265
  3. Arif T, Fatima R, Sami M. Extragenital lichen sclerosus: a comprehensive review [published online August 11, 2022]. Australas J Dermatol. doi:10.1111/ajd.13890
  4. Heibel HD, Styles AR, Cockerell CJ. A case of acral lichen sclerosus et atrophicus. JAAD Case Rep. 2020;8:26-27. doi:10.1016/j.jdcr.2020.12.008
  5. Seyffert J, Bibliowicz N, Harding T, et al. Palmar lichen sclerosus et atrophicus. JAAD Case Rep. 2020;6:697-699. doi:10.1016/j.jdcr.2020.06.005
  6. Jacobe H. Extragenital lichen sclerosus: clinical features and diagnosis. UpToDate. Updated July 11, 2023. Accessed December 14, 2023. https://www.uptodate.com/contents/extragenital-lichen-sclerosus?search=Lichen%20sclerosus&source =search_result&selectedTitle=2~66&usage_type=default&display_ rank=2
  7. Goldstein BG, Goldstein AO, Mostow E. Lichen planus. UpToDate. Updated October 25, 2021. Accessed December 14, 2023. https://www.uptodate.com/contents/lichen-planus?search=lichen%20 sclerosus&topicRef=15838&source=see_link
  8. Tallon B. Lichen sclerosus pathology. DermNet NZ website. Accessed December 5, 2023. https://dermnetnz.org/topics/lichen-sclerosus-pathology
  9. Jacobe H. Pathogenesis, clinical manifestations, and diagnosis of morphea (localized scleroderma) in adults. UpToDate. Updated November 15, 2021. Accessed December 14, 2023. https://medilib.ir/uptodate/show/13776
  10. McDaniel B, Sukumaran S, Koritala T, et al. Discoid lupus erythematosus. StatPearls [Internet]. StatPearls Publishing; 2022. Updated August 28, 2023. Accessed December 14, 2023. http://www.ncbi.nlm.nih.gov/books/NBK493145/
  11. Veronesi G, Scarfì F, Misciali C, et al. An unusual skin reaction in uveal melanoma during treatment with nivolumab: extragenital lichen sclerosus. Anticancer Drugs. 2019;30:969-972. doi:10.1097/ CAD.0000000000000819
Article PDF
Author and Disclosure Information

From the University of Florida College of Medicine, Gainesville. Drs. Gurnani and Montañez-Wiscovich are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Marjorie E. Montañez-Wiscovich, MD, PhD, 4037 NW 86 Terr, 4th Floor, Gainesville, FL 32606 (m.montanez@dermatology.med.ufl.edu).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E24-E26
Sections
Author and Disclosure Information

From the University of Florida College of Medicine, Gainesville. Drs. Gurnani and Montañez-Wiscovich are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Marjorie E. Montañez-Wiscovich, MD, PhD, 4037 NW 86 Terr, 4th Floor, Gainesville, FL 32606 (m.montanez@dermatology.med.ufl.edu).

Author and Disclosure Information

From the University of Florida College of Medicine, Gainesville. Drs. Gurnani and Montañez-Wiscovich are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Marjorie E. Montañez-Wiscovich, MD, PhD, 4037 NW 86 Terr, 4th Floor, Gainesville, FL 32606 (m.montanez@dermatology.med.ufl.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Lichen Sclerosus

Histopathology revealed a thin epidermis with homogenization of the upper dermal collagen. By contrast, the lower dermis was sclerotic with patchy chronic dermal infiltrate (Figure). Ultimately, the patient’s clinical presentation and histopathologic findings led to a diagnosis of lichen sclerosus (LS).

Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).
A and B, Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).

Lichen sclerosus is a rare chronic inflammatory skin condition that typically is characterized by porcelainwhite atrophic plaques on the skin, most often involving the external female genitalia including the vulva and perianal area.1 It is thought to be underdiagnosed and underreported.2 Extragenital manifestations may occur, though some cases are characterized by concomitant genital involvement.3,4 Our patient presented with palmoplantar distribution of plaques without genitalia involvement. Approximately 6% to 10% of patients with extragenital LS do not have genital involvement at the time of diagnosis.3,5 Furthermore, LS involving the palms and soles is exceedingly rare.2 Although extragenital LS may be asymptomatic, patients can experience debilitating pruritus; bullae with hemorrhage and erosion; plaque thickening with repeated excoriations; and painful fissuring, especially if lesions are in areas that are susceptible to friction or tension.3,6 New lesions on previously unaffected skin also may develop secondary to trauma through the Koebner phenomenon.1,6

Histologically, LS is characterized by epidermal hyperkeratosis accompanied by follicular plugging, epidermal atrophy with flattened rete ridges, vacuolization of the basal epidermis, marked edema in the superficial dermis (in early lesions) or homogenized collagen in the upper dermis (in established lesions), and a lymphohistiocytic infiltrate beneath the homogenized collagen. Although the pathogenesis of LS is unclear, purported etiologic factors from studies in genital disease include immune dysfunction, genetic predisposition, infection, and trauma.6 Lichen sclerosus is associated strongly with autoimmune diseases including alopecia areata, vitiligo, autoimmune thyroiditis, diabetes mellitus, and pernicious anemia, indicating its potential multifactorial etiology and linkage to T-lymphocyte dysfunction.1 Early LS lesions often appear as flat-topped and slightly scaly, hypopigmented, white or mildly erythematous, polygonal papules that coalesce to form larger plaques with peripheral erythema. With time, the inflammation subsides, and lesions become porcelain-white with varying degrees of palpable sclerosis, resembling thin paperlike wrinkles indicative of epidermal atrophy.6

The differential diagnosis of LS includes lichen planus (LP), morphea, discoid lupus erythematosus (DLE), and vitiligo.3 Lesions of LP commonly are described as flat-topped, polygonal, pink-purple papules localized mostly along the volar wrists, shins, presacral area, and hands.7 Lichen planus is considered to be more pruritic3 than LS and can be further distinguished by biopsy through identifying a well-formed granular layer and numerous cytoid bodies. Unlike LS, LP is not characterized by basement membrane thickening or epidermal atrophy.8

Skin lesions seen in morphea may resemble the classic atrophic white lesions of extragenital LS; however, it is unclear if the appearance of LS-like lesions with morphea is a simultaneous occurrence of 2 separate disorders or the development of clinical findings resembling LS in lesions of morphea.6 Furthermore, morphea involves deep inflammation and sclerosis of the dermis that may extend into subcutaneous fat without follicular plugging of the epidermis.3,9 In contrast, LS primarily affects the epidermis and dermis with the presence of epidermal follicular plugging.6

Lesions seen in DLE are characterized as well-defined, annular, erythematous patches and plaques followed by follicular hyperkeratosis with adherent scaling. Upon removal of the scale, follicle-sized keratotic spikes (carpet tacks) are present.10 Scaling of lesions and the carpet tack sign were absent in our patient. In addition, DLE typically reveals surrounding pigmentation and scarring over plaques,3 which were not observed in our patient.

Vitiligo commonly is associated with extragenital LS. As with LS, vitiligo can be explained by mechanisms of immune checkpoint inhibitor–induced cytotoxicity as well as perforin and granzyme-B expression.11 Although vitiligo resembles the late hypopigmented lesions of extragenital LS, there are no plaques or surface changes, and a larger, more generalized area of the skin typically is involved.3

The Diagnosis: Lichen Sclerosus

Histopathology revealed a thin epidermis with homogenization of the upper dermal collagen. By contrast, the lower dermis was sclerotic with patchy chronic dermal infiltrate (Figure). Ultimately, the patient’s clinical presentation and histopathologic findings led to a diagnosis of lichen sclerosus (LS).

Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).
A and B, Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).

Lichen sclerosus is a rare chronic inflammatory skin condition that typically is characterized by porcelainwhite atrophic plaques on the skin, most often involving the external female genitalia including the vulva and perianal area.1 It is thought to be underdiagnosed and underreported.2 Extragenital manifestations may occur, though some cases are characterized by concomitant genital involvement.3,4 Our patient presented with palmoplantar distribution of plaques without genitalia involvement. Approximately 6% to 10% of patients with extragenital LS do not have genital involvement at the time of diagnosis.3,5 Furthermore, LS involving the palms and soles is exceedingly rare.2 Although extragenital LS may be asymptomatic, patients can experience debilitating pruritus; bullae with hemorrhage and erosion; plaque thickening with repeated excoriations; and painful fissuring, especially if lesions are in areas that are susceptible to friction or tension.3,6 New lesions on previously unaffected skin also may develop secondary to trauma through the Koebner phenomenon.1,6

Histologically, LS is characterized by epidermal hyperkeratosis accompanied by follicular plugging, epidermal atrophy with flattened rete ridges, vacuolization of the basal epidermis, marked edema in the superficial dermis (in early lesions) or homogenized collagen in the upper dermis (in established lesions), and a lymphohistiocytic infiltrate beneath the homogenized collagen. Although the pathogenesis of LS is unclear, purported etiologic factors from studies in genital disease include immune dysfunction, genetic predisposition, infection, and trauma.6 Lichen sclerosus is associated strongly with autoimmune diseases including alopecia areata, vitiligo, autoimmune thyroiditis, diabetes mellitus, and pernicious anemia, indicating its potential multifactorial etiology and linkage to T-lymphocyte dysfunction.1 Early LS lesions often appear as flat-topped and slightly scaly, hypopigmented, white or mildly erythematous, polygonal papules that coalesce to form larger plaques with peripheral erythema. With time, the inflammation subsides, and lesions become porcelain-white with varying degrees of palpable sclerosis, resembling thin paperlike wrinkles indicative of epidermal atrophy.6

The differential diagnosis of LS includes lichen planus (LP), morphea, discoid lupus erythematosus (DLE), and vitiligo.3 Lesions of LP commonly are described as flat-topped, polygonal, pink-purple papules localized mostly along the volar wrists, shins, presacral area, and hands.7 Lichen planus is considered to be more pruritic3 than LS and can be further distinguished by biopsy through identifying a well-formed granular layer and numerous cytoid bodies. Unlike LS, LP is not characterized by basement membrane thickening or epidermal atrophy.8

Skin lesions seen in morphea may resemble the classic atrophic white lesions of extragenital LS; however, it is unclear if the appearance of LS-like lesions with morphea is a simultaneous occurrence of 2 separate disorders or the development of clinical findings resembling LS in lesions of morphea.6 Furthermore, morphea involves deep inflammation and sclerosis of the dermis that may extend into subcutaneous fat without follicular plugging of the epidermis.3,9 In contrast, LS primarily affects the epidermis and dermis with the presence of epidermal follicular plugging.6

Lesions seen in DLE are characterized as well-defined, annular, erythematous patches and plaques followed by follicular hyperkeratosis with adherent scaling. Upon removal of the scale, follicle-sized keratotic spikes (carpet tacks) are present.10 Scaling of lesions and the carpet tack sign were absent in our patient. In addition, DLE typically reveals surrounding pigmentation and scarring over plaques,3 which were not observed in our patient.

Vitiligo commonly is associated with extragenital LS. As with LS, vitiligo can be explained by mechanisms of immune checkpoint inhibitor–induced cytotoxicity as well as perforin and granzyme-B expression.11 Although vitiligo resembles the late hypopigmented lesions of extragenital LS, there are no plaques or surface changes, and a larger, more generalized area of the skin typically is involved.3

References
  1. Chamli A, Souissi A. Lichen sclerosus. StatPearls [Internet]. StatPearls Publishing; 2022. http://www.ncbi.nlm.nih.gov/books/NBK538246/
  2. Gaddis KJ, Huang J, Haun PL. An atrophic and spiny eruption of the palms. JAMA Dermatol. 2018;154:1344-1345. doi:10.1001 /jamadermatol.2018.1265
  3. Arif T, Fatima R, Sami M. Extragenital lichen sclerosus: a comprehensive review [published online August 11, 2022]. Australas J Dermatol. doi:10.1111/ajd.13890
  4. Heibel HD, Styles AR, Cockerell CJ. A case of acral lichen sclerosus et atrophicus. JAAD Case Rep. 2020;8:26-27. doi:10.1016/j.jdcr.2020.12.008
  5. Seyffert J, Bibliowicz N, Harding T, et al. Palmar lichen sclerosus et atrophicus. JAAD Case Rep. 2020;6:697-699. doi:10.1016/j.jdcr.2020.06.005
  6. Jacobe H. Extragenital lichen sclerosus: clinical features and diagnosis. UpToDate. Updated July 11, 2023. Accessed December 14, 2023. https://www.uptodate.com/contents/extragenital-lichen-sclerosus?search=Lichen%20sclerosus&source =search_result&selectedTitle=2~66&usage_type=default&display_ rank=2
  7. Goldstein BG, Goldstein AO, Mostow E. Lichen planus. UpToDate. Updated October 25, 2021. Accessed December 14, 2023. https://www.uptodate.com/contents/lichen-planus?search=lichen%20 sclerosus&topicRef=15838&source=see_link
  8. Tallon B. Lichen sclerosus pathology. DermNet NZ website. Accessed December 5, 2023. https://dermnetnz.org/topics/lichen-sclerosus-pathology
  9. Jacobe H. Pathogenesis, clinical manifestations, and diagnosis of morphea (localized scleroderma) in adults. UpToDate. Updated November 15, 2021. Accessed December 14, 2023. https://medilib.ir/uptodate/show/13776
  10. McDaniel B, Sukumaran S, Koritala T, et al. Discoid lupus erythematosus. StatPearls [Internet]. StatPearls Publishing; 2022. Updated August 28, 2023. Accessed December 14, 2023. http://www.ncbi.nlm.nih.gov/books/NBK493145/
  11. Veronesi G, Scarfì F, Misciali C, et al. An unusual skin reaction in uveal melanoma during treatment with nivolumab: extragenital lichen sclerosus. Anticancer Drugs. 2019;30:969-972. doi:10.1097/ CAD.0000000000000819
References
  1. Chamli A, Souissi A. Lichen sclerosus. StatPearls [Internet]. StatPearls Publishing; 2022. http://www.ncbi.nlm.nih.gov/books/NBK538246/
  2. Gaddis KJ, Huang J, Haun PL. An atrophic and spiny eruption of the palms. JAMA Dermatol. 2018;154:1344-1345. doi:10.1001 /jamadermatol.2018.1265
  3. Arif T, Fatima R, Sami M. Extragenital lichen sclerosus: a comprehensive review [published online August 11, 2022]. Australas J Dermatol. doi:10.1111/ajd.13890
  4. Heibel HD, Styles AR, Cockerell CJ. A case of acral lichen sclerosus et atrophicus. JAAD Case Rep. 2020;8:26-27. doi:10.1016/j.jdcr.2020.12.008
  5. Seyffert J, Bibliowicz N, Harding T, et al. Palmar lichen sclerosus et atrophicus. JAAD Case Rep. 2020;6:697-699. doi:10.1016/j.jdcr.2020.06.005
  6. Jacobe H. Extragenital lichen sclerosus: clinical features and diagnosis. UpToDate. Updated July 11, 2023. Accessed December 14, 2023. https://www.uptodate.com/contents/extragenital-lichen-sclerosus?search=Lichen%20sclerosus&source =search_result&selectedTitle=2~66&usage_type=default&display_ rank=2
  7. Goldstein BG, Goldstein AO, Mostow E. Lichen planus. UpToDate. Updated October 25, 2021. Accessed December 14, 2023. https://www.uptodate.com/contents/lichen-planus?search=lichen%20 sclerosus&topicRef=15838&source=see_link
  8. Tallon B. Lichen sclerosus pathology. DermNet NZ website. Accessed December 5, 2023. https://dermnetnz.org/topics/lichen-sclerosus-pathology
  9. Jacobe H. Pathogenesis, clinical manifestations, and diagnosis of morphea (localized scleroderma) in adults. UpToDate. Updated November 15, 2021. Accessed December 14, 2023. https://medilib.ir/uptodate/show/13776
  10. McDaniel B, Sukumaran S, Koritala T, et al. Discoid lupus erythematosus. StatPearls [Internet]. StatPearls Publishing; 2022. Updated August 28, 2023. Accessed December 14, 2023. http://www.ncbi.nlm.nih.gov/books/NBK493145/
  11. Veronesi G, Scarfì F, Misciali C, et al. An unusual skin reaction in uveal melanoma during treatment with nivolumab: extragenital lichen sclerosus. Anticancer Drugs. 2019;30:969-972. doi:10.1097/ CAD.0000000000000819
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E24-E26
Page Number
E24-E26
Publications
Publications
Topics
Article Type
Display Headline
Bilateral Burning Palmoplantar Lesions
Display Headline
Bilateral Burning Palmoplantar Lesions
Sections
Questionnaire Body

A 59-year-old woman presented with atrophic, hypopigmented, ivory papules and plaques localized to the central palms and soles of 3 years’ duration. The lesions were associated with burning that was most notable after extended periods of ambulation. The lesions initially were diagnosed as plaque psoriasis by an external dermatology clinic. At the time of presentation to our clinic, treatment with several highpotency topical steroids and biologics approved for plaque psoriasis had failed. Her medical history and concurrent medical workup were notable for type 2 diabetes mellitus, liver dysfunction, thyroid nodules overseen by an endocrinologist, vitamin B12 and vitamin D deficiencies managed with supplementation, and diffuse androgenic alopecia with suspected telogen effluvium. Physical examination revealed no plaque fissuring, pruritus, or scaling. She had no history of radiation therapy or organ transplantation. A punch biopsy of the left palm was performed.

Bilateral burning palmoplantar lesions

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 12/19/2023 - 11:45
Un-Gate On Date
Tue, 12/19/2023 - 11:45
Use ProPublica
CFC Schedule Remove Status
Tue, 12/19/2023 - 11:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Diffuse Capillary Malformation With Undergrowth of a Limb in a Boy

Article Type
Changed
Mon, 12/18/2023 - 08:19
Display Headline
Diffuse Capillary Malformation With Undergrowth of a Limb in a Boy

To the Editor:

Capillary malformations (CMs), the most common vascular malformations that can affect the skin,1 present clinically as macules and patches of various colors, shapes, and sizes. Congenital structural abnormalities are associated with conditions such as Klippel-Trenaunay syndrome (KTS), cutis marmorata telangiectatica congenita (CMTC), and megalencephaly–capillary malformation syndrome.2 Diffuse CM with overgrowth (DCMO) of the soft tissue and bones is an established association of CMs; however, diffuse capillary malformation with undergrowth (DCMU) is a more recent term that describes the lesser-recognized counterpart to DCMO.3 Herein, we describe a case of CM with left-sided undergrowth.

Reticulated violaceous patches on the left abdomen and left anterior thigh, respectively.
FIGURE 1. A and B, Reticulated violaceous patches on the left abdomen and left anterior thigh, respectively.

An 11-year-old boy presented to our clinic with asymptomatic vascular patterning on the left side of the body that had been present since birth. He previously was diagnosed with congenital right hemihypertrophy. He reported that the areas gradually lightened over time, and he denied any history of ulceration or venous or lymphatic malformations. Additionally, he explained how the left arm and leg have been noticeably smaller than the right extremities throughout his life. Physical examination revealed superficial, violaceous, reticulated patches along the left upper back tracking down the arm, abdomen (Figure 1A), and anterior thigh (Figure 1B) without crossing the midline. A few dilated veins were noted in the same region as the patches. There was no evidence of scarring or depression found in the skin. The right arms and legs were visibly larger compared to the left side (Figure 2A), and there also was macrodactyly of the third digit of the left hand (Figure 2B). Radiography confirmed the limb length discrepancy and showed the right and left legs to measure 73.2 cm and 71.3 cm, respectively. Given the patient’s multifocal reticulated CMs and ipsilateral undergrowth, a diagnosis of DCMU was rendered. The superficial vascular pattern is likely to fade over time, which will partially be hidden by his darker complexion. He also was advised to continue to see an orthopedist to monitor the limb length incongruity. Surgical intervention was not recommended.

Hypotrophy of the left arm and hand as well as macrodactyly of the left third digit, respectively.
FIGURE 2. A and B, Hypotrophy of the left arm and hand as well as macrodactyly of the left third digit, respectively.

It ordinarily is thought that vascular anomalies of a limb may result in hypertrophy due to increased blood flow such as in KTS, but there are occasions where the affected limb(s) are inexplicably smaller.2,4 Cubiró et al3 observed that in 6 patients with unilateral CMs, all had ipsilateral limb undergrowth. They proposed the term diffuse capillary malformation with undergrowth as a distinct counterpart to DCMO. Diffuse capillary malformation with undergrowth is most similar to CMTC, as both can present with patchy or reticulated capillary staining with ipsilateral limb hypotrophy, but girth more often is affected than length; CMTC also may be associated with dermal atrophy and ulceration.2 The lesions of CMTC typically diminish within the first few years of life whereas those in DCMU tend to persist. Patients with KTS also can exhibit soft-tissue and bony undergrowth, which is termed inverse Klippel-Trenaunay syndrome3; however, the lack of the triad of capillary-lymphatic-venous malformation in our patient made this condition less likely. Additionally, it appears that our patient had left-sided undergrowth rather than the previously diagnosed right hemihypertrophy. The ipsilateral macrodactyly of the third digit of the left hand was an interesting observation and contrasted the undergrowth apparent in the rest of the left limb, which could be caused by increased blood flow specifically to the third digit resembling DCMO.4

Of note, genetic mutations have been implicated as a cause of vascular malformations and growth abnormalities. Specifically, mutations in the phosphoinositide-3-kinase–AKT pathway have been reported in these cases likely due its role in cell growth, proliferation, and angiogenesis.3,4 Future studies should investigate genetic associations in patients with DCMU to determine if there is a robust genotypic-phenotypic link.

Although CMs are a common occurrence in pediatric dermatology, CMs with concurrent limb undergrowth are rare. Our patient’s unique features included involvement of both an arm and leg as well as the presence of macrodactyly. We agree with the terminology for DCMU to describe multifocal reticulated vascular patterning with ipsilateral undergrowth.3

References
  1. Huang JT, Liang MG. Vascular malformations. Pediatr Clin North Am. 2010;57:1091-1110. doi:10.1016/j.pcl.2010.08.003
  2. Lee MS, Liang MG, Mulliken JB. Diffuse capillary malformation with overgrowth: a clinical subtype of vascular anomalies with hypertrophy. J Am Acad Dermatol. 2013;69:589-594. doi:10.1016/j.jaad.2013.05.030
  3. Cubiró X, Rozas‐Muñoz E, Castel P, et al. Clinical and genetic evaluation of six children with diffuse capillary malformation and undergrowth. Pediatr Dermatol. 2020;37:833-838. doi:10.1111/pde.14252
  4. Uihlein LC, Liang MG, Fishman SJ, et al. Capillary-venous malformation in the lower limb. Pediatr Dermatol. 2013;30:541-548. doi:10.1111/pde.12186
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Baylor Scott & White, Temple, Texas.

The authors report no conflict of interest.

Correspondence: Ronnie M. Youssef, MD, Texas A&M College of Medicine, 3500 Gaston Ave, Dallas, TX 75246 (ryoussef14@exchange.tamu.edu).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E17-E19
Sections
Author and Disclosure Information

From the Department of Dermatology, Baylor Scott & White, Temple, Texas.

The authors report no conflict of interest.

Correspondence: Ronnie M. Youssef, MD, Texas A&M College of Medicine, 3500 Gaston Ave, Dallas, TX 75246 (ryoussef14@exchange.tamu.edu).

Author and Disclosure Information

From the Department of Dermatology, Baylor Scott & White, Temple, Texas.

The authors report no conflict of interest.

Correspondence: Ronnie M. Youssef, MD, Texas A&M College of Medicine, 3500 Gaston Ave, Dallas, TX 75246 (ryoussef14@exchange.tamu.edu).

Article PDF
Article PDF

To the Editor:

Capillary malformations (CMs), the most common vascular malformations that can affect the skin,1 present clinically as macules and patches of various colors, shapes, and sizes. Congenital structural abnormalities are associated with conditions such as Klippel-Trenaunay syndrome (KTS), cutis marmorata telangiectatica congenita (CMTC), and megalencephaly–capillary malformation syndrome.2 Diffuse CM with overgrowth (DCMO) of the soft tissue and bones is an established association of CMs; however, diffuse capillary malformation with undergrowth (DCMU) is a more recent term that describes the lesser-recognized counterpart to DCMO.3 Herein, we describe a case of CM with left-sided undergrowth.

Reticulated violaceous patches on the left abdomen and left anterior thigh, respectively.
FIGURE 1. A and B, Reticulated violaceous patches on the left abdomen and left anterior thigh, respectively.

An 11-year-old boy presented to our clinic with asymptomatic vascular patterning on the left side of the body that had been present since birth. He previously was diagnosed with congenital right hemihypertrophy. He reported that the areas gradually lightened over time, and he denied any history of ulceration or venous or lymphatic malformations. Additionally, he explained how the left arm and leg have been noticeably smaller than the right extremities throughout his life. Physical examination revealed superficial, violaceous, reticulated patches along the left upper back tracking down the arm, abdomen (Figure 1A), and anterior thigh (Figure 1B) without crossing the midline. A few dilated veins were noted in the same region as the patches. There was no evidence of scarring or depression found in the skin. The right arms and legs were visibly larger compared to the left side (Figure 2A), and there also was macrodactyly of the third digit of the left hand (Figure 2B). Radiography confirmed the limb length discrepancy and showed the right and left legs to measure 73.2 cm and 71.3 cm, respectively. Given the patient’s multifocal reticulated CMs and ipsilateral undergrowth, a diagnosis of DCMU was rendered. The superficial vascular pattern is likely to fade over time, which will partially be hidden by his darker complexion. He also was advised to continue to see an orthopedist to monitor the limb length incongruity. Surgical intervention was not recommended.

Hypotrophy of the left arm and hand as well as macrodactyly of the left third digit, respectively.
FIGURE 2. A and B, Hypotrophy of the left arm and hand as well as macrodactyly of the left third digit, respectively.

It ordinarily is thought that vascular anomalies of a limb may result in hypertrophy due to increased blood flow such as in KTS, but there are occasions where the affected limb(s) are inexplicably smaller.2,4 Cubiró et al3 observed that in 6 patients with unilateral CMs, all had ipsilateral limb undergrowth. They proposed the term diffuse capillary malformation with undergrowth as a distinct counterpart to DCMO. Diffuse capillary malformation with undergrowth is most similar to CMTC, as both can present with patchy or reticulated capillary staining with ipsilateral limb hypotrophy, but girth more often is affected than length; CMTC also may be associated with dermal atrophy and ulceration.2 The lesions of CMTC typically diminish within the first few years of life whereas those in DCMU tend to persist. Patients with KTS also can exhibit soft-tissue and bony undergrowth, which is termed inverse Klippel-Trenaunay syndrome3; however, the lack of the triad of capillary-lymphatic-venous malformation in our patient made this condition less likely. Additionally, it appears that our patient had left-sided undergrowth rather than the previously diagnosed right hemihypertrophy. The ipsilateral macrodactyly of the third digit of the left hand was an interesting observation and contrasted the undergrowth apparent in the rest of the left limb, which could be caused by increased blood flow specifically to the third digit resembling DCMO.4

Of note, genetic mutations have been implicated as a cause of vascular malformations and growth abnormalities. Specifically, mutations in the phosphoinositide-3-kinase–AKT pathway have been reported in these cases likely due its role in cell growth, proliferation, and angiogenesis.3,4 Future studies should investigate genetic associations in patients with DCMU to determine if there is a robust genotypic-phenotypic link.

Although CMs are a common occurrence in pediatric dermatology, CMs with concurrent limb undergrowth are rare. Our patient’s unique features included involvement of both an arm and leg as well as the presence of macrodactyly. We agree with the terminology for DCMU to describe multifocal reticulated vascular patterning with ipsilateral undergrowth.3

To the Editor:

Capillary malformations (CMs), the most common vascular malformations that can affect the skin,1 present clinically as macules and patches of various colors, shapes, and sizes. Congenital structural abnormalities are associated with conditions such as Klippel-Trenaunay syndrome (KTS), cutis marmorata telangiectatica congenita (CMTC), and megalencephaly–capillary malformation syndrome.2 Diffuse CM with overgrowth (DCMO) of the soft tissue and bones is an established association of CMs; however, diffuse capillary malformation with undergrowth (DCMU) is a more recent term that describes the lesser-recognized counterpart to DCMO.3 Herein, we describe a case of CM with left-sided undergrowth.

Reticulated violaceous patches on the left abdomen and left anterior thigh, respectively.
FIGURE 1. A and B, Reticulated violaceous patches on the left abdomen and left anterior thigh, respectively.

An 11-year-old boy presented to our clinic with asymptomatic vascular patterning on the left side of the body that had been present since birth. He previously was diagnosed with congenital right hemihypertrophy. He reported that the areas gradually lightened over time, and he denied any history of ulceration or venous or lymphatic malformations. Additionally, he explained how the left arm and leg have been noticeably smaller than the right extremities throughout his life. Physical examination revealed superficial, violaceous, reticulated patches along the left upper back tracking down the arm, abdomen (Figure 1A), and anterior thigh (Figure 1B) without crossing the midline. A few dilated veins were noted in the same region as the patches. There was no evidence of scarring or depression found in the skin. The right arms and legs were visibly larger compared to the left side (Figure 2A), and there also was macrodactyly of the third digit of the left hand (Figure 2B). Radiography confirmed the limb length discrepancy and showed the right and left legs to measure 73.2 cm and 71.3 cm, respectively. Given the patient’s multifocal reticulated CMs and ipsilateral undergrowth, a diagnosis of DCMU was rendered. The superficial vascular pattern is likely to fade over time, which will partially be hidden by his darker complexion. He also was advised to continue to see an orthopedist to monitor the limb length incongruity. Surgical intervention was not recommended.

Hypotrophy of the left arm and hand as well as macrodactyly of the left third digit, respectively.
FIGURE 2. A and B, Hypotrophy of the left arm and hand as well as macrodactyly of the left third digit, respectively.

It ordinarily is thought that vascular anomalies of a limb may result in hypertrophy due to increased blood flow such as in KTS, but there are occasions where the affected limb(s) are inexplicably smaller.2,4 Cubiró et al3 observed that in 6 patients with unilateral CMs, all had ipsilateral limb undergrowth. They proposed the term diffuse capillary malformation with undergrowth as a distinct counterpart to DCMO. Diffuse capillary malformation with undergrowth is most similar to CMTC, as both can present with patchy or reticulated capillary staining with ipsilateral limb hypotrophy, but girth more often is affected than length; CMTC also may be associated with dermal atrophy and ulceration.2 The lesions of CMTC typically diminish within the first few years of life whereas those in DCMU tend to persist. Patients with KTS also can exhibit soft-tissue and bony undergrowth, which is termed inverse Klippel-Trenaunay syndrome3; however, the lack of the triad of capillary-lymphatic-venous malformation in our patient made this condition less likely. Additionally, it appears that our patient had left-sided undergrowth rather than the previously diagnosed right hemihypertrophy. The ipsilateral macrodactyly of the third digit of the left hand was an interesting observation and contrasted the undergrowth apparent in the rest of the left limb, which could be caused by increased blood flow specifically to the third digit resembling DCMO.4

Of note, genetic mutations have been implicated as a cause of vascular malformations and growth abnormalities. Specifically, mutations in the phosphoinositide-3-kinase–AKT pathway have been reported in these cases likely due its role in cell growth, proliferation, and angiogenesis.3,4 Future studies should investigate genetic associations in patients with DCMU to determine if there is a robust genotypic-phenotypic link.

Although CMs are a common occurrence in pediatric dermatology, CMs with concurrent limb undergrowth are rare. Our patient’s unique features included involvement of both an arm and leg as well as the presence of macrodactyly. We agree with the terminology for DCMU to describe multifocal reticulated vascular patterning with ipsilateral undergrowth.3

References
  1. Huang JT, Liang MG. Vascular malformations. Pediatr Clin North Am. 2010;57:1091-1110. doi:10.1016/j.pcl.2010.08.003
  2. Lee MS, Liang MG, Mulliken JB. Diffuse capillary malformation with overgrowth: a clinical subtype of vascular anomalies with hypertrophy. J Am Acad Dermatol. 2013;69:589-594. doi:10.1016/j.jaad.2013.05.030
  3. Cubiró X, Rozas‐Muñoz E, Castel P, et al. Clinical and genetic evaluation of six children with diffuse capillary malformation and undergrowth. Pediatr Dermatol. 2020;37:833-838. doi:10.1111/pde.14252
  4. Uihlein LC, Liang MG, Fishman SJ, et al. Capillary-venous malformation in the lower limb. Pediatr Dermatol. 2013;30:541-548. doi:10.1111/pde.12186
References
  1. Huang JT, Liang MG. Vascular malformations. Pediatr Clin North Am. 2010;57:1091-1110. doi:10.1016/j.pcl.2010.08.003
  2. Lee MS, Liang MG, Mulliken JB. Diffuse capillary malformation with overgrowth: a clinical subtype of vascular anomalies with hypertrophy. J Am Acad Dermatol. 2013;69:589-594. doi:10.1016/j.jaad.2013.05.030
  3. Cubiró X, Rozas‐Muñoz E, Castel P, et al. Clinical and genetic evaluation of six children with diffuse capillary malformation and undergrowth. Pediatr Dermatol. 2020;37:833-838. doi:10.1111/pde.14252
  4. Uihlein LC, Liang MG, Fishman SJ, et al. Capillary-venous malformation in the lower limb. Pediatr Dermatol. 2013;30:541-548. doi:10.1111/pde.12186
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E17-E19
Page Number
E17-E19
Publications
Publications
Topics
Article Type
Display Headline
Diffuse Capillary Malformation With Undergrowth of a Limb in a Boy
Display Headline
Diffuse Capillary Malformation With Undergrowth of a Limb in a Boy
Sections
Inside the Article

Practice Points

  • The term diffuse capillary malformation with undergrowth (DCMU) describes a distinct counterpart to diffuse capillary malformation with overgrowth. It can be challenging to distinguish from other vascular malformations associated with congenital structural abnormalities.
  • The vascular patterning of DCMU may fade over time, but patients should continue to be monitored for their structural incongruity.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

What’s Eating You? Update on the Sticktight Flea (Echidnophaga gallinacea)

Article Type
Changed
Thu, 12/14/2023 - 11:04
Display Headline
What’s Eating You? Update on the Sticktight Flea (Echidnophaga gallinacea)

Fleas (order Siphonaptera) are vectors for various diseases, such as plague (as carriers of Yersinia pestis) and rickettsial infections.1-4 The sticktight flea (Echidnophaga gallinacea) commonly is seen on birds and mammals, including ground squirrels, dogs, cats, and rodents, and can attach to its host for days at a time by burrowing its head into the skin. Similar to other fleas, the sticktight flea needs a blood supply to reproduce.5 Therefore, it is important to study the sticktight flea, its habitat, and infection patterns to improve public health and prevent infestation.

Identification

Echidnophaga gallinacea is named for the female flea’s behavior—it “sticks tight” to the surface of the host by embedding its head into the skin for days at a time.5 The sticktight flea and the rat flea (Xenopsylla cheopis) can be differentiated by the sticktight’s reduced thorax and lack of a pleural rod (the vertical ridge that divides the mesosternum above the second pair of legs)(Figure, A and B). The sticktight flea can be differentiated from the dog flea (Ctenocephalides canis) and the cat flea (Ctenocephalides felis) by its lack of genal ctenidia (horizontal combs in the mustache area) and pronotal ctenidia (vertical combs behind the head)(Figure, B and C).6,7 Other defining features of E gallinacea include 2 pairs of large postantennal setae (hairs) on its anteriorly flattened head; a C-shaped reproductive organ known as the spermatheca; and broad maxillary lacinia (Figure, C).8

Anatomy of the sticktight flea (Echidnophaga gallinacea), rat flea (Xenopsylla cheopis), and cat flea (Ctenocephalides felis), respectively
A-C, Anatomy of the sticktight flea (Echidnophaga gallinacea), rat flea (Xenopsylla cheopis), and cat flea (Ctenocephalides felis), respectively. The rat flea has a pleural rod and the cat flea has genal and pronotal ctenidia (combs), which are absent in E gallinacean.

Habitat, Seasonality, and Behavior

Echidnophaga gallinacea commonly infests the comb, wattles, and surrounding ears of chickens; the flea also has been found on dogs, cats, rodents, and other species of birds.9 The sticktight flea is more prevalent in summer and autumn, which may explain its predominance in warmer climates, including California, Florida, Mexico, Egypt, Africa, and Iran.1,9-11

When a female sticktight flea begins to feed, it stays on the host for days at a time, waiting for a male.5 The female deposits its fertilized eggs in nests on the host or in lesions caused by infestation. Eventually, eggs hatch and fall into soil, where they lay dormant or grow to adulthood.5

Cutaneous Reaction to Infestation

Flea bites cause a hypersensitivity reaction, with pruritic pustules and erythematous papules that have a central punctum.12 In a reported case in Los Angeles, California, a female sticktight flea buried itself into the cheek of a young boy for more than 12 hours. The lesion was not marked by surrounding erythema, tenderness, pruritus, or swelling; however, several days after the flea was removed, erythema developed at the site then spontaneously resolved.7 In a study of dogs that were infested with E gallinacea, the flea never disengaged to attach to a human; when the flea was deliberately placed on a human, it fed and left hastily.11

Management

Because E gallinacea burrows its head into the skin, the best removal method is applying slow gentle traction under sterile conditions to ensure removal of mouthparts.7 An oral antihistamine can be administered or a topical antihistamine or corticosteroid can be applied to the affected area.12 Flea infestation should be treated with an insecticide. Affected animals should be treated by a veterinarian using a pesticide, such as fipronil, selamectin, imidacloprid, metaflumizone, nitenpyram, lufenuron, methoprene, or pyriproxyfen.13

References
  1. Hubbart JA, Jachowski DS, Eads DA. Seasonal and among-site variation in the occurrence and abundance of fleas on California ground squirrels (Otospermophilus beecheyi). J Vector Ecol. 2011;36:117-123. doi:10.1111/j.1948-7134.2011.00148.x
  2. Jiang J, Maina AN, Knobel DL, et al. Molecular detection of Rickettsia felis and Candidatus Rickettsia asemboensis in fleas from human habitats, Asembo, Kenya. Vector Borne Zoonotic Dis. 2013;13:550-558. doi:10.1089/vbz.2012.1123
  3. López-Pérez AM, Chaves A, Sánchez-Montes S, et al. Diversity of rickettsiae in domestic, synanthropic, and sylvatic mammals and their ectoparasites in a spotted fever-epidemic region at the western US-Mexico border. Transbound Emerg Dis. 2022;69:609-622. doi:10.1111/tbed.14027
  4. Ehlers J, Krüger A, Rakotondranary SJ, et al. Molecular detection of Rickettsia spp., Borrelia spp., Bartonella spp. and Yersinia pestis in ectoparasites of endemic and domestic animals in southwest Madagascar. Acta Trop. 2020;205:105339. doi:10.1016/j.actatropica.2020.105339
  5. Boughton RK, Atwell JW, Schoech SJ. An introduced generalist parasite, the sticktight flea (Echidnophaga gallinacea), and its pathology in the threatened Florida scrub-jay (Aphelocoma coerulescens). J Parasitol. 2006;92:941-948. doi:10.1645/GE-769R.1
  6. Bitam I, Dittmar K, Parola P, et al. Fleas and flea-borne diseases. Int J Infect Dis. 2010;14:e667-e676. doi:10.1016/j.ijid.2009.11.011
  7. Linardi PM, Santos JLC. Ctenocephalides felis felis vs. Ctenocephalides canis (Siphonaptera: Pulicidae): some issues in correctly identify these species. Rev Bras Parasitol Vet. 2012;21:345-354. doi:10.1590/s1984-29612012000400002
  8. Carlson JC, Fox MS. A sticktight flea removed from the cheek of a two-year-old boy from Los Angeles. Dermatol Online J. 2009;15:4. https://doi.org/10.5070/D36vb8p1b1
  9. Mirzaei M, Ghashghaei O, Yakhchali M. Prevalence of ectoparasites of indigenous chickens from Dalahu region, Kermanshah province, Iran. Turkiye Parazitol Derg. 2016;40:13-16. doi:10.5152/tpd.2016.4185
  10. Farid DS, Sallam NH, Eldein AMS, et al. Cross-sectional seasonal prevalence and relative risk of ectoparasitic infestations of rodents in North Sinai, Egypt. Vet World. 2021;14:2996-3006. doi:10.14202/vetworld.2021.2996-3006
  11. Harman DW, Halliwell RE, Greiner EC. Flea species from dogs and cats in north-central Florida. Vet Parasitol. 1987;23:135-140. doi:10.1016/0304-4017(87)90031-8
  12. Anderson J, Paterek E. Flea bites. StatPearls [Internet]. StatPearls Publishing; 2023. Updated August 8, 2023. Accessed November 27, 2023. https://www.ncbi.nlm.nih.gov/books/NBK541118/
  13. Gyimesi ZS, Hayden ER, Greiner EC. Sticktight flea (Echidnophaga gallinacea) infestation in a Victoria crowned pigeon (Goura victoria). J Zoo Wildl Med. 2007;38:594-596. doi:10.1638/2007-0062.1
Article PDF
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Penny Lane Huebsch is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Penny Lane Huebsch, MS, 96 Jonathan Lucas St, Ste 601, Charleston, SC 29425 (huebsch@musc.edu).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E15-E16
Sections
Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Penny Lane Huebsch is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Penny Lane Huebsch, MS, 96 Jonathan Lucas St, Ste 601, Charleston, SC 29425 (huebsch@musc.edu).

Author and Disclosure Information

From the Medical University of South Carolina, Charleston. Penny Lane Huebsch is from the College of Medicine, and Dr. Elston is from the Department of Dermatology and Dermatologic Surgery.

The authors report no conflict of interest.

The images are in the public domain.

Correspondence: Penny Lane Huebsch, MS, 96 Jonathan Lucas St, Ste 601, Charleston, SC 29425 (huebsch@musc.edu).

Article PDF
Article PDF

Fleas (order Siphonaptera) are vectors for various diseases, such as plague (as carriers of Yersinia pestis) and rickettsial infections.1-4 The sticktight flea (Echidnophaga gallinacea) commonly is seen on birds and mammals, including ground squirrels, dogs, cats, and rodents, and can attach to its host for days at a time by burrowing its head into the skin. Similar to other fleas, the sticktight flea needs a blood supply to reproduce.5 Therefore, it is important to study the sticktight flea, its habitat, and infection patterns to improve public health and prevent infestation.

Identification

Echidnophaga gallinacea is named for the female flea’s behavior—it “sticks tight” to the surface of the host by embedding its head into the skin for days at a time.5 The sticktight flea and the rat flea (Xenopsylla cheopis) can be differentiated by the sticktight’s reduced thorax and lack of a pleural rod (the vertical ridge that divides the mesosternum above the second pair of legs)(Figure, A and B). The sticktight flea can be differentiated from the dog flea (Ctenocephalides canis) and the cat flea (Ctenocephalides felis) by its lack of genal ctenidia (horizontal combs in the mustache area) and pronotal ctenidia (vertical combs behind the head)(Figure, B and C).6,7 Other defining features of E gallinacea include 2 pairs of large postantennal setae (hairs) on its anteriorly flattened head; a C-shaped reproductive organ known as the spermatheca; and broad maxillary lacinia (Figure, C).8

Anatomy of the sticktight flea (Echidnophaga gallinacea), rat flea (Xenopsylla cheopis), and cat flea (Ctenocephalides felis), respectively
A-C, Anatomy of the sticktight flea (Echidnophaga gallinacea), rat flea (Xenopsylla cheopis), and cat flea (Ctenocephalides felis), respectively. The rat flea has a pleural rod and the cat flea has genal and pronotal ctenidia (combs), which are absent in E gallinacean.

Habitat, Seasonality, and Behavior

Echidnophaga gallinacea commonly infests the comb, wattles, and surrounding ears of chickens; the flea also has been found on dogs, cats, rodents, and other species of birds.9 The sticktight flea is more prevalent in summer and autumn, which may explain its predominance in warmer climates, including California, Florida, Mexico, Egypt, Africa, and Iran.1,9-11

When a female sticktight flea begins to feed, it stays on the host for days at a time, waiting for a male.5 The female deposits its fertilized eggs in nests on the host or in lesions caused by infestation. Eventually, eggs hatch and fall into soil, where they lay dormant or grow to adulthood.5

Cutaneous Reaction to Infestation

Flea bites cause a hypersensitivity reaction, with pruritic pustules and erythematous papules that have a central punctum.12 In a reported case in Los Angeles, California, a female sticktight flea buried itself into the cheek of a young boy for more than 12 hours. The lesion was not marked by surrounding erythema, tenderness, pruritus, or swelling; however, several days after the flea was removed, erythema developed at the site then spontaneously resolved.7 In a study of dogs that were infested with E gallinacea, the flea never disengaged to attach to a human; when the flea was deliberately placed on a human, it fed and left hastily.11

Management

Because E gallinacea burrows its head into the skin, the best removal method is applying slow gentle traction under sterile conditions to ensure removal of mouthparts.7 An oral antihistamine can be administered or a topical antihistamine or corticosteroid can be applied to the affected area.12 Flea infestation should be treated with an insecticide. Affected animals should be treated by a veterinarian using a pesticide, such as fipronil, selamectin, imidacloprid, metaflumizone, nitenpyram, lufenuron, methoprene, or pyriproxyfen.13

Fleas (order Siphonaptera) are vectors for various diseases, such as plague (as carriers of Yersinia pestis) and rickettsial infections.1-4 The sticktight flea (Echidnophaga gallinacea) commonly is seen on birds and mammals, including ground squirrels, dogs, cats, and rodents, and can attach to its host for days at a time by burrowing its head into the skin. Similar to other fleas, the sticktight flea needs a blood supply to reproduce.5 Therefore, it is important to study the sticktight flea, its habitat, and infection patterns to improve public health and prevent infestation.

Identification

Echidnophaga gallinacea is named for the female flea’s behavior—it “sticks tight” to the surface of the host by embedding its head into the skin for days at a time.5 The sticktight flea and the rat flea (Xenopsylla cheopis) can be differentiated by the sticktight’s reduced thorax and lack of a pleural rod (the vertical ridge that divides the mesosternum above the second pair of legs)(Figure, A and B). The sticktight flea can be differentiated from the dog flea (Ctenocephalides canis) and the cat flea (Ctenocephalides felis) by its lack of genal ctenidia (horizontal combs in the mustache area) and pronotal ctenidia (vertical combs behind the head)(Figure, B and C).6,7 Other defining features of E gallinacea include 2 pairs of large postantennal setae (hairs) on its anteriorly flattened head; a C-shaped reproductive organ known as the spermatheca; and broad maxillary lacinia (Figure, C).8

Anatomy of the sticktight flea (Echidnophaga gallinacea), rat flea (Xenopsylla cheopis), and cat flea (Ctenocephalides felis), respectively
A-C, Anatomy of the sticktight flea (Echidnophaga gallinacea), rat flea (Xenopsylla cheopis), and cat flea (Ctenocephalides felis), respectively. The rat flea has a pleural rod and the cat flea has genal and pronotal ctenidia (combs), which are absent in E gallinacean.

Habitat, Seasonality, and Behavior

Echidnophaga gallinacea commonly infests the comb, wattles, and surrounding ears of chickens; the flea also has been found on dogs, cats, rodents, and other species of birds.9 The sticktight flea is more prevalent in summer and autumn, which may explain its predominance in warmer climates, including California, Florida, Mexico, Egypt, Africa, and Iran.1,9-11

When a female sticktight flea begins to feed, it stays on the host for days at a time, waiting for a male.5 The female deposits its fertilized eggs in nests on the host or in lesions caused by infestation. Eventually, eggs hatch and fall into soil, where they lay dormant or grow to adulthood.5

Cutaneous Reaction to Infestation

Flea bites cause a hypersensitivity reaction, with pruritic pustules and erythematous papules that have a central punctum.12 In a reported case in Los Angeles, California, a female sticktight flea buried itself into the cheek of a young boy for more than 12 hours. The lesion was not marked by surrounding erythema, tenderness, pruritus, or swelling; however, several days after the flea was removed, erythema developed at the site then spontaneously resolved.7 In a study of dogs that were infested with E gallinacea, the flea never disengaged to attach to a human; when the flea was deliberately placed on a human, it fed and left hastily.11

Management

Because E gallinacea burrows its head into the skin, the best removal method is applying slow gentle traction under sterile conditions to ensure removal of mouthparts.7 An oral antihistamine can be administered or a topical antihistamine or corticosteroid can be applied to the affected area.12 Flea infestation should be treated with an insecticide. Affected animals should be treated by a veterinarian using a pesticide, such as fipronil, selamectin, imidacloprid, metaflumizone, nitenpyram, lufenuron, methoprene, or pyriproxyfen.13

References
  1. Hubbart JA, Jachowski DS, Eads DA. Seasonal and among-site variation in the occurrence and abundance of fleas on California ground squirrels (Otospermophilus beecheyi). J Vector Ecol. 2011;36:117-123. doi:10.1111/j.1948-7134.2011.00148.x
  2. Jiang J, Maina AN, Knobel DL, et al. Molecular detection of Rickettsia felis and Candidatus Rickettsia asemboensis in fleas from human habitats, Asembo, Kenya. Vector Borne Zoonotic Dis. 2013;13:550-558. doi:10.1089/vbz.2012.1123
  3. López-Pérez AM, Chaves A, Sánchez-Montes S, et al. Diversity of rickettsiae in domestic, synanthropic, and sylvatic mammals and their ectoparasites in a spotted fever-epidemic region at the western US-Mexico border. Transbound Emerg Dis. 2022;69:609-622. doi:10.1111/tbed.14027
  4. Ehlers J, Krüger A, Rakotondranary SJ, et al. Molecular detection of Rickettsia spp., Borrelia spp., Bartonella spp. and Yersinia pestis in ectoparasites of endemic and domestic animals in southwest Madagascar. Acta Trop. 2020;205:105339. doi:10.1016/j.actatropica.2020.105339
  5. Boughton RK, Atwell JW, Schoech SJ. An introduced generalist parasite, the sticktight flea (Echidnophaga gallinacea), and its pathology in the threatened Florida scrub-jay (Aphelocoma coerulescens). J Parasitol. 2006;92:941-948. doi:10.1645/GE-769R.1
  6. Bitam I, Dittmar K, Parola P, et al. Fleas and flea-borne diseases. Int J Infect Dis. 2010;14:e667-e676. doi:10.1016/j.ijid.2009.11.011
  7. Linardi PM, Santos JLC. Ctenocephalides felis felis vs. Ctenocephalides canis (Siphonaptera: Pulicidae): some issues in correctly identify these species. Rev Bras Parasitol Vet. 2012;21:345-354. doi:10.1590/s1984-29612012000400002
  8. Carlson JC, Fox MS. A sticktight flea removed from the cheek of a two-year-old boy from Los Angeles. Dermatol Online J. 2009;15:4. https://doi.org/10.5070/D36vb8p1b1
  9. Mirzaei M, Ghashghaei O, Yakhchali M. Prevalence of ectoparasites of indigenous chickens from Dalahu region, Kermanshah province, Iran. Turkiye Parazitol Derg. 2016;40:13-16. doi:10.5152/tpd.2016.4185
  10. Farid DS, Sallam NH, Eldein AMS, et al. Cross-sectional seasonal prevalence and relative risk of ectoparasitic infestations of rodents in North Sinai, Egypt. Vet World. 2021;14:2996-3006. doi:10.14202/vetworld.2021.2996-3006
  11. Harman DW, Halliwell RE, Greiner EC. Flea species from dogs and cats in north-central Florida. Vet Parasitol. 1987;23:135-140. doi:10.1016/0304-4017(87)90031-8
  12. Anderson J, Paterek E. Flea bites. StatPearls [Internet]. StatPearls Publishing; 2023. Updated August 8, 2023. Accessed November 27, 2023. https://www.ncbi.nlm.nih.gov/books/NBK541118/
  13. Gyimesi ZS, Hayden ER, Greiner EC. Sticktight flea (Echidnophaga gallinacea) infestation in a Victoria crowned pigeon (Goura victoria). J Zoo Wildl Med. 2007;38:594-596. doi:10.1638/2007-0062.1
References
  1. Hubbart JA, Jachowski DS, Eads DA. Seasonal and among-site variation in the occurrence and abundance of fleas on California ground squirrels (Otospermophilus beecheyi). J Vector Ecol. 2011;36:117-123. doi:10.1111/j.1948-7134.2011.00148.x
  2. Jiang J, Maina AN, Knobel DL, et al. Molecular detection of Rickettsia felis and Candidatus Rickettsia asemboensis in fleas from human habitats, Asembo, Kenya. Vector Borne Zoonotic Dis. 2013;13:550-558. doi:10.1089/vbz.2012.1123
  3. López-Pérez AM, Chaves A, Sánchez-Montes S, et al. Diversity of rickettsiae in domestic, synanthropic, and sylvatic mammals and their ectoparasites in a spotted fever-epidemic region at the western US-Mexico border. Transbound Emerg Dis. 2022;69:609-622. doi:10.1111/tbed.14027
  4. Ehlers J, Krüger A, Rakotondranary SJ, et al. Molecular detection of Rickettsia spp., Borrelia spp., Bartonella spp. and Yersinia pestis in ectoparasites of endemic and domestic animals in southwest Madagascar. Acta Trop. 2020;205:105339. doi:10.1016/j.actatropica.2020.105339
  5. Boughton RK, Atwell JW, Schoech SJ. An introduced generalist parasite, the sticktight flea (Echidnophaga gallinacea), and its pathology in the threatened Florida scrub-jay (Aphelocoma coerulescens). J Parasitol. 2006;92:941-948. doi:10.1645/GE-769R.1
  6. Bitam I, Dittmar K, Parola P, et al. Fleas and flea-borne diseases. Int J Infect Dis. 2010;14:e667-e676. doi:10.1016/j.ijid.2009.11.011
  7. Linardi PM, Santos JLC. Ctenocephalides felis felis vs. Ctenocephalides canis (Siphonaptera: Pulicidae): some issues in correctly identify these species. Rev Bras Parasitol Vet. 2012;21:345-354. doi:10.1590/s1984-29612012000400002
  8. Carlson JC, Fox MS. A sticktight flea removed from the cheek of a two-year-old boy from Los Angeles. Dermatol Online J. 2009;15:4. https://doi.org/10.5070/D36vb8p1b1
  9. Mirzaei M, Ghashghaei O, Yakhchali M. Prevalence of ectoparasites of indigenous chickens from Dalahu region, Kermanshah province, Iran. Turkiye Parazitol Derg. 2016;40:13-16. doi:10.5152/tpd.2016.4185
  10. Farid DS, Sallam NH, Eldein AMS, et al. Cross-sectional seasonal prevalence and relative risk of ectoparasitic infestations of rodents in North Sinai, Egypt. Vet World. 2021;14:2996-3006. doi:10.14202/vetworld.2021.2996-3006
  11. Harman DW, Halliwell RE, Greiner EC. Flea species from dogs and cats in north-central Florida. Vet Parasitol. 1987;23:135-140. doi:10.1016/0304-4017(87)90031-8
  12. Anderson J, Paterek E. Flea bites. StatPearls [Internet]. StatPearls Publishing; 2023. Updated August 8, 2023. Accessed November 27, 2023. https://www.ncbi.nlm.nih.gov/books/NBK541118/
  13. Gyimesi ZS, Hayden ER, Greiner EC. Sticktight flea (Echidnophaga gallinacea) infestation in a Victoria crowned pigeon (Goura victoria). J Zoo Wildl Med. 2007;38:594-596. doi:10.1638/2007-0062.1
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E15-E16
Page Number
E15-E16
Publications
Publications
Topics
Article Type
Display Headline
What’s Eating You? Update on the Sticktight Flea (Echidnophaga gallinacea)
Display Headline
What’s Eating You? Update on the Sticktight Flea (Echidnophaga gallinacea)
Sections
Inside the Article

Practice Points

  • The sticktight flea (Echidnophaga gallinacea) attaches to its host by embedding its head in the skin for days at a time.
  • Unlike other fleas that bite and run, the sticktight flea can be identified dermoscopically.
  • The sticktight flea serves as a vector for plague as a carrier of Yersinia pestis, rickettsial infections, and other diseases.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Large Indurated Plaque on the Chest With Ulceration and Necrosis

Article Type
Changed
Wed, 12/13/2023 - 10:34
Display Headline
Large Indurated Plaque on the Chest With Ulceration and Necrosis

The Diagnosis: Carcinoma en Cuirasse

Histopathology demonstrated a cellular infiltrate filling the dermis with sparing of the papillary and superficial reticular dermis (Figure 1A). The cells were arranged in strands and cords that infiltrated between sclerotic collagen bundles. Cytomorphologically, the cells ranged from epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (Figure 1B). Occasional mitotic figures were identified, and cells demonstrated diffuse nuclear positivity for GATA-3 (Figure 1C); 55% of the cells demonstrated estrogen receptor positivity, and immunohistochemistry of progesterone receptors was negative. These findings confirmed our patient’s diagnosis of breast carcinoma en cuirasse (CeC) as the primary manifestation of metastatic invasive ductal carcinoma. Our patient was treated with intravenous chemotherapy and tamoxifen.

A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboid
FIGURE 1. A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (H&E, original magnification ×400). C, The cells also demonstrated diffuse nuclear positivity for GATA-3 (original magnification ×40).

Histopathologic findings of morphea include thickened hyalinized collagen bundles and loss of adventitial fat.1 A diagnosis of chronic radiation dermatitis was inconsistent with our patient’s medical history and biopsy results, as pathology should reveal hyalinized collagen or stellate radiation fibroblasts.2,3 Nests of squamous epithelial cells with abundant eosinophilic cytoplasm and large vesicular nuclei were not seen, excluding squamous cell carcinoma as a possible diagnosis.4 Although sclerosing sweat duct carcinoma is characterized by infiltrating cords in sclerotic dermis, the cells were not arranged in ductlike structures 1– to 2–cell layers thick, excluding this diagnosis.5

Carcinoma en cuirasse—named for skin involvement that appears similar to the metal breastplate of a cuirassier—is a rare form of cutaneous metastasis that typically presents with extensive infiltrative plaques resulting in fibrosis of the skin and subcutaneous tissue.6,7 Carcinoma en cuirasse most commonly metastasizes from the breast but also may represent metastases from the lungs, gastrointestinal tract, or genitourinary systems.8 In the setting of a primary breast malignancy, metastatic plaques of CeC tend to represent tumor recurrence following a mastectomy procedure; however, in rare cases CeC can present as the primary manifestation of breast cancer or as a result of untreated malignancy.6,9 In our patient, CeC was the primary manifestation of metastatic invasive ductal carcinoma with additional paraneoplastic ichthyosis (Figure 2).

Ichthyotic plaques with brown scaling on the leg.
FIGURE 2. Ichthyotic plaques with brown scaling on the leg.

Carcinoma en cuirasse comprises 3% to 6% of cutaneous metastases originating from the breast.10,11 Breast cancer is the most common primary neoplasm displaying extracutaneous metastasis, comprising 70% of all cutaneous metastases in females.11 Cutaneous metastasis often indicates late stage of disease, portending a poor prognosis. In our patient, the cutaneous nodules were present for approximately 3 years prior to the diagnosis of stage IV invasive ductal cell carcinoma with metastasis to the skin and lungs. Prior to admission, she had not been diagnosed with breast cancer, thus no treatments had been administered. It is uncommon for CeC to present as the initial finding and without prior treatment of the underlying malignancy. The median length of survival after diagnosis of cutaneous metastasis from breast cancer is 13.8 months, with a 10-year survival rate of 3.1%.12

In addition to cutaneous metastasis, breast cancer also may present with paraneoplastic dermatoses such as ichthyosis.13 Ichthyosis is characterized by extreme dryness, flaking, thickening, and mild pruritus.14 It most commonly is an inherited condition, but it may be acquired due to malignancy. Acquired ichthyosis may manifest in systemic diseases including systemic lupus erythematosus, sarcoidosis, and hypothyroidism.15 Although acquired ichthyosis is rare, it has been reported in cases of internal malignancy, most commonly lymphoproliferative malignancies and less frequently carcinoma of the breasts, cervix, and lungs. Patients who acquire ichthyosis in association with malignancy usually present with late-stage disease.15 Our patient acquired ichthyosis 3 months prior to admission and had never experienced it previously. Although the exact mechanism for acquiring ichthyosis remains unknown, it is uncertain if ichthyosis associated with malignancy is paraneoplastic or a result of chemotherapy.14,16 In this case, the patient had not yet started chemotherapy at the time of the ichthyosis diagnosis, suggesting a paraneoplastic etiology.

Carcinoma en cuirasse and paraneoplastic ichthyosis individually are extremely rare manifestations of breast cancer. Thus, it is even rarer for these conditions to present concurrently. Treatment options for CeC include chemotherapy, radiotherapy, hormonal antagonists, and snake venom.11 Systemic chemotherapy targeting the histopathologic type of the primary tumor is the treatment of choice. Other treatment methods usually are chosen for late stages of disease progression.10 Paraneoplastic ichthyosis has been reported to show improvement with treatment of the underlying primary malignancy by surgical removal or chemotherapy.14,17 Tamoxifen less commonly is used for systemic treatment of CeC, but one case in the literature reported favorable outcomes.18

We describe 2 rare cutaneous manifestations of breast cancer occurring concomitantly: CeC and paraneoplastic ichthyosis. The combination of clinical and pathologic findings presented in this case solidified the diagnosis of metastatic invasive ductal carcinoma. We aim to improve recognition of paraneoplastic skin findings to accelerate the process of effective and efficient treatment.

References
  1. Walker D, Susa JS, Currimbhoy S, et al. Histopathological changes in morphea and their clinical correlates: results from the Morphea in Adults and Children Cohort V. J Am Acad Dermatol. 2017;76:1124-1130. https://doi.org/10.1016/j.jaad.2016.12.020
  2. Borrelli MR, Shen AH, Lee GK, et al. Radiation-induced skin fibrosis: pathogenesis, current treatment options, and emerging therapeutics. Ann Plast Surg. 2019;83(4 suppl 1):S59-S64. https://doi.org/10.1097/SAP.0000000000002098
  3. Boncher J, Bergfeld WF. Fluoroscopy-induced chronic radiation dermatitis: a report of two additional cases and a brief review of the literature. J Cutan Pathol. 2012;39:63-67. https://doi.org/10.1111/j .1600-0560.2011.01754.x
  4. Cassarino DS, Derienzo DP, Barr RJ. Cutaneous squamous cell carcinoma: a comprehensive clinicopathologic classification. part one. J Cutan Pathol. 2006;33:191-206. https://doi.org/10.1111 /j.0303-6987.2006.00516_1.x
  5. Harvey DT, Hu J, Long JA, et al. Sclerosing sweat duct carcinoma of the lower extremity treated with Mohs micrographic surgery. JAAD Case Rep. 2016;2:284-286. https://doi.org/10.1016/j.jdcr.2016.05.017
  6. Sharma V, Kumar A. Carcinoma en cuirasse. N Engl J Med. 2021;385:2562. doi:10.1056/NEJMicm2111669
  7. Oliveira GM, Zachetti DB, Barros HR, et al. Breast carcinoma en cuirasse—case report. An Bras Dermatol. 2013;88:608-610. doi:10.1590/abd1806-4841.20131926
  8. Alcaraz I, Cerroni L, Rütten A, et al. Cutaneous metastases from internal malignancies: a clinicopathologic and immunohistochemical review. Am J Dermatopathol. 2012;34:347-393. doi:10.1097 /DAD.0b013e31823069cf
  9. Glazebrook AJ, Tomaszewski W. Ichthyosiform atrophy of the skin in Hodgkin’s disease: report of a case, with reference to vitamin A metabolism. Arch Derm Syphilol. 1944;50:85-89. doi:10.1001 /archderm.1944.01510140008002
  10. Mordenti C, Concetta F, Cerroni M, et al. Cutaneous metastatic breast carcinoma: a study of 164 patients. Acta Dermatovenerol Alp Pannonica Adriat. 2000;9:143-148.
  11. Culver AL, Metter DM, Pippen JE Jr. Carcinoma en cuirasse. Proc (Bayl Univ Med Cent). 2019;32:263-265. doi:10.1080/08998280.2018.1564966
  12. Schoenlaub P, Sarraux A, Grosshans E, et al. Survival after cutaneous metastasis: a study of 200 cases [in French]. Ann Dermatol Venereol. 2001;128:1310-1315.
  13. Tan AR. Cutaneous manifestations of breast cancer. Semin Oncol. 2016;43:331-334. doi:10.1053/j.seminoncol.2016.02.030
  14. Song Y, Wu Y, Fan T. Dermatosis as the initial manifestation of malignant breast tumors: retrospective analysis of 4 cases. Breast Care. 2010;5:174-176. doi:10.1159/000314265
  15. Polisky RB, Bronson DM. Acquired ichthyosis in a patient with adenocarcinoma of the breast. Cutis. 1986;38:359-360.
  16. Haste AR. Acquired ichthyosis from breast cancer. Br Med J. 1967;4:96-98.
  17. Riesco Martínez MC, Muñoz Martín AJ, Zamberk Majlis P, et al. Acquired ichthyosis as a paraneoplastic syndrome in Hodgkin’s disease. Clin Transl Oncol. 2009;11:552-553. doi:10.1007/s12094-009-0402-2
  18. Siddiqui MA, Zaman MN. Primary carcinoma en cuirasse. J Am Geriatr Soc. 1996;44:221-222. doi:10.1111/j.1532-5415.1996.tb02455.xssss
Article PDF
Author and Disclosure Information

Caroline E. Roberts and Dr. Cascardo are from the William Beaumont School of Medicine, Oakland University, Rochester, Michigan. Dr. Cornell is from the Department of Dermatology, Trinity Health Ann Arbor, Ypsilanti, Michigan.

The authors report no conflict of interest.

Correspondence: Caroline E. Roberts, BM, 586 Pioneer Dr, Rochester, MI 48309 (carolineroberts@oakland.edu).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E12-E14
Sections
Author and Disclosure Information

Caroline E. Roberts and Dr. Cascardo are from the William Beaumont School of Medicine, Oakland University, Rochester, Michigan. Dr. Cornell is from the Department of Dermatology, Trinity Health Ann Arbor, Ypsilanti, Michigan.

The authors report no conflict of interest.

Correspondence: Caroline E. Roberts, BM, 586 Pioneer Dr, Rochester, MI 48309 (carolineroberts@oakland.edu).

Author and Disclosure Information

Caroline E. Roberts and Dr. Cascardo are from the William Beaumont School of Medicine, Oakland University, Rochester, Michigan. Dr. Cornell is from the Department of Dermatology, Trinity Health Ann Arbor, Ypsilanti, Michigan.

The authors report no conflict of interest.

Correspondence: Caroline E. Roberts, BM, 586 Pioneer Dr, Rochester, MI 48309 (carolineroberts@oakland.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Carcinoma en Cuirasse

Histopathology demonstrated a cellular infiltrate filling the dermis with sparing of the papillary and superficial reticular dermis (Figure 1A). The cells were arranged in strands and cords that infiltrated between sclerotic collagen bundles. Cytomorphologically, the cells ranged from epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (Figure 1B). Occasional mitotic figures were identified, and cells demonstrated diffuse nuclear positivity for GATA-3 (Figure 1C); 55% of the cells demonstrated estrogen receptor positivity, and immunohistochemistry of progesterone receptors was negative. These findings confirmed our patient’s diagnosis of breast carcinoma en cuirasse (CeC) as the primary manifestation of metastatic invasive ductal carcinoma. Our patient was treated with intravenous chemotherapy and tamoxifen.

A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboid
FIGURE 1. A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (H&E, original magnification ×400). C, The cells also demonstrated diffuse nuclear positivity for GATA-3 (original magnification ×40).

Histopathologic findings of morphea include thickened hyalinized collagen bundles and loss of adventitial fat.1 A diagnosis of chronic radiation dermatitis was inconsistent with our patient’s medical history and biopsy results, as pathology should reveal hyalinized collagen or stellate radiation fibroblasts.2,3 Nests of squamous epithelial cells with abundant eosinophilic cytoplasm and large vesicular nuclei were not seen, excluding squamous cell carcinoma as a possible diagnosis.4 Although sclerosing sweat duct carcinoma is characterized by infiltrating cords in sclerotic dermis, the cells were not arranged in ductlike structures 1– to 2–cell layers thick, excluding this diagnosis.5

Carcinoma en cuirasse—named for skin involvement that appears similar to the metal breastplate of a cuirassier—is a rare form of cutaneous metastasis that typically presents with extensive infiltrative plaques resulting in fibrosis of the skin and subcutaneous tissue.6,7 Carcinoma en cuirasse most commonly metastasizes from the breast but also may represent metastases from the lungs, gastrointestinal tract, or genitourinary systems.8 In the setting of a primary breast malignancy, metastatic plaques of CeC tend to represent tumor recurrence following a mastectomy procedure; however, in rare cases CeC can present as the primary manifestation of breast cancer or as a result of untreated malignancy.6,9 In our patient, CeC was the primary manifestation of metastatic invasive ductal carcinoma with additional paraneoplastic ichthyosis (Figure 2).

Ichthyotic plaques with brown scaling on the leg.
FIGURE 2. Ichthyotic plaques with brown scaling on the leg.

Carcinoma en cuirasse comprises 3% to 6% of cutaneous metastases originating from the breast.10,11 Breast cancer is the most common primary neoplasm displaying extracutaneous metastasis, comprising 70% of all cutaneous metastases in females.11 Cutaneous metastasis often indicates late stage of disease, portending a poor prognosis. In our patient, the cutaneous nodules were present for approximately 3 years prior to the diagnosis of stage IV invasive ductal cell carcinoma with metastasis to the skin and lungs. Prior to admission, she had not been diagnosed with breast cancer, thus no treatments had been administered. It is uncommon for CeC to present as the initial finding and without prior treatment of the underlying malignancy. The median length of survival after diagnosis of cutaneous metastasis from breast cancer is 13.8 months, with a 10-year survival rate of 3.1%.12

In addition to cutaneous metastasis, breast cancer also may present with paraneoplastic dermatoses such as ichthyosis.13 Ichthyosis is characterized by extreme dryness, flaking, thickening, and mild pruritus.14 It most commonly is an inherited condition, but it may be acquired due to malignancy. Acquired ichthyosis may manifest in systemic diseases including systemic lupus erythematosus, sarcoidosis, and hypothyroidism.15 Although acquired ichthyosis is rare, it has been reported in cases of internal malignancy, most commonly lymphoproliferative malignancies and less frequently carcinoma of the breasts, cervix, and lungs. Patients who acquire ichthyosis in association with malignancy usually present with late-stage disease.15 Our patient acquired ichthyosis 3 months prior to admission and had never experienced it previously. Although the exact mechanism for acquiring ichthyosis remains unknown, it is uncertain if ichthyosis associated with malignancy is paraneoplastic or a result of chemotherapy.14,16 In this case, the patient had not yet started chemotherapy at the time of the ichthyosis diagnosis, suggesting a paraneoplastic etiology.

Carcinoma en cuirasse and paraneoplastic ichthyosis individually are extremely rare manifestations of breast cancer. Thus, it is even rarer for these conditions to present concurrently. Treatment options for CeC include chemotherapy, radiotherapy, hormonal antagonists, and snake venom.11 Systemic chemotherapy targeting the histopathologic type of the primary tumor is the treatment of choice. Other treatment methods usually are chosen for late stages of disease progression.10 Paraneoplastic ichthyosis has been reported to show improvement with treatment of the underlying primary malignancy by surgical removal or chemotherapy.14,17 Tamoxifen less commonly is used for systemic treatment of CeC, but one case in the literature reported favorable outcomes.18

We describe 2 rare cutaneous manifestations of breast cancer occurring concomitantly: CeC and paraneoplastic ichthyosis. The combination of clinical and pathologic findings presented in this case solidified the diagnosis of metastatic invasive ductal carcinoma. We aim to improve recognition of paraneoplastic skin findings to accelerate the process of effective and efficient treatment.

The Diagnosis: Carcinoma en Cuirasse

Histopathology demonstrated a cellular infiltrate filling the dermis with sparing of the papillary and superficial reticular dermis (Figure 1A). The cells were arranged in strands and cords that infiltrated between sclerotic collagen bundles. Cytomorphologically, the cells ranged from epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (Figure 1B). Occasional mitotic figures were identified, and cells demonstrated diffuse nuclear positivity for GATA-3 (Figure 1C); 55% of the cells demonstrated estrogen receptor positivity, and immunohistochemistry of progesterone receptors was negative. These findings confirmed our patient’s diagnosis of breast carcinoma en cuirasse (CeC) as the primary manifestation of metastatic invasive ductal carcinoma. Our patient was treated with intravenous chemotherapy and tamoxifen.

A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboid
FIGURE 1. A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (H&E, original magnification ×400). C, The cells also demonstrated diffuse nuclear positivity for GATA-3 (original magnification ×40).

Histopathologic findings of morphea include thickened hyalinized collagen bundles and loss of adventitial fat.1 A diagnosis of chronic radiation dermatitis was inconsistent with our patient’s medical history and biopsy results, as pathology should reveal hyalinized collagen or stellate radiation fibroblasts.2,3 Nests of squamous epithelial cells with abundant eosinophilic cytoplasm and large vesicular nuclei were not seen, excluding squamous cell carcinoma as a possible diagnosis.4 Although sclerosing sweat duct carcinoma is characterized by infiltrating cords in sclerotic dermis, the cells were not arranged in ductlike structures 1– to 2–cell layers thick, excluding this diagnosis.5

Carcinoma en cuirasse—named for skin involvement that appears similar to the metal breastplate of a cuirassier—is a rare form of cutaneous metastasis that typically presents with extensive infiltrative plaques resulting in fibrosis of the skin and subcutaneous tissue.6,7 Carcinoma en cuirasse most commonly metastasizes from the breast but also may represent metastases from the lungs, gastrointestinal tract, or genitourinary systems.8 In the setting of a primary breast malignancy, metastatic plaques of CeC tend to represent tumor recurrence following a mastectomy procedure; however, in rare cases CeC can present as the primary manifestation of breast cancer or as a result of untreated malignancy.6,9 In our patient, CeC was the primary manifestation of metastatic invasive ductal carcinoma with additional paraneoplastic ichthyosis (Figure 2).

Ichthyotic plaques with brown scaling on the leg.
FIGURE 2. Ichthyotic plaques with brown scaling on the leg.

Carcinoma en cuirasse comprises 3% to 6% of cutaneous metastases originating from the breast.10,11 Breast cancer is the most common primary neoplasm displaying extracutaneous metastasis, comprising 70% of all cutaneous metastases in females.11 Cutaneous metastasis often indicates late stage of disease, portending a poor prognosis. In our patient, the cutaneous nodules were present for approximately 3 years prior to the diagnosis of stage IV invasive ductal cell carcinoma with metastasis to the skin and lungs. Prior to admission, she had not been diagnosed with breast cancer, thus no treatments had been administered. It is uncommon for CeC to present as the initial finding and without prior treatment of the underlying malignancy. The median length of survival after diagnosis of cutaneous metastasis from breast cancer is 13.8 months, with a 10-year survival rate of 3.1%.12

In addition to cutaneous metastasis, breast cancer also may present with paraneoplastic dermatoses such as ichthyosis.13 Ichthyosis is characterized by extreme dryness, flaking, thickening, and mild pruritus.14 It most commonly is an inherited condition, but it may be acquired due to malignancy. Acquired ichthyosis may manifest in systemic diseases including systemic lupus erythematosus, sarcoidosis, and hypothyroidism.15 Although acquired ichthyosis is rare, it has been reported in cases of internal malignancy, most commonly lymphoproliferative malignancies and less frequently carcinoma of the breasts, cervix, and lungs. Patients who acquire ichthyosis in association with malignancy usually present with late-stage disease.15 Our patient acquired ichthyosis 3 months prior to admission and had never experienced it previously. Although the exact mechanism for acquiring ichthyosis remains unknown, it is uncertain if ichthyosis associated with malignancy is paraneoplastic or a result of chemotherapy.14,16 In this case, the patient had not yet started chemotherapy at the time of the ichthyosis diagnosis, suggesting a paraneoplastic etiology.

Carcinoma en cuirasse and paraneoplastic ichthyosis individually are extremely rare manifestations of breast cancer. Thus, it is even rarer for these conditions to present concurrently. Treatment options for CeC include chemotherapy, radiotherapy, hormonal antagonists, and snake venom.11 Systemic chemotherapy targeting the histopathologic type of the primary tumor is the treatment of choice. Other treatment methods usually are chosen for late stages of disease progression.10 Paraneoplastic ichthyosis has been reported to show improvement with treatment of the underlying primary malignancy by surgical removal or chemotherapy.14,17 Tamoxifen less commonly is used for systemic treatment of CeC, but one case in the literature reported favorable outcomes.18

We describe 2 rare cutaneous manifestations of breast cancer occurring concomitantly: CeC and paraneoplastic ichthyosis. The combination of clinical and pathologic findings presented in this case solidified the diagnosis of metastatic invasive ductal carcinoma. We aim to improve recognition of paraneoplastic skin findings to accelerate the process of effective and efficient treatment.

References
  1. Walker D, Susa JS, Currimbhoy S, et al. Histopathological changes in morphea and their clinical correlates: results from the Morphea in Adults and Children Cohort V. J Am Acad Dermatol. 2017;76:1124-1130. https://doi.org/10.1016/j.jaad.2016.12.020
  2. Borrelli MR, Shen AH, Lee GK, et al. Radiation-induced skin fibrosis: pathogenesis, current treatment options, and emerging therapeutics. Ann Plast Surg. 2019;83(4 suppl 1):S59-S64. https://doi.org/10.1097/SAP.0000000000002098
  3. Boncher J, Bergfeld WF. Fluoroscopy-induced chronic radiation dermatitis: a report of two additional cases and a brief review of the literature. J Cutan Pathol. 2012;39:63-67. https://doi.org/10.1111/j .1600-0560.2011.01754.x
  4. Cassarino DS, Derienzo DP, Barr RJ. Cutaneous squamous cell carcinoma: a comprehensive clinicopathologic classification. part one. J Cutan Pathol. 2006;33:191-206. https://doi.org/10.1111 /j.0303-6987.2006.00516_1.x
  5. Harvey DT, Hu J, Long JA, et al. Sclerosing sweat duct carcinoma of the lower extremity treated with Mohs micrographic surgery. JAAD Case Rep. 2016;2:284-286. https://doi.org/10.1016/j.jdcr.2016.05.017
  6. Sharma V, Kumar A. Carcinoma en cuirasse. N Engl J Med. 2021;385:2562. doi:10.1056/NEJMicm2111669
  7. Oliveira GM, Zachetti DB, Barros HR, et al. Breast carcinoma en cuirasse—case report. An Bras Dermatol. 2013;88:608-610. doi:10.1590/abd1806-4841.20131926
  8. Alcaraz I, Cerroni L, Rütten A, et al. Cutaneous metastases from internal malignancies: a clinicopathologic and immunohistochemical review. Am J Dermatopathol. 2012;34:347-393. doi:10.1097 /DAD.0b013e31823069cf
  9. Glazebrook AJ, Tomaszewski W. Ichthyosiform atrophy of the skin in Hodgkin’s disease: report of a case, with reference to vitamin A metabolism. Arch Derm Syphilol. 1944;50:85-89. doi:10.1001 /archderm.1944.01510140008002
  10. Mordenti C, Concetta F, Cerroni M, et al. Cutaneous metastatic breast carcinoma: a study of 164 patients. Acta Dermatovenerol Alp Pannonica Adriat. 2000;9:143-148.
  11. Culver AL, Metter DM, Pippen JE Jr. Carcinoma en cuirasse. Proc (Bayl Univ Med Cent). 2019;32:263-265. doi:10.1080/08998280.2018.1564966
  12. Schoenlaub P, Sarraux A, Grosshans E, et al. Survival after cutaneous metastasis: a study of 200 cases [in French]. Ann Dermatol Venereol. 2001;128:1310-1315.
  13. Tan AR. Cutaneous manifestations of breast cancer. Semin Oncol. 2016;43:331-334. doi:10.1053/j.seminoncol.2016.02.030
  14. Song Y, Wu Y, Fan T. Dermatosis as the initial manifestation of malignant breast tumors: retrospective analysis of 4 cases. Breast Care. 2010;5:174-176. doi:10.1159/000314265
  15. Polisky RB, Bronson DM. Acquired ichthyosis in a patient with adenocarcinoma of the breast. Cutis. 1986;38:359-360.
  16. Haste AR. Acquired ichthyosis from breast cancer. Br Med J. 1967;4:96-98.
  17. Riesco Martínez MC, Muñoz Martín AJ, Zamberk Majlis P, et al. Acquired ichthyosis as a paraneoplastic syndrome in Hodgkin’s disease. Clin Transl Oncol. 2009;11:552-553. doi:10.1007/s12094-009-0402-2
  18. Siddiqui MA, Zaman MN. Primary carcinoma en cuirasse. J Am Geriatr Soc. 1996;44:221-222. doi:10.1111/j.1532-5415.1996.tb02455.xssss
References
  1. Walker D, Susa JS, Currimbhoy S, et al. Histopathological changes in morphea and their clinical correlates: results from the Morphea in Adults and Children Cohort V. J Am Acad Dermatol. 2017;76:1124-1130. https://doi.org/10.1016/j.jaad.2016.12.020
  2. Borrelli MR, Shen AH, Lee GK, et al. Radiation-induced skin fibrosis: pathogenesis, current treatment options, and emerging therapeutics. Ann Plast Surg. 2019;83(4 suppl 1):S59-S64. https://doi.org/10.1097/SAP.0000000000002098
  3. Boncher J, Bergfeld WF. Fluoroscopy-induced chronic radiation dermatitis: a report of two additional cases and a brief review of the literature. J Cutan Pathol. 2012;39:63-67. https://doi.org/10.1111/j .1600-0560.2011.01754.x
  4. Cassarino DS, Derienzo DP, Barr RJ. Cutaneous squamous cell carcinoma: a comprehensive clinicopathologic classification. part one. J Cutan Pathol. 2006;33:191-206. https://doi.org/10.1111 /j.0303-6987.2006.00516_1.x
  5. Harvey DT, Hu J, Long JA, et al. Sclerosing sweat duct carcinoma of the lower extremity treated with Mohs micrographic surgery. JAAD Case Rep. 2016;2:284-286. https://doi.org/10.1016/j.jdcr.2016.05.017
  6. Sharma V, Kumar A. Carcinoma en cuirasse. N Engl J Med. 2021;385:2562. doi:10.1056/NEJMicm2111669
  7. Oliveira GM, Zachetti DB, Barros HR, et al. Breast carcinoma en cuirasse—case report. An Bras Dermatol. 2013;88:608-610. doi:10.1590/abd1806-4841.20131926
  8. Alcaraz I, Cerroni L, Rütten A, et al. Cutaneous metastases from internal malignancies: a clinicopathologic and immunohistochemical review. Am J Dermatopathol. 2012;34:347-393. doi:10.1097 /DAD.0b013e31823069cf
  9. Glazebrook AJ, Tomaszewski W. Ichthyosiform atrophy of the skin in Hodgkin’s disease: report of a case, with reference to vitamin A metabolism. Arch Derm Syphilol. 1944;50:85-89. doi:10.1001 /archderm.1944.01510140008002
  10. Mordenti C, Concetta F, Cerroni M, et al. Cutaneous metastatic breast carcinoma: a study of 164 patients. Acta Dermatovenerol Alp Pannonica Adriat. 2000;9:143-148.
  11. Culver AL, Metter DM, Pippen JE Jr. Carcinoma en cuirasse. Proc (Bayl Univ Med Cent). 2019;32:263-265. doi:10.1080/08998280.2018.1564966
  12. Schoenlaub P, Sarraux A, Grosshans E, et al. Survival after cutaneous metastasis: a study of 200 cases [in French]. Ann Dermatol Venereol. 2001;128:1310-1315.
  13. Tan AR. Cutaneous manifestations of breast cancer. Semin Oncol. 2016;43:331-334. doi:10.1053/j.seminoncol.2016.02.030
  14. Song Y, Wu Y, Fan T. Dermatosis as the initial manifestation of malignant breast tumors: retrospective analysis of 4 cases. Breast Care. 2010;5:174-176. doi:10.1159/000314265
  15. Polisky RB, Bronson DM. Acquired ichthyosis in a patient with adenocarcinoma of the breast. Cutis. 1986;38:359-360.
  16. Haste AR. Acquired ichthyosis from breast cancer. Br Med J. 1967;4:96-98.
  17. Riesco Martínez MC, Muñoz Martín AJ, Zamberk Majlis P, et al. Acquired ichthyosis as a paraneoplastic syndrome in Hodgkin’s disease. Clin Transl Oncol. 2009;11:552-553. doi:10.1007/s12094-009-0402-2
  18. Siddiqui MA, Zaman MN. Primary carcinoma en cuirasse. J Am Geriatr Soc. 1996;44:221-222. doi:10.1111/j.1532-5415.1996.tb02455.xssss
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E12-E14
Page Number
E12-E14
Publications
Publications
Topics
Article Type
Display Headline
Large Indurated Plaque on the Chest With Ulceration and Necrosis
Display Headline
Large Indurated Plaque on the Chest With Ulceration and Necrosis
Sections
Questionnaire Body

A 47-year-old woman with no notable medical history presented to the emergency department with shortness of breath on simple exertion as well as a large lesion on the chest that had slowly increased in size over the last 3 years. The lesion was not painful or pruritic, and she had been treating it with topical emollients without substantial improvement. Physical examination revealed a large indurated plaque with areas of ulceration and necrosis spanning the mid to lateral chest. Additionally, ichthyotic brown scaling was present on the arms and legs. Upon further questioning, the patient reported that the scales on the extremities appeared in the last 3 months and were not previously noted. She had no recent routine cancer screenings, and her family history was notable for a brother with brain cancer. A punch biopsy of the chest plaque was performed.

Large indurated plaque on the chest with ulceration and necrosis

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 12/13/2023 - 08:30
Un-Gate On Date
Wed, 12/13/2023 - 08:30
Use ProPublica
CFC Schedule Remove Status
Wed, 12/13/2023 - 08:30
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Fellowships in Complex Medical Dermatology

Article Type
Changed
Wed, 12/13/2023 - 10:31
Display Headline
Fellowships in Complex Medical Dermatology

Complex medical dermatology has become an emerging field in dermatology. Although a rather protean and broad term, complex medical dermatology encompasses patients with autoimmune conditions, bullous disease, connective tissue disease, vasculitis, severe dermatoses requiring immunomodulation, and inpatient consultations. Importantly, dermatology inpatient consultations aid in lowering health care costs due to accurate diagnoses, correct treatment, and decreased hospital stays.1 A fellowship is not required for holding an inpatient role in the hospital system as a dermatologist but can be beneficial. There are combined internal medicine–dermatology programs available for medical students applying to dermatology residency, but a complex medical dermatology fellowship is an option after residency for those who are interested. I believe that a focused complex medical dermatology fellowship differs from the training offered in combined internal medicine–dermatology residency. My fellow colleagues in combined internal medicine–dermatology programs are exposed to systemic manifestations of cutaneous disease and are experts in the interplay between the skin and other organ systems. However, the focus of their programs is with the intention of becoming double boarded in internal medicine and dermatology with comprehensive exposure to both fields. In my fellowship, I am able to tailor my schedule to focus on any dermatologic disease such as connective tissue disease, pruritus, graft vs host disease, and Merkel cell carcinoma. I ultimately can determine a niche in dermatology and hone my skills for a year under supervision.

Available Fellowships

Fellowship Locations—Importantly, the complex medical dermatology fellowship is not accredited by the Accreditation Council for Graduate Medical Education, which can make it difficult to identify and apply to programs. The complex medical dermatology fellowship is different than a rheumatology-dermatology fellowship, cutaneous oncology fellowship, pediatric dermatology fellowship, or other subspecialty fellowships such as those in itch or autoimmune blistering diseases. The fellowship often encompasses gaining clinical expertise in many of these conditions. I performed a thorough search online and spoke with complex medical dermatologists to compile a list of programs that offer a complex medical dermatology fellowship: Brigham and Women’s Hospital (Boston, Massachusetts); University of California San Francisco (San Francisco, California); University of Pennsylvania (Philadelphia, Pennsylvania); Cleveland Clinic (Cleveland, Ohio); and New York University (New York, New York)(Table). Only 1 spot is offered at each of these programs.

Complex Medical Dermatology Fellowship Information

Reason to Pursue the Fellowship—There are many reasons to pursue a fellowship in complex medical dermatology such as a desire to enhance exposure to the field, to practice in an academic center and develop a niche within dermatology, to practice dermatology in an inpatient setting, to improve delivery of health care to medically challenging populations in a community setting, and to become an expert on cutaneous manifestations of internal and systemic disease.

Application—There is no standardized application or deadline for this fellowship; however, there is a concerted attempt from some of the programs to offer interviews and decisions at a similar time. Deadlines and contact information are listed on the program websites, along with more details (Table).

Recommendations—I would recommend reaching out at the beginning of postgraduate year (PGY) 4 to these programs and voicing your interest in the fellowship. It is possible to set up an away rotation at some of the programs, and if your program offers elective time, pursuing an away rotation during PGY-3 or early in PGY-4 can prove to be advantageous. Furthermore, during my application cycle I toured the University of California San Francisco, University of Pennsylvania, and Brigham and Women’s Hospital to gain further insight into each program.

Brigham and Women’s Complex Medical Dermatology Fellowship

I am currently the complex medical dermatology fellow at Brigham and Women’s Hospital, and it has been an outstanding experience thus far. The program offers numerous subspecialty clinics focusing solely on cutaneous-oncodermatology, psoriasis, rheumatology-dermatology, skin of color, mole mapping backed by artificial intelligence, cosmetics, high-risk skin cancer, neutrophilic dermatoses, patch testing, phototherapy, psychodermatology, and transplant dermatology. In addition to a wide variety of subspecialty clinics, fellows have the opportunity to participate in inpatient dermatology rounds and act as a junior attending. I appreciate the flexibility of this program combined with the ability to work alongside worldwide experts. There are numerous teaching opportunities, and all of the faculty are amiable and intelligent and emphasize wellness, education, and autonomy. Overall, my experience and decision to pursue a complex medical dermatology fellowship has been extremely rewarding and invaluable. I am gaining additional skills to aid medically challenging patients while pursuing my true passion in dermatology.

References

1. Sahni DR. Inpatient dermatology consultation services in hospital institutions. Cutis. 2023;111:E11-E12. doi:10.12788/cutis.0776.

Article PDF
Author and Disclosure Information

From Brigham and Women’s Hospital, Boston, Massachusetts.

The author reports no conflict of interest.

Correspondence: Dev Ram Sahni, MD, MHA, 75 Francis St, Boston, MA 02115.

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E3-E4
Sections
Author and Disclosure Information

From Brigham and Women’s Hospital, Boston, Massachusetts.

The author reports no conflict of interest.

Correspondence: Dev Ram Sahni, MD, MHA, 75 Francis St, Boston, MA 02115.

Author and Disclosure Information

From Brigham and Women’s Hospital, Boston, Massachusetts.

The author reports no conflict of interest.

Correspondence: Dev Ram Sahni, MD, MHA, 75 Francis St, Boston, MA 02115.

Article PDF
Article PDF

Complex medical dermatology has become an emerging field in dermatology. Although a rather protean and broad term, complex medical dermatology encompasses patients with autoimmune conditions, bullous disease, connective tissue disease, vasculitis, severe dermatoses requiring immunomodulation, and inpatient consultations. Importantly, dermatology inpatient consultations aid in lowering health care costs due to accurate diagnoses, correct treatment, and decreased hospital stays.1 A fellowship is not required for holding an inpatient role in the hospital system as a dermatologist but can be beneficial. There are combined internal medicine–dermatology programs available for medical students applying to dermatology residency, but a complex medical dermatology fellowship is an option after residency for those who are interested. I believe that a focused complex medical dermatology fellowship differs from the training offered in combined internal medicine–dermatology residency. My fellow colleagues in combined internal medicine–dermatology programs are exposed to systemic manifestations of cutaneous disease and are experts in the interplay between the skin and other organ systems. However, the focus of their programs is with the intention of becoming double boarded in internal medicine and dermatology with comprehensive exposure to both fields. In my fellowship, I am able to tailor my schedule to focus on any dermatologic disease such as connective tissue disease, pruritus, graft vs host disease, and Merkel cell carcinoma. I ultimately can determine a niche in dermatology and hone my skills for a year under supervision.

Available Fellowships

Fellowship Locations—Importantly, the complex medical dermatology fellowship is not accredited by the Accreditation Council for Graduate Medical Education, which can make it difficult to identify and apply to programs. The complex medical dermatology fellowship is different than a rheumatology-dermatology fellowship, cutaneous oncology fellowship, pediatric dermatology fellowship, or other subspecialty fellowships such as those in itch or autoimmune blistering diseases. The fellowship often encompasses gaining clinical expertise in many of these conditions. I performed a thorough search online and spoke with complex medical dermatologists to compile a list of programs that offer a complex medical dermatology fellowship: Brigham and Women’s Hospital (Boston, Massachusetts); University of California San Francisco (San Francisco, California); University of Pennsylvania (Philadelphia, Pennsylvania); Cleveland Clinic (Cleveland, Ohio); and New York University (New York, New York)(Table). Only 1 spot is offered at each of these programs.

Complex Medical Dermatology Fellowship Information

Reason to Pursue the Fellowship—There are many reasons to pursue a fellowship in complex medical dermatology such as a desire to enhance exposure to the field, to practice in an academic center and develop a niche within dermatology, to practice dermatology in an inpatient setting, to improve delivery of health care to medically challenging populations in a community setting, and to become an expert on cutaneous manifestations of internal and systemic disease.

Application—There is no standardized application or deadline for this fellowship; however, there is a concerted attempt from some of the programs to offer interviews and decisions at a similar time. Deadlines and contact information are listed on the program websites, along with more details (Table).

Recommendations—I would recommend reaching out at the beginning of postgraduate year (PGY) 4 to these programs and voicing your interest in the fellowship. It is possible to set up an away rotation at some of the programs, and if your program offers elective time, pursuing an away rotation during PGY-3 or early in PGY-4 can prove to be advantageous. Furthermore, during my application cycle I toured the University of California San Francisco, University of Pennsylvania, and Brigham and Women’s Hospital to gain further insight into each program.

Brigham and Women’s Complex Medical Dermatology Fellowship

I am currently the complex medical dermatology fellow at Brigham and Women’s Hospital, and it has been an outstanding experience thus far. The program offers numerous subspecialty clinics focusing solely on cutaneous-oncodermatology, psoriasis, rheumatology-dermatology, skin of color, mole mapping backed by artificial intelligence, cosmetics, high-risk skin cancer, neutrophilic dermatoses, patch testing, phototherapy, psychodermatology, and transplant dermatology. In addition to a wide variety of subspecialty clinics, fellows have the opportunity to participate in inpatient dermatology rounds and act as a junior attending. I appreciate the flexibility of this program combined with the ability to work alongside worldwide experts. There are numerous teaching opportunities, and all of the faculty are amiable and intelligent and emphasize wellness, education, and autonomy. Overall, my experience and decision to pursue a complex medical dermatology fellowship has been extremely rewarding and invaluable. I am gaining additional skills to aid medically challenging patients while pursuing my true passion in dermatology.

Complex medical dermatology has become an emerging field in dermatology. Although a rather protean and broad term, complex medical dermatology encompasses patients with autoimmune conditions, bullous disease, connective tissue disease, vasculitis, severe dermatoses requiring immunomodulation, and inpatient consultations. Importantly, dermatology inpatient consultations aid in lowering health care costs due to accurate diagnoses, correct treatment, and decreased hospital stays.1 A fellowship is not required for holding an inpatient role in the hospital system as a dermatologist but can be beneficial. There are combined internal medicine–dermatology programs available for medical students applying to dermatology residency, but a complex medical dermatology fellowship is an option after residency for those who are interested. I believe that a focused complex medical dermatology fellowship differs from the training offered in combined internal medicine–dermatology residency. My fellow colleagues in combined internal medicine–dermatology programs are exposed to systemic manifestations of cutaneous disease and are experts in the interplay between the skin and other organ systems. However, the focus of their programs is with the intention of becoming double boarded in internal medicine and dermatology with comprehensive exposure to both fields. In my fellowship, I am able to tailor my schedule to focus on any dermatologic disease such as connective tissue disease, pruritus, graft vs host disease, and Merkel cell carcinoma. I ultimately can determine a niche in dermatology and hone my skills for a year under supervision.

Available Fellowships

Fellowship Locations—Importantly, the complex medical dermatology fellowship is not accredited by the Accreditation Council for Graduate Medical Education, which can make it difficult to identify and apply to programs. The complex medical dermatology fellowship is different than a rheumatology-dermatology fellowship, cutaneous oncology fellowship, pediatric dermatology fellowship, or other subspecialty fellowships such as those in itch or autoimmune blistering diseases. The fellowship often encompasses gaining clinical expertise in many of these conditions. I performed a thorough search online and spoke with complex medical dermatologists to compile a list of programs that offer a complex medical dermatology fellowship: Brigham and Women’s Hospital (Boston, Massachusetts); University of California San Francisco (San Francisco, California); University of Pennsylvania (Philadelphia, Pennsylvania); Cleveland Clinic (Cleveland, Ohio); and New York University (New York, New York)(Table). Only 1 spot is offered at each of these programs.

Complex Medical Dermatology Fellowship Information

Reason to Pursue the Fellowship—There are many reasons to pursue a fellowship in complex medical dermatology such as a desire to enhance exposure to the field, to practice in an academic center and develop a niche within dermatology, to practice dermatology in an inpatient setting, to improve delivery of health care to medically challenging populations in a community setting, and to become an expert on cutaneous manifestations of internal and systemic disease.

Application—There is no standardized application or deadline for this fellowship; however, there is a concerted attempt from some of the programs to offer interviews and decisions at a similar time. Deadlines and contact information are listed on the program websites, along with more details (Table).

Recommendations—I would recommend reaching out at the beginning of postgraduate year (PGY) 4 to these programs and voicing your interest in the fellowship. It is possible to set up an away rotation at some of the programs, and if your program offers elective time, pursuing an away rotation during PGY-3 or early in PGY-4 can prove to be advantageous. Furthermore, during my application cycle I toured the University of California San Francisco, University of Pennsylvania, and Brigham and Women’s Hospital to gain further insight into each program.

Brigham and Women’s Complex Medical Dermatology Fellowship

I am currently the complex medical dermatology fellow at Brigham and Women’s Hospital, and it has been an outstanding experience thus far. The program offers numerous subspecialty clinics focusing solely on cutaneous-oncodermatology, psoriasis, rheumatology-dermatology, skin of color, mole mapping backed by artificial intelligence, cosmetics, high-risk skin cancer, neutrophilic dermatoses, patch testing, phototherapy, psychodermatology, and transplant dermatology. In addition to a wide variety of subspecialty clinics, fellows have the opportunity to participate in inpatient dermatology rounds and act as a junior attending. I appreciate the flexibility of this program combined with the ability to work alongside worldwide experts. There are numerous teaching opportunities, and all of the faculty are amiable and intelligent and emphasize wellness, education, and autonomy. Overall, my experience and decision to pursue a complex medical dermatology fellowship has been extremely rewarding and invaluable. I am gaining additional skills to aid medically challenging patients while pursuing my true passion in dermatology.

References

1. Sahni DR. Inpatient dermatology consultation services in hospital institutions. Cutis. 2023;111:E11-E12. doi:10.12788/cutis.0776.

References

1. Sahni DR. Inpatient dermatology consultation services in hospital institutions. Cutis. 2023;111:E11-E12. doi:10.12788/cutis.0776.

Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E3-E4
Page Number
E3-E4
Publications
Publications
Topics
Article Type
Display Headline
Fellowships in Complex Medical Dermatology
Display Headline
Fellowships in Complex Medical Dermatology
Sections
Inside the Article

RESIDENT PEARL

  • Complex medical dermatology is a rewarding and fascinating subspecialty of dermatology, and additional training can be accomplished through a fellowship at a variety of prestigious institutions.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Painful Growing Nodule on the Right Calf

Article Type
Changed
Tue, 12/05/2023 - 16:49
Display Headline
Painful Growing Nodule on the Right Calf

The Diagnosis: Merkel Cell Carcinoma

Multiple diagnoses should be considered for a small, round, blue cell neoplasm of the skin, including both primary and metastatic entities. In our patient, histopathology revealed sheets and nests of infiltrative neoplastic cells with dispersed chromatin, minimal cytoplasm, and multiple mitoses (quiz image 1).1 The lesional cells were in the dermis and superficial subcutaneous tissue but did not appear to be arising from the epidermis. Lymphovascular invasion also was evident on additional sections. Metastatic disease was identified in 3 sentinel lymph nodes from the right inguinal and right iliac regions. These features were compatible with a diagnosis of Merkel cell carcinoma (MCC).

Merkel cell carcinoma is a rare malignant neuroendocrine cutaneous tumor with a worldwide incidence of 0.1 to 1.6 cases per 100,000 individuals annually.2 The typical patient is older than 75 years with fair skin and a history of extensive sun exposure. Immunocompromised individuals are predisposed and more susceptible to infection with the Merkel cell polyomavirus, which promotes oncogenesis in the majority of MCCs. Our patient’s history of combined variable immunodeficiency likely explains her presentation at a younger age.

The prognosis in patients with MCC is poor, with 5-year survival rates of 51% for local disease, 35% for nodal disease, and 14% for systemic metastases. Survival also is reduced in cases with head/ neck primary tumors and polyomavirus-negative tumors, as well as in immunocompromised patients.2 Treatment of resectable MCC consists of Mohs micrographic surgery or wide local excision depending on the patient’s cosmetic concerns. Radiation therapy is recommended for cases with increased risk for recurrence or positive surgical margins, as well as when additional resection is impossible. A study investigating immunotherapy with nivolumab demonstrated complete pathologic response and radiographic tumor regression in nearly half of patients when given 4 weeks prior to surgery.3

Immunohistochemistry is essential in discerning MCC from other small blue cell tumors. Most MCC cases show positive expression of neuroendocrine markers such as synaptophysin, chromogranin, and insulinomaassociated protein 1. Perinuclear dotlike staining with cytokeratin (CK) 20 (quiz image 2) commonly is seen, but up to 15% of cases may be CK20 negative. Many of these CK20-negative cases also express CK7. This tumor also may stain with paired box 5 (PAX-5), CD99, terminal deoxynucleotidyl transferase, Ber-EP4, and CD1171,4; melanoma stains (ie, human melanoma black [HMB] 45, SRYrelated HMB-box 10 [SOX-10], S-100, melanoma antigen recognized by T-cells 1 [MART-1]) should be negative. However, PAX-5 expression may be a potential pitfall given that B-cell lymphomas also would express that marker and could mimic MCC histologically. Therefore, other universal lymphoid markers such as CD45 should be ordered to rule out this entity. Even with one or a few aberrant stains, a diagnosis of MCC still can be rendered using the histomorphology and the overall staining profile.4 Of prognostic significance, p63 expression is associated with more aggressive tumors, while Bcl-2 expression is favorable, as it offers an additional targeted treatment option.5,6

Basal cell carcinoma (BCC) is linked to excessive sun exposure and is the most common skin cancer. Similar to MCC, it typically is mitotically active and hyperchromatic; however, lymphovascular invasion or metastasis almost never is observed in BCC, whereas approximately one-third of MCC cases have metastasized by the time of diagnosis. Additionally, BCC lacks the perinuclear dotlike staining seen with CK20.2,7 Features present in BCC that are unusual for MCC include peripheral nuclear palisading, mucin, and retraction artifact on paraffin-embedded sections (Figure 1).7

Basal cell carcinoma
FIGURE 1. Basal cell carcinoma. Nodular growth with classic peripheral nuclear palisading, retraction, and focal mucin (H&E, original magnification ×200).

Leukemia cutis (or cutaneous infiltrates of leukemia) commonly displays a perivascular and periadnexal pattern in the dermis and subcutis. These infiltrates of neoplastic leukocytes can congregate into sheets, sometimes with an overlying Grenz zone, or form single-file infiltrates (Figure 2).1,4 The neoplastic cells can be monomorphic or atypical and commonly are susceptible to crush artifact.4 Although the immunohistochemical profile varies depending on the etiology of the underlying leukemia, broad hematologic markers such as CD43 and CD45 are helpful to discern these malignancies from MCC.4

Leukemia cutis
FIGURE 2. Leukemia cutis. Infiltration of metastatic leukemia cells in the dermis with a single-file infiltration pattern and atypical nuclei (H&E, original magnification ×400).

Being neuroendocrine in origin, metastatic small cell carcinoma (Figure 3) strongly mimics MCC histologically and usually stains with synaptophysin, chromogranin, and insulinoma-associated protein 1. Both tumor cells typically exhibit nuclear molding and high mitotic rates. Although small cell carcinoma is more likely to stain with high-molecular-weight cytokeratins (ie, CK7), it is not uncommon for these tumors to express lowmolecular- weight cytokeratins such as CK20. Because most cases originate from the lungs, these lesions should be positive for thyroid transcription factor 1 and negative for PAX-5, whereas MCC would show the reverse for those stains.1 Ultimately, however, clinical correlation with imaging results is the single best methodology for differentiation.

Metastatic small cell carcinoma
FIGURE 3. Metastatic small cell carcinoma. Sheets of infiltrative basophilic cells with fine chromatin, nuclear molding, and brisk mitoses (H&E, original magnification ×200).

Small cell melanoma, a variant of nevoid melanoma, can strongly resemble an MCC or a lymphoma. Usually located on the scalp or arising from a congenital nevus, small cell melanomas are aggressive and confer an unfavorable prognosis. Histologically, they consist of nests to sheets of atypical cells within the epidermis and dermis. These cells typically exhibit hyperchromatic nuclei, minimal cytoplasm, and frequent mitoses (Figure 4). Furthermore, the cells do not display maturation based on depth.8 These tumors usually are positive for HMB45, S-100, MART-1, SOX-10, and tyrosinase, all of which are extremely unlikely to stain an MCC.1

Small cell melanoma
FIGURE 4. Small cell melanoma. Infiltrative nests and individual cells involving the epidermis and dermis (H&E, original magnification ×400).

References
  1. Patterson JW, Hosler GA. Weedon’s Skin Pathology. 4th ed. Churchill Livingstone/Elsevier; 2016.
  2. Walsh NM, Cerroni L. Merkel cell carcinoma: a review. J Cutan Pathol. 2021;48:411-421.
  3. Topalian SL, Bhatia S, Amin A, et al. Neoadjuvant nivolumab for patients with resectable Merkel cell carcinoma in the CheckMate 358 Trial. J Clin Oncol. 2020;38:2476-2488.
  4. Rapini RP. Practical Dermatopathology. 3rd ed. Elsevier; 2021.
  5. Asioli S, Righi A, Volante M, et al. p63 expression as a new prognostic marker in Merkel cell carcinoma. Cancer. 2007;110:640-647.
  6. Verhaegen ME, Mangelberger D, Weick JW, et al. Merkel cell carcinoma dependence on Bcl-2 family members for survival. J Invest Dermatol. 2014;134:2241-2250.
  7. Le MD, O’Steen LH, Cassarino DS. A rare case of CK20/CK7 double negative Merkel cell carcinoma. Am J Dermatopathol. 2017;39:208-211.
  8. North JP, Bastian BC, Lazar AJ. Melanoma. In: Calonje E, Brenn T, Lazar AJ, et al, eds. McKee’s Pathology of the Skin With Clinical Correlations. 5th ed. Elsevier; 2020.
Article PDF
Author and Disclosure Information

From the Department of Pathology, San Antonio Military Medical Center, Fort Sam Houston, Texas.

The authors report no conflict of interest.

The views expressed herein are those of the authors and do not reflect the official policy or position of Brooke Army Medical Center, the US Army Medical Department, the US Army Office of the Surgeon General, the Department of the Army, the Department of the Air Force, or the Department of Defense of the US Government.

Correspondence: Jesse Lee Fitzgerald, DO, San Antonio Military Medical Center, 3551 Roger Brooke Dr, Fort Sam Houston, TX 78234 (jesse.l.fitzgerald3.mil@health.mil).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
287,297-298
Sections
Author and Disclosure Information

From the Department of Pathology, San Antonio Military Medical Center, Fort Sam Houston, Texas.

The authors report no conflict of interest.

The views expressed herein are those of the authors and do not reflect the official policy or position of Brooke Army Medical Center, the US Army Medical Department, the US Army Office of the Surgeon General, the Department of the Army, the Department of the Air Force, or the Department of Defense of the US Government.

Correspondence: Jesse Lee Fitzgerald, DO, San Antonio Military Medical Center, 3551 Roger Brooke Dr, Fort Sam Houston, TX 78234 (jesse.l.fitzgerald3.mil@health.mil).

Author and Disclosure Information

From the Department of Pathology, San Antonio Military Medical Center, Fort Sam Houston, Texas.

The authors report no conflict of interest.

The views expressed herein are those of the authors and do not reflect the official policy or position of Brooke Army Medical Center, the US Army Medical Department, the US Army Office of the Surgeon General, the Department of the Army, the Department of the Air Force, or the Department of Defense of the US Government.

Correspondence: Jesse Lee Fitzgerald, DO, San Antonio Military Medical Center, 3551 Roger Brooke Dr, Fort Sam Houston, TX 78234 (jesse.l.fitzgerald3.mil@health.mil).

Article PDF
Article PDF
Related Articles

The Diagnosis: Merkel Cell Carcinoma

Multiple diagnoses should be considered for a small, round, blue cell neoplasm of the skin, including both primary and metastatic entities. In our patient, histopathology revealed sheets and nests of infiltrative neoplastic cells with dispersed chromatin, minimal cytoplasm, and multiple mitoses (quiz image 1).1 The lesional cells were in the dermis and superficial subcutaneous tissue but did not appear to be arising from the epidermis. Lymphovascular invasion also was evident on additional sections. Metastatic disease was identified in 3 sentinel lymph nodes from the right inguinal and right iliac regions. These features were compatible with a diagnosis of Merkel cell carcinoma (MCC).

Merkel cell carcinoma is a rare malignant neuroendocrine cutaneous tumor with a worldwide incidence of 0.1 to 1.6 cases per 100,000 individuals annually.2 The typical patient is older than 75 years with fair skin and a history of extensive sun exposure. Immunocompromised individuals are predisposed and more susceptible to infection with the Merkel cell polyomavirus, which promotes oncogenesis in the majority of MCCs. Our patient’s history of combined variable immunodeficiency likely explains her presentation at a younger age.

The prognosis in patients with MCC is poor, with 5-year survival rates of 51% for local disease, 35% for nodal disease, and 14% for systemic metastases. Survival also is reduced in cases with head/ neck primary tumors and polyomavirus-negative tumors, as well as in immunocompromised patients.2 Treatment of resectable MCC consists of Mohs micrographic surgery or wide local excision depending on the patient’s cosmetic concerns. Radiation therapy is recommended for cases with increased risk for recurrence or positive surgical margins, as well as when additional resection is impossible. A study investigating immunotherapy with nivolumab demonstrated complete pathologic response and radiographic tumor regression in nearly half of patients when given 4 weeks prior to surgery.3

Immunohistochemistry is essential in discerning MCC from other small blue cell tumors. Most MCC cases show positive expression of neuroendocrine markers such as synaptophysin, chromogranin, and insulinomaassociated protein 1. Perinuclear dotlike staining with cytokeratin (CK) 20 (quiz image 2) commonly is seen, but up to 15% of cases may be CK20 negative. Many of these CK20-negative cases also express CK7. This tumor also may stain with paired box 5 (PAX-5), CD99, terminal deoxynucleotidyl transferase, Ber-EP4, and CD1171,4; melanoma stains (ie, human melanoma black [HMB] 45, SRYrelated HMB-box 10 [SOX-10], S-100, melanoma antigen recognized by T-cells 1 [MART-1]) should be negative. However, PAX-5 expression may be a potential pitfall given that B-cell lymphomas also would express that marker and could mimic MCC histologically. Therefore, other universal lymphoid markers such as CD45 should be ordered to rule out this entity. Even with one or a few aberrant stains, a diagnosis of MCC still can be rendered using the histomorphology and the overall staining profile.4 Of prognostic significance, p63 expression is associated with more aggressive tumors, while Bcl-2 expression is favorable, as it offers an additional targeted treatment option.5,6

Basal cell carcinoma (BCC) is linked to excessive sun exposure and is the most common skin cancer. Similar to MCC, it typically is mitotically active and hyperchromatic; however, lymphovascular invasion or metastasis almost never is observed in BCC, whereas approximately one-third of MCC cases have metastasized by the time of diagnosis. Additionally, BCC lacks the perinuclear dotlike staining seen with CK20.2,7 Features present in BCC that are unusual for MCC include peripheral nuclear palisading, mucin, and retraction artifact on paraffin-embedded sections (Figure 1).7

Basal cell carcinoma
FIGURE 1. Basal cell carcinoma. Nodular growth with classic peripheral nuclear palisading, retraction, and focal mucin (H&E, original magnification ×200).

Leukemia cutis (or cutaneous infiltrates of leukemia) commonly displays a perivascular and periadnexal pattern in the dermis and subcutis. These infiltrates of neoplastic leukocytes can congregate into sheets, sometimes with an overlying Grenz zone, or form single-file infiltrates (Figure 2).1,4 The neoplastic cells can be monomorphic or atypical and commonly are susceptible to crush artifact.4 Although the immunohistochemical profile varies depending on the etiology of the underlying leukemia, broad hematologic markers such as CD43 and CD45 are helpful to discern these malignancies from MCC.4

Leukemia cutis
FIGURE 2. Leukemia cutis. Infiltration of metastatic leukemia cells in the dermis with a single-file infiltration pattern and atypical nuclei (H&E, original magnification ×400).

Being neuroendocrine in origin, metastatic small cell carcinoma (Figure 3) strongly mimics MCC histologically and usually stains with synaptophysin, chromogranin, and insulinoma-associated protein 1. Both tumor cells typically exhibit nuclear molding and high mitotic rates. Although small cell carcinoma is more likely to stain with high-molecular-weight cytokeratins (ie, CK7), it is not uncommon for these tumors to express lowmolecular- weight cytokeratins such as CK20. Because most cases originate from the lungs, these lesions should be positive for thyroid transcription factor 1 and negative for PAX-5, whereas MCC would show the reverse for those stains.1 Ultimately, however, clinical correlation with imaging results is the single best methodology for differentiation.

Metastatic small cell carcinoma
FIGURE 3. Metastatic small cell carcinoma. Sheets of infiltrative basophilic cells with fine chromatin, nuclear molding, and brisk mitoses (H&E, original magnification ×200).

Small cell melanoma, a variant of nevoid melanoma, can strongly resemble an MCC or a lymphoma. Usually located on the scalp or arising from a congenital nevus, small cell melanomas are aggressive and confer an unfavorable prognosis. Histologically, they consist of nests to sheets of atypical cells within the epidermis and dermis. These cells typically exhibit hyperchromatic nuclei, minimal cytoplasm, and frequent mitoses (Figure 4). Furthermore, the cells do not display maturation based on depth.8 These tumors usually are positive for HMB45, S-100, MART-1, SOX-10, and tyrosinase, all of which are extremely unlikely to stain an MCC.1

Small cell melanoma
FIGURE 4. Small cell melanoma. Infiltrative nests and individual cells involving the epidermis and dermis (H&E, original magnification ×400).

The Diagnosis: Merkel Cell Carcinoma

Multiple diagnoses should be considered for a small, round, blue cell neoplasm of the skin, including both primary and metastatic entities. In our patient, histopathology revealed sheets and nests of infiltrative neoplastic cells with dispersed chromatin, minimal cytoplasm, and multiple mitoses (quiz image 1).1 The lesional cells were in the dermis and superficial subcutaneous tissue but did not appear to be arising from the epidermis. Lymphovascular invasion also was evident on additional sections. Metastatic disease was identified in 3 sentinel lymph nodes from the right inguinal and right iliac regions. These features were compatible with a diagnosis of Merkel cell carcinoma (MCC).

Merkel cell carcinoma is a rare malignant neuroendocrine cutaneous tumor with a worldwide incidence of 0.1 to 1.6 cases per 100,000 individuals annually.2 The typical patient is older than 75 years with fair skin and a history of extensive sun exposure. Immunocompromised individuals are predisposed and more susceptible to infection with the Merkel cell polyomavirus, which promotes oncogenesis in the majority of MCCs. Our patient’s history of combined variable immunodeficiency likely explains her presentation at a younger age.

The prognosis in patients with MCC is poor, with 5-year survival rates of 51% for local disease, 35% for nodal disease, and 14% for systemic metastases. Survival also is reduced in cases with head/ neck primary tumors and polyomavirus-negative tumors, as well as in immunocompromised patients.2 Treatment of resectable MCC consists of Mohs micrographic surgery or wide local excision depending on the patient’s cosmetic concerns. Radiation therapy is recommended for cases with increased risk for recurrence or positive surgical margins, as well as when additional resection is impossible. A study investigating immunotherapy with nivolumab demonstrated complete pathologic response and radiographic tumor regression in nearly half of patients when given 4 weeks prior to surgery.3

Immunohistochemistry is essential in discerning MCC from other small blue cell tumors. Most MCC cases show positive expression of neuroendocrine markers such as synaptophysin, chromogranin, and insulinomaassociated protein 1. Perinuclear dotlike staining with cytokeratin (CK) 20 (quiz image 2) commonly is seen, but up to 15% of cases may be CK20 negative. Many of these CK20-negative cases also express CK7. This tumor also may stain with paired box 5 (PAX-5), CD99, terminal deoxynucleotidyl transferase, Ber-EP4, and CD1171,4; melanoma stains (ie, human melanoma black [HMB] 45, SRYrelated HMB-box 10 [SOX-10], S-100, melanoma antigen recognized by T-cells 1 [MART-1]) should be negative. However, PAX-5 expression may be a potential pitfall given that B-cell lymphomas also would express that marker and could mimic MCC histologically. Therefore, other universal lymphoid markers such as CD45 should be ordered to rule out this entity. Even with one or a few aberrant stains, a diagnosis of MCC still can be rendered using the histomorphology and the overall staining profile.4 Of prognostic significance, p63 expression is associated with more aggressive tumors, while Bcl-2 expression is favorable, as it offers an additional targeted treatment option.5,6

Basal cell carcinoma (BCC) is linked to excessive sun exposure and is the most common skin cancer. Similar to MCC, it typically is mitotically active and hyperchromatic; however, lymphovascular invasion or metastasis almost never is observed in BCC, whereas approximately one-third of MCC cases have metastasized by the time of diagnosis. Additionally, BCC lacks the perinuclear dotlike staining seen with CK20.2,7 Features present in BCC that are unusual for MCC include peripheral nuclear palisading, mucin, and retraction artifact on paraffin-embedded sections (Figure 1).7

Basal cell carcinoma
FIGURE 1. Basal cell carcinoma. Nodular growth with classic peripheral nuclear palisading, retraction, and focal mucin (H&E, original magnification ×200).

Leukemia cutis (or cutaneous infiltrates of leukemia) commonly displays a perivascular and periadnexal pattern in the dermis and subcutis. These infiltrates of neoplastic leukocytes can congregate into sheets, sometimes with an overlying Grenz zone, or form single-file infiltrates (Figure 2).1,4 The neoplastic cells can be monomorphic or atypical and commonly are susceptible to crush artifact.4 Although the immunohistochemical profile varies depending on the etiology of the underlying leukemia, broad hematologic markers such as CD43 and CD45 are helpful to discern these malignancies from MCC.4

Leukemia cutis
FIGURE 2. Leukemia cutis. Infiltration of metastatic leukemia cells in the dermis with a single-file infiltration pattern and atypical nuclei (H&E, original magnification ×400).

Being neuroendocrine in origin, metastatic small cell carcinoma (Figure 3) strongly mimics MCC histologically and usually stains with synaptophysin, chromogranin, and insulinoma-associated protein 1. Both tumor cells typically exhibit nuclear molding and high mitotic rates. Although small cell carcinoma is more likely to stain with high-molecular-weight cytokeratins (ie, CK7), it is not uncommon for these tumors to express lowmolecular- weight cytokeratins such as CK20. Because most cases originate from the lungs, these lesions should be positive for thyroid transcription factor 1 and negative for PAX-5, whereas MCC would show the reverse for those stains.1 Ultimately, however, clinical correlation with imaging results is the single best methodology for differentiation.

Metastatic small cell carcinoma
FIGURE 3. Metastatic small cell carcinoma. Sheets of infiltrative basophilic cells with fine chromatin, nuclear molding, and brisk mitoses (H&E, original magnification ×200).

Small cell melanoma, a variant of nevoid melanoma, can strongly resemble an MCC or a lymphoma. Usually located on the scalp or arising from a congenital nevus, small cell melanomas are aggressive and confer an unfavorable prognosis. Histologically, they consist of nests to sheets of atypical cells within the epidermis and dermis. These cells typically exhibit hyperchromatic nuclei, minimal cytoplasm, and frequent mitoses (Figure 4). Furthermore, the cells do not display maturation based on depth.8 These tumors usually are positive for HMB45, S-100, MART-1, SOX-10, and tyrosinase, all of which are extremely unlikely to stain an MCC.1

Small cell melanoma
FIGURE 4. Small cell melanoma. Infiltrative nests and individual cells involving the epidermis and dermis (H&E, original magnification ×400).

References
  1. Patterson JW, Hosler GA. Weedon’s Skin Pathology. 4th ed. Churchill Livingstone/Elsevier; 2016.
  2. Walsh NM, Cerroni L. Merkel cell carcinoma: a review. J Cutan Pathol. 2021;48:411-421.
  3. Topalian SL, Bhatia S, Amin A, et al. Neoadjuvant nivolumab for patients with resectable Merkel cell carcinoma in the CheckMate 358 Trial. J Clin Oncol. 2020;38:2476-2488.
  4. Rapini RP. Practical Dermatopathology. 3rd ed. Elsevier; 2021.
  5. Asioli S, Righi A, Volante M, et al. p63 expression as a new prognostic marker in Merkel cell carcinoma. Cancer. 2007;110:640-647.
  6. Verhaegen ME, Mangelberger D, Weick JW, et al. Merkel cell carcinoma dependence on Bcl-2 family members for survival. J Invest Dermatol. 2014;134:2241-2250.
  7. Le MD, O’Steen LH, Cassarino DS. A rare case of CK20/CK7 double negative Merkel cell carcinoma. Am J Dermatopathol. 2017;39:208-211.
  8. North JP, Bastian BC, Lazar AJ. Melanoma. In: Calonje E, Brenn T, Lazar AJ, et al, eds. McKee’s Pathology of the Skin With Clinical Correlations. 5th ed. Elsevier; 2020.
References
  1. Patterson JW, Hosler GA. Weedon’s Skin Pathology. 4th ed. Churchill Livingstone/Elsevier; 2016.
  2. Walsh NM, Cerroni L. Merkel cell carcinoma: a review. J Cutan Pathol. 2021;48:411-421.
  3. Topalian SL, Bhatia S, Amin A, et al. Neoadjuvant nivolumab for patients with resectable Merkel cell carcinoma in the CheckMate 358 Trial. J Clin Oncol. 2020;38:2476-2488.
  4. Rapini RP. Practical Dermatopathology. 3rd ed. Elsevier; 2021.
  5. Asioli S, Righi A, Volante M, et al. p63 expression as a new prognostic marker in Merkel cell carcinoma. Cancer. 2007;110:640-647.
  6. Verhaegen ME, Mangelberger D, Weick JW, et al. Merkel cell carcinoma dependence on Bcl-2 family members for survival. J Invest Dermatol. 2014;134:2241-2250.
  7. Le MD, O’Steen LH, Cassarino DS. A rare case of CK20/CK7 double negative Merkel cell carcinoma. Am J Dermatopathol. 2017;39:208-211.
  8. North JP, Bastian BC, Lazar AJ. Melanoma. In: Calonje E, Brenn T, Lazar AJ, et al, eds. McKee’s Pathology of the Skin With Clinical Correlations. 5th ed. Elsevier; 2020.
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
287,297-298
Page Number
287,297-298
Publications
Publications
Topics
Article Type
Display Headline
Painful Growing Nodule on the Right Calf
Display Headline
Painful Growing Nodule on the Right Calf
Sections
Questionnaire Body

A 47-year-old woman with a history of combined variable immunodeficiency presented with a 2.6×2.4-cm nodule on the lateral aspect of the right calf that was first noticed 2 years prior as a smaller nodule. It increased in size and became painful to touch over the last 3 to 4 months. Following diagnostic biopsy, the nodule was removed by wide local excision and was tan-brown on gross dissection. The lesion showed dotlike perinuclear positivity with cytokeratin 20 immunostaining. Positron emission tomography–computed tomography showed no evidence of lung lesions. A complete blood cell count was within reference range.

Painful growing nodule on the right calf

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 12/05/2023 - 08:00
Un-Gate On Date
Tue, 12/05/2023 - 08:00
Use ProPublica
CFC Schedule Remove Status
Tue, 12/05/2023 - 08:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media