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
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
Wed, 01/29/2025 - 13:41
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
Gating Strategy
First Page Free
Challenge Center
Disable Inline Native ads
survey writer start date
Wed, 01/29/2025 - 13:41
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

Exophytic Firm Papulonodule on the Labia in a Patient With Nonspecific Gastrointestinal Symptoms

Article Type
Changed
Tue, 01/17/2023 - 14:09
Display Headline
Exophytic Firm Papulonodule on the Labia in a Patient With Nonspecific Gastrointestinal Symptoms

The Diagnosis: Cutaneous Crohn Disease

Kinyoun and Grocott-Gomori methenamine-silver staining of the labial biopsy were negative for mycobacteria and fungi, respectively. A complete blood cell count, erythrocyte sedimentation rate, C-reactive protein, celiac disease serologies, stool occult blood, and stool calprotectin laboratory test results were within reference range. Magnetic resonance imaging of the pelvis demonstrated an anal fissure extending from the anal verge at the 6 o’clock position, abnormal T2 bright signal in the skin of the buttocks and perineum extending to the labia, and mild mucosal enhancement of the rectal and anal mucosa. Esophagogastroduodenoscopy and magnetic resonance elastography were unremarkable. Colonoscopy demonstrated scattered superficial erythematous patches and erosions in the rectum. Histologically, there was mild to moderately active colitis in the rectum with no evidence of chronicity. Given our patient’s labial edema and exophytic papulonodule (Figure 1) in the setting of nonspecific gastrointestinal symptoms and granulomatous dermatitis seen on pathology (Figure 2), she was diagnosed with cutaneous Crohn disease (CD).

Enlarged view of the patient’s right labia on initial presentation to dermatology, characterized by a flesh-colored, exophytic, firm papulonodule.
FIGURE 1. Enlarged view of the patient’s right labia on initial presentation to dermatology, characterized by a flesh-colored, exophytic, firm papulonodule.

In our patient, labial biopsy was necessary to definitively diagnose CD. Prior to biopsy of the lesion, our patient was diagnosed with irritable bowel syndrome with constipation leading to an anal fissure and skin tag due to lack of laboratory, imaging, and colonoscopy findings commonly associated with CD. Her biopsy results and gastrointestinal symptoms made these diagnoses, as well as condyloma or a large sentinel skin tag, less likely.

Histopathology of a biopsy from the right labia showed granulomatous dermatitis (H&E, original magnifications ×4 and ×20). Reference bars indicate 500 μm.
FIGURE 2. A and B, Histopathology of a biopsy from the right labia showed granulomatous dermatitis (H&E, original magnifications ×4 and ×20). Reference bars indicate 500 μm.

Extraintestinal findings of CD, especially cutaneous manifestations, are relatively frequent and may be present in as many as 44% of patients.1,2 Cutaneous CD often is characterized based on pathogenic mechanisms as either reactive, associated, or CD specific. Reactive cutaneous manifestations include erythema nodosum, pyoderma gangrenosum, and oral aphthae. Associated cutaneous manifestations include vitiligo, palmar erythema, and palmoplantar pustulosis.2 Crohn disease–specific manifestations, including genital or extragenital metastatic CD (MCD), fistulas, and oral involvement, are granulomatous in nature, similar to intestinal CD. Genital manifestations of MCD include edema, erythema, fissures, and/or ulceration of the vulva, penis, or scrotum. Labial swelling is the most common presenting symptom of MCD in females in both pediatric and adult age groups.2 Lymphedema, skin tags, and condylomalike growths also can be seen but are relatively less common.2

Given the labial edema, exophytic papulonodule, and granulomatous dermatitis seen on histopathology, our patient likely fit into the MCD category.2 In adults, most instances of MCD arise in the setting of well-established intestinal CD disease,3 whereas in children 86% of cases occur in patients without concurrent intestinal CD.2

Given the nonspecific and variable presentation of MCD, the differential diagnosis is broad. The differential diagnosis could include infectious etiologies such as condyloma acuminatum (human papillomavirus); syphilitic chancre; or mycobacterial, bacterial, fungal, or parasitic vulvovaginitis. Sexual abuse, sarcoidosis, Behçet disease, or hidradenitis suppurativa, among other diagnoses, also should be considered. Diagnostic workup should include biopsy of the lesion with special stains, polarizing microscopy, and tissue cultures.4 A thorough evaluation for gastrointestinal CD should be completed after diagnosis.3

The clinical course of vulvar CD can be unpredictable, with some cases healing spontaneously but most persisting despite treatment and sometimes prompting surgical removal.2,4 Early recognition is crucial, as long-standing MCD lesions can be therapy resistant.5 Due to the rarity of the condition and lack of data, there is a lack of treatment consensus for MCD. In 2014, the American Academy of Dermatology published treatment guidelines recommending superpotent topical steroids or topical tacrolimus as first-line therapy. Next-line therapy includes oral metronidazole, followed by prednisolone if still symptomatic.3 Treatment-resistant disease can warrant treatment with immunomodulators or tumor necrosis factor α inhibitors. Our patient was started on adalimumab; after just 2 months of therapy, the labial swelling decreased and the exophytic nodule was less firm and smaller.

Metastatic CD is a rare manifestation of cutaneous CD and can be present in the absence of gastrointestinal disease.3 This case demonstrates the importance of recognizing the cutaneous signs of CD and the necessity of lesional biopsy for the diagnosis of MCD, as our patient presented with nonspecific gastrointestinal symptoms and a diagnostic workup, including endoscopies, that proved inconclusive for the diagnosis of CD.

References
  1. Antonelli E, Bassotti G, Tramontana M, et al. Dermatological manifestations in inflammatory bowel diseases. J Clin Med. 2021;10:1-16. doi:10.3390/JCM10020364
  2. Schneider SL, Foster K, Patel D, et al. Cutaneous manifestations of metastatic Crohn’s disease. Pediatr Dermatol. 2018;35:566-574. doi:10.1111/PDE.13565
  3. Kurtzman DJB, Jones T, Lian F, et al. Metastatic Crohn’s disease: a review and approach to therapy. J Am Acad Dermatol. 2014;71:804-813. doi:10.1016/J.JAAD.2014.04.002
  4. Barret M, De Parades V, Battistella M, et al. Crohn’s disease of the vulva. J Crohns Colitis. 2014;8:563-570. doi:10.1016/J.CROHNS.2013.10.009
  5. Aberumand B, Howard J, Howard J. Metastatic Crohn’s disease: an approach to an uncommon but important cutaneous disorder [published online January 3, 2017]. Biomed Res Int. 2017;2017:8192150. doi:10.1155/2017/8192150
Article PDF
Author and Disclosure Information

Ms. Peck is from the College of Medicine, University of Cincinnati, Ohio. Dr. Mosser-Goldfarb is from the Department of Dermatology, Nationwide Children’s Hospital, The Ohio State University Wexner Medical Center, Columbus.

The authors report no conflict of interest.

Correspondence: Gabrielle M. Peck, BA, 3230 Eden Ave, Cincinnati, OH 45267 ([email protected]).

Issue
Cutis - 111(1)
Publications
Topics
Page Number
E16-E18
Sections
Author and Disclosure Information

Ms. Peck is from the College of Medicine, University of Cincinnati, Ohio. Dr. Mosser-Goldfarb is from the Department of Dermatology, Nationwide Children’s Hospital, The Ohio State University Wexner Medical Center, Columbus.

The authors report no conflict of interest.

Correspondence: Gabrielle M. Peck, BA, 3230 Eden Ave, Cincinnati, OH 45267 ([email protected]).

Author and Disclosure Information

Ms. Peck is from the College of Medicine, University of Cincinnati, Ohio. Dr. Mosser-Goldfarb is from the Department of Dermatology, Nationwide Children’s Hospital, The Ohio State University Wexner Medical Center, Columbus.

The authors report no conflict of interest.

Correspondence: Gabrielle M. Peck, BA, 3230 Eden Ave, Cincinnati, OH 45267 ([email protected]).

Article PDF
Article PDF

The Diagnosis: Cutaneous Crohn Disease

Kinyoun and Grocott-Gomori methenamine-silver staining of the labial biopsy were negative for mycobacteria and fungi, respectively. A complete blood cell count, erythrocyte sedimentation rate, C-reactive protein, celiac disease serologies, stool occult blood, and stool calprotectin laboratory test results were within reference range. Magnetic resonance imaging of the pelvis demonstrated an anal fissure extending from the anal verge at the 6 o’clock position, abnormal T2 bright signal in the skin of the buttocks and perineum extending to the labia, and mild mucosal enhancement of the rectal and anal mucosa. Esophagogastroduodenoscopy and magnetic resonance elastography were unremarkable. Colonoscopy demonstrated scattered superficial erythematous patches and erosions in the rectum. Histologically, there was mild to moderately active colitis in the rectum with no evidence of chronicity. Given our patient’s labial edema and exophytic papulonodule (Figure 1) in the setting of nonspecific gastrointestinal symptoms and granulomatous dermatitis seen on pathology (Figure 2), she was diagnosed with cutaneous Crohn disease (CD).

Enlarged view of the patient’s right labia on initial presentation to dermatology, characterized by a flesh-colored, exophytic, firm papulonodule.
FIGURE 1. Enlarged view of the patient’s right labia on initial presentation to dermatology, characterized by a flesh-colored, exophytic, firm papulonodule.

In our patient, labial biopsy was necessary to definitively diagnose CD. Prior to biopsy of the lesion, our patient was diagnosed with irritable bowel syndrome with constipation leading to an anal fissure and skin tag due to lack of laboratory, imaging, and colonoscopy findings commonly associated with CD. Her biopsy results and gastrointestinal symptoms made these diagnoses, as well as condyloma or a large sentinel skin tag, less likely.

Histopathology of a biopsy from the right labia showed granulomatous dermatitis (H&E, original magnifications ×4 and ×20). Reference bars indicate 500 μm.
FIGURE 2. A and B, Histopathology of a biopsy from the right labia showed granulomatous dermatitis (H&E, original magnifications ×4 and ×20). Reference bars indicate 500 μm.

Extraintestinal findings of CD, especially cutaneous manifestations, are relatively frequent and may be present in as many as 44% of patients.1,2 Cutaneous CD often is characterized based on pathogenic mechanisms as either reactive, associated, or CD specific. Reactive cutaneous manifestations include erythema nodosum, pyoderma gangrenosum, and oral aphthae. Associated cutaneous manifestations include vitiligo, palmar erythema, and palmoplantar pustulosis.2 Crohn disease–specific manifestations, including genital or extragenital metastatic CD (MCD), fistulas, and oral involvement, are granulomatous in nature, similar to intestinal CD. Genital manifestations of MCD include edema, erythema, fissures, and/or ulceration of the vulva, penis, or scrotum. Labial swelling is the most common presenting symptom of MCD in females in both pediatric and adult age groups.2 Lymphedema, skin tags, and condylomalike growths also can be seen but are relatively less common.2

Given the labial edema, exophytic papulonodule, and granulomatous dermatitis seen on histopathology, our patient likely fit into the MCD category.2 In adults, most instances of MCD arise in the setting of well-established intestinal CD disease,3 whereas in children 86% of cases occur in patients without concurrent intestinal CD.2

Given the nonspecific and variable presentation of MCD, the differential diagnosis is broad. The differential diagnosis could include infectious etiologies such as condyloma acuminatum (human papillomavirus); syphilitic chancre; or mycobacterial, bacterial, fungal, or parasitic vulvovaginitis. Sexual abuse, sarcoidosis, Behçet disease, or hidradenitis suppurativa, among other diagnoses, also should be considered. Diagnostic workup should include biopsy of the lesion with special stains, polarizing microscopy, and tissue cultures.4 A thorough evaluation for gastrointestinal CD should be completed after diagnosis.3

The clinical course of vulvar CD can be unpredictable, with some cases healing spontaneously but most persisting despite treatment and sometimes prompting surgical removal.2,4 Early recognition is crucial, as long-standing MCD lesions can be therapy resistant.5 Due to the rarity of the condition and lack of data, there is a lack of treatment consensus for MCD. In 2014, the American Academy of Dermatology published treatment guidelines recommending superpotent topical steroids or topical tacrolimus as first-line therapy. Next-line therapy includes oral metronidazole, followed by prednisolone if still symptomatic.3 Treatment-resistant disease can warrant treatment with immunomodulators or tumor necrosis factor α inhibitors. Our patient was started on adalimumab; after just 2 months of therapy, the labial swelling decreased and the exophytic nodule was less firm and smaller.

Metastatic CD is a rare manifestation of cutaneous CD and can be present in the absence of gastrointestinal disease.3 This case demonstrates the importance of recognizing the cutaneous signs of CD and the necessity of lesional biopsy for the diagnosis of MCD, as our patient presented with nonspecific gastrointestinal symptoms and a diagnostic workup, including endoscopies, that proved inconclusive for the diagnosis of CD.

The Diagnosis: Cutaneous Crohn Disease

Kinyoun and Grocott-Gomori methenamine-silver staining of the labial biopsy were negative for mycobacteria and fungi, respectively. A complete blood cell count, erythrocyte sedimentation rate, C-reactive protein, celiac disease serologies, stool occult blood, and stool calprotectin laboratory test results were within reference range. Magnetic resonance imaging of the pelvis demonstrated an anal fissure extending from the anal verge at the 6 o’clock position, abnormal T2 bright signal in the skin of the buttocks and perineum extending to the labia, and mild mucosal enhancement of the rectal and anal mucosa. Esophagogastroduodenoscopy and magnetic resonance elastography were unremarkable. Colonoscopy demonstrated scattered superficial erythematous patches and erosions in the rectum. Histologically, there was mild to moderately active colitis in the rectum with no evidence of chronicity. Given our patient’s labial edema and exophytic papulonodule (Figure 1) in the setting of nonspecific gastrointestinal symptoms and granulomatous dermatitis seen on pathology (Figure 2), she was diagnosed with cutaneous Crohn disease (CD).

Enlarged view of the patient’s right labia on initial presentation to dermatology, characterized by a flesh-colored, exophytic, firm papulonodule.
FIGURE 1. Enlarged view of the patient’s right labia on initial presentation to dermatology, characterized by a flesh-colored, exophytic, firm papulonodule.

In our patient, labial biopsy was necessary to definitively diagnose CD. Prior to biopsy of the lesion, our patient was diagnosed with irritable bowel syndrome with constipation leading to an anal fissure and skin tag due to lack of laboratory, imaging, and colonoscopy findings commonly associated with CD. Her biopsy results and gastrointestinal symptoms made these diagnoses, as well as condyloma or a large sentinel skin tag, less likely.

Histopathology of a biopsy from the right labia showed granulomatous dermatitis (H&E, original magnifications ×4 and ×20). Reference bars indicate 500 μm.
FIGURE 2. A and B, Histopathology of a biopsy from the right labia showed granulomatous dermatitis (H&E, original magnifications ×4 and ×20). Reference bars indicate 500 μm.

Extraintestinal findings of CD, especially cutaneous manifestations, are relatively frequent and may be present in as many as 44% of patients.1,2 Cutaneous CD often is characterized based on pathogenic mechanisms as either reactive, associated, or CD specific. Reactive cutaneous manifestations include erythema nodosum, pyoderma gangrenosum, and oral aphthae. Associated cutaneous manifestations include vitiligo, palmar erythema, and palmoplantar pustulosis.2 Crohn disease–specific manifestations, including genital or extragenital metastatic CD (MCD), fistulas, and oral involvement, are granulomatous in nature, similar to intestinal CD. Genital manifestations of MCD include edema, erythema, fissures, and/or ulceration of the vulva, penis, or scrotum. Labial swelling is the most common presenting symptom of MCD in females in both pediatric and adult age groups.2 Lymphedema, skin tags, and condylomalike growths also can be seen but are relatively less common.2

Given the labial edema, exophytic papulonodule, and granulomatous dermatitis seen on histopathology, our patient likely fit into the MCD category.2 In adults, most instances of MCD arise in the setting of well-established intestinal CD disease,3 whereas in children 86% of cases occur in patients without concurrent intestinal CD.2

Given the nonspecific and variable presentation of MCD, the differential diagnosis is broad. The differential diagnosis could include infectious etiologies such as condyloma acuminatum (human papillomavirus); syphilitic chancre; or mycobacterial, bacterial, fungal, or parasitic vulvovaginitis. Sexual abuse, sarcoidosis, Behçet disease, or hidradenitis suppurativa, among other diagnoses, also should be considered. Diagnostic workup should include biopsy of the lesion with special stains, polarizing microscopy, and tissue cultures.4 A thorough evaluation for gastrointestinal CD should be completed after diagnosis.3

The clinical course of vulvar CD can be unpredictable, with some cases healing spontaneously but most persisting despite treatment and sometimes prompting surgical removal.2,4 Early recognition is crucial, as long-standing MCD lesions can be therapy resistant.5 Due to the rarity of the condition and lack of data, there is a lack of treatment consensus for MCD. In 2014, the American Academy of Dermatology published treatment guidelines recommending superpotent topical steroids or topical tacrolimus as first-line therapy. Next-line therapy includes oral metronidazole, followed by prednisolone if still symptomatic.3 Treatment-resistant disease can warrant treatment with immunomodulators or tumor necrosis factor α inhibitors. Our patient was started on adalimumab; after just 2 months of therapy, the labial swelling decreased and the exophytic nodule was less firm and smaller.

Metastatic CD is a rare manifestation of cutaneous CD and can be present in the absence of gastrointestinal disease.3 This case demonstrates the importance of recognizing the cutaneous signs of CD and the necessity of lesional biopsy for the diagnosis of MCD, as our patient presented with nonspecific gastrointestinal symptoms and a diagnostic workup, including endoscopies, that proved inconclusive for the diagnosis of CD.

References
  1. Antonelli E, Bassotti G, Tramontana M, et al. Dermatological manifestations in inflammatory bowel diseases. J Clin Med. 2021;10:1-16. doi:10.3390/JCM10020364
  2. Schneider SL, Foster K, Patel D, et al. Cutaneous manifestations of metastatic Crohn’s disease. Pediatr Dermatol. 2018;35:566-574. doi:10.1111/PDE.13565
  3. Kurtzman DJB, Jones T, Lian F, et al. Metastatic Crohn’s disease: a review and approach to therapy. J Am Acad Dermatol. 2014;71:804-813. doi:10.1016/J.JAAD.2014.04.002
  4. Barret M, De Parades V, Battistella M, et al. Crohn’s disease of the vulva. J Crohns Colitis. 2014;8:563-570. doi:10.1016/J.CROHNS.2013.10.009
  5. Aberumand B, Howard J, Howard J. Metastatic Crohn’s disease: an approach to an uncommon but important cutaneous disorder [published online January 3, 2017]. Biomed Res Int. 2017;2017:8192150. doi:10.1155/2017/8192150
References
  1. Antonelli E, Bassotti G, Tramontana M, et al. Dermatological manifestations in inflammatory bowel diseases. J Clin Med. 2021;10:1-16. doi:10.3390/JCM10020364
  2. Schneider SL, Foster K, Patel D, et al. Cutaneous manifestations of metastatic Crohn’s disease. Pediatr Dermatol. 2018;35:566-574. doi:10.1111/PDE.13565
  3. Kurtzman DJB, Jones T, Lian F, et al. Metastatic Crohn’s disease: a review and approach to therapy. J Am Acad Dermatol. 2014;71:804-813. doi:10.1016/J.JAAD.2014.04.002
  4. Barret M, De Parades V, Battistella M, et al. Crohn’s disease of the vulva. J Crohns Colitis. 2014;8:563-570. doi:10.1016/J.CROHNS.2013.10.009
  5. Aberumand B, Howard J, Howard J. Metastatic Crohn’s disease: an approach to an uncommon but important cutaneous disorder [published online January 3, 2017]. Biomed Res Int. 2017;2017:8192150. doi:10.1155/2017/8192150
Issue
Cutis - 111(1)
Issue
Cutis - 111(1)
Page Number
E16-E18
Page Number
E16-E18
Publications
Publications
Topics
Article Type
Display Headline
Exophytic Firm Papulonodule on the Labia in a Patient With Nonspecific Gastrointestinal Symptoms
Display Headline
Exophytic Firm Papulonodule on the Labia in a Patient With Nonspecific Gastrointestinal Symptoms
Sections
Questionnaire Body

An 18-year-old woman with chronic constipation presented with an enlarging, painful, and edematous “lump” in the perineum of 1 year’s duration. The lesion became firmer and more painful with bowel movements. Physical examination revealed an enlarged right labia majora, as well as a pink to flesh-colored, exophytic, firm papulonodule in the perineum posterior to the right labia. The patient concomitantly was following with gastroenterology due to abdominal pain that worsened with eating, as well as constipation, nausea, weight loss, and rectal bleeding of 5 years’ duration. The patient denied rash, joint arthralgia, or oral ulcers. A biopsy from the labial lesion was performed.

Exophytic firm papulonodule on the labia

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 01/17/2023 - 11:30
Un-Gate On Date
Tue, 01/17/2023 - 11:30
Use ProPublica
CFC Schedule Remove Status
Tue, 01/17/2023 - 11:30
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

Methacrylate Polymer Powder Dressing for a Lower Leg Surgical Defect

Article Type
Changed
Mon, 01/16/2023 - 15:13
Display Headline
Methacrylate Polymer Powder Dressing for a Lower Leg Surgical Defect

To the Editor:

Surgical wounds on the lower leg are challenging to manage because venous stasis, bacterial colonization, and high tension may contribute to protracted healing. Advances in technology led to the development of novel, polymer-based wound-healing modalities that hold promise for the management of these wounds.

A 75-year-old man presented with a well-differentiated squamous cell carcinoma with a 3-mm depth of invasion on the left pretibial region. His comorbidities were notable for hypertension, hypercholesterolemia, varicose veins, myocardial infarction, peripheral vascular disease, and a 32 pack-year cigarette smoking history. Current medications included clopidogrel bisulfate and warfarin sodium to manage a recently placed coronary artery stent.

The tumor was cleared after 2 stages of Mohs micrographic surgery with excision down to tibialis anterior fascia (Figure 1A). The resultant defect measured 43×33 mm in area and 9 mm in depth (wound size, 12,771 mm3). Reconstructive options were discussed, including random-pattern flap repair and skin graft. Given the patient’s risk of bleeding, the decision was made to forego a flap repair. Additionally, the patient was a heavy smoker and could not comply with the wound care and elevation and ambulation restrictions required for optimal skin graft care. Therefore, a decision was made to proceed with secondary intention healing using a methacrylate polymer powder dressing.

After achieving hemostasis, a novel 10-mg sterile, biologically inert methacrylate polymer powder dressing was poured over the wound in a uniform layer to fill and seal the entire wound surface (Figure 1B). Sterile normal saline 0.1 mL was sprayed onto the powder to activate particle aggregation. No secondary dressing was used, and the patient was permitted to get the dressing wet after 48 hours.

The dressing was changed in a similar fashion 4 weeks after application, following gentle debridement with gauze and normal saline. Eight weeks after surgery, the wound exhibited healthy granulation tissue and measured 5×6 mm in area and 2 mm in depth (wound size, 60 mm3), which represented a 99.5% reduction in wound size (Figure 1C). The dressing was not painful, and there were no reported adverse effects. The patient continued to smoke and ambulate fully throughout this period. No antibiotics were used.

A, A wound on the left pretibial region following Mohs micrographic surgery. B, A methacrylate polymer powder dressing was applied to the wound. C, Eight weeks after surgery, the methacrylate polymer was no longer intact
FIGURE 1. A, A wound on the left pretibial region following Mohs micrographic surgery. B, A methacrylate polymer powder dressing was applied to the wound. C, Eight weeks after surgery, the methacrylate polymer was no longer intact, and moist wound healing was encouraged by daily cleaning with soap and water and application of liquid petroleum jelly. The wound reduced in size by 99.5%.

Methacrylate polymer powder dressings are a novel and sophisticated dressing modality with great promise for the management of surgical wounds on the lower limb. The dressing is a sterile powder consisting of 84.8% poly-2-hydroxyethylmethacrylate, 14.9% poly-2-hydroxypropylmethacrylate, and 0.3% sodium deoxycholate. These hydrophilic polymers have a covalent methacrylate backbone with a hydroxyl aliphatic side chain. When saline or wound exudate contacts the powder, the spheres hydrate and nonreversibly aggregate to form a moist, flexible dressing that conforms to the topography of the wound and seals it (Figure 2).1

A, Methacrylate polymer powder. B, Aggregation of the methacrylate polymer powder after application of normal saline medium.
FIGURE 2. A, Methacrylate polymer powder. B, Aggregation of the methacrylate polymer powder after application of normal saline medium.

Once the spheres have aggregated, they are designed to orient in a honeycomb formation with 4- to 10-nm openings that serve as capillary channels (Figure 3). This porous architecture of the polymer is essential for adequate moisture management. It allows for vapor transpiration at a rate of 12 L/m2 per day, which ensures the capillary flow from the moist wound surface is evenly distributed through the dressing, contributing to its 68% water content. Notably, this approximately three-fifths water composition is similar to the water makeup of human skin. Optimized moisture management is theorized to enhance epithelial migration, stimulate angiogenesis, retain growth factors, promote autolytic debridement, and maintain ideal voltage and oxygen gradients for wound healing. The risk for infection is not increased by the existence of these pores, as their small size does not allow for bacterial migration.1

Mechanism of methacrylate polymer powder
FIGURE 3. Mechanism of methacrylate polymer powder. When saline is added to the methacrylate polymer powder, the particles form an aggregated, organized honeycomb structure with pores 4 to 10 nm in diameter that serves as capillary channels. The small size allows for wound moisture management but does not permit bacterial transmigration. Illustration courtesy of Ni-ka Ford, MS (New York, New York).

This case demonstrates the effectiveness of using a methacrylate polymer powder dressing to promote timely wound healing in a poorly vascularized lower leg surgical wound. The low maintenance, user-friendly dressing was changed at monthly intervals, which spared the patient the inconvenience and pain associated with the repeated application of more conventional primary and secondary dressings. The dressing was well tolerated and resulted in a 99.5% reduction in wound size. Further studies are needed to investigate the utility of this promising technology.

References

1. Fitzgerald RH, Bharara M, Mills JL, et al. Use of a nanoflex powder dressing for wound management following debridement for necrotising fasciitis in the diabetic foot. Int Wound J. 2009;6:133-139.

Article PDF
Author and Disclosure Information

From the Division of Dermatologic Surgery, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York.

The authors report no conflict of interest.

Correspondence: Matthew J. Lin, MD, Division of Dermatologic Surgery, Department of Dermatology, Icahn School of Medicine at Mount Sinai, 234 E 85th St, 5th Floor, New York, NY 10028 ([email protected]).

Issue
Cutis - 111(1)
Publications
Topics
Page Number
E5-E7
Sections
Author and Disclosure Information

From the Division of Dermatologic Surgery, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York.

The authors report no conflict of interest.

Correspondence: Matthew J. Lin, MD, Division of Dermatologic Surgery, Department of Dermatology, Icahn School of Medicine at Mount Sinai, 234 E 85th St, 5th Floor, New York, NY 10028 ([email protected]).

Author and Disclosure Information

From the Division of Dermatologic Surgery, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York.

The authors report no conflict of interest.

Correspondence: Matthew J. Lin, MD, Division of Dermatologic Surgery, Department of Dermatology, Icahn School of Medicine at Mount Sinai, 234 E 85th St, 5th Floor, New York, NY 10028 ([email protected]).

Article PDF
Article PDF

To the Editor:

Surgical wounds on the lower leg are challenging to manage because venous stasis, bacterial colonization, and high tension may contribute to protracted healing. Advances in technology led to the development of novel, polymer-based wound-healing modalities that hold promise for the management of these wounds.

A 75-year-old man presented with a well-differentiated squamous cell carcinoma with a 3-mm depth of invasion on the left pretibial region. His comorbidities were notable for hypertension, hypercholesterolemia, varicose veins, myocardial infarction, peripheral vascular disease, and a 32 pack-year cigarette smoking history. Current medications included clopidogrel bisulfate and warfarin sodium to manage a recently placed coronary artery stent.

The tumor was cleared after 2 stages of Mohs micrographic surgery with excision down to tibialis anterior fascia (Figure 1A). The resultant defect measured 43×33 mm in area and 9 mm in depth (wound size, 12,771 mm3). Reconstructive options were discussed, including random-pattern flap repair and skin graft. Given the patient’s risk of bleeding, the decision was made to forego a flap repair. Additionally, the patient was a heavy smoker and could not comply with the wound care and elevation and ambulation restrictions required for optimal skin graft care. Therefore, a decision was made to proceed with secondary intention healing using a methacrylate polymer powder dressing.

After achieving hemostasis, a novel 10-mg sterile, biologically inert methacrylate polymer powder dressing was poured over the wound in a uniform layer to fill and seal the entire wound surface (Figure 1B). Sterile normal saline 0.1 mL was sprayed onto the powder to activate particle aggregation. No secondary dressing was used, and the patient was permitted to get the dressing wet after 48 hours.

The dressing was changed in a similar fashion 4 weeks after application, following gentle debridement with gauze and normal saline. Eight weeks after surgery, the wound exhibited healthy granulation tissue and measured 5×6 mm in area and 2 mm in depth (wound size, 60 mm3), which represented a 99.5% reduction in wound size (Figure 1C). The dressing was not painful, and there were no reported adverse effects. The patient continued to smoke and ambulate fully throughout this period. No antibiotics were used.

A, A wound on the left pretibial region following Mohs micrographic surgery. B, A methacrylate polymer powder dressing was applied to the wound. C, Eight weeks after surgery, the methacrylate polymer was no longer intact
FIGURE 1. A, A wound on the left pretibial region following Mohs micrographic surgery. B, A methacrylate polymer powder dressing was applied to the wound. C, Eight weeks after surgery, the methacrylate polymer was no longer intact, and moist wound healing was encouraged by daily cleaning with soap and water and application of liquid petroleum jelly. The wound reduced in size by 99.5%.

Methacrylate polymer powder dressings are a novel and sophisticated dressing modality with great promise for the management of surgical wounds on the lower limb. The dressing is a sterile powder consisting of 84.8% poly-2-hydroxyethylmethacrylate, 14.9% poly-2-hydroxypropylmethacrylate, and 0.3% sodium deoxycholate. These hydrophilic polymers have a covalent methacrylate backbone with a hydroxyl aliphatic side chain. When saline or wound exudate contacts the powder, the spheres hydrate and nonreversibly aggregate to form a moist, flexible dressing that conforms to the topography of the wound and seals it (Figure 2).1

A, Methacrylate polymer powder. B, Aggregation of the methacrylate polymer powder after application of normal saline medium.
FIGURE 2. A, Methacrylate polymer powder. B, Aggregation of the methacrylate polymer powder after application of normal saline medium.

Once the spheres have aggregated, they are designed to orient in a honeycomb formation with 4- to 10-nm openings that serve as capillary channels (Figure 3). This porous architecture of the polymer is essential for adequate moisture management. It allows for vapor transpiration at a rate of 12 L/m2 per day, which ensures the capillary flow from the moist wound surface is evenly distributed through the dressing, contributing to its 68% water content. Notably, this approximately three-fifths water composition is similar to the water makeup of human skin. Optimized moisture management is theorized to enhance epithelial migration, stimulate angiogenesis, retain growth factors, promote autolytic debridement, and maintain ideal voltage and oxygen gradients for wound healing. The risk for infection is not increased by the existence of these pores, as their small size does not allow for bacterial migration.1

Mechanism of methacrylate polymer powder
FIGURE 3. Mechanism of methacrylate polymer powder. When saline is added to the methacrylate polymer powder, the particles form an aggregated, organized honeycomb structure with pores 4 to 10 nm in diameter that serves as capillary channels. The small size allows for wound moisture management but does not permit bacterial transmigration. Illustration courtesy of Ni-ka Ford, MS (New York, New York).

This case demonstrates the effectiveness of using a methacrylate polymer powder dressing to promote timely wound healing in a poorly vascularized lower leg surgical wound. The low maintenance, user-friendly dressing was changed at monthly intervals, which spared the patient the inconvenience and pain associated with the repeated application of more conventional primary and secondary dressings. The dressing was well tolerated and resulted in a 99.5% reduction in wound size. Further studies are needed to investigate the utility of this promising technology.

To the Editor:

Surgical wounds on the lower leg are challenging to manage because venous stasis, bacterial colonization, and high tension may contribute to protracted healing. Advances in technology led to the development of novel, polymer-based wound-healing modalities that hold promise for the management of these wounds.

A 75-year-old man presented with a well-differentiated squamous cell carcinoma with a 3-mm depth of invasion on the left pretibial region. His comorbidities were notable for hypertension, hypercholesterolemia, varicose veins, myocardial infarction, peripheral vascular disease, and a 32 pack-year cigarette smoking history. Current medications included clopidogrel bisulfate and warfarin sodium to manage a recently placed coronary artery stent.

The tumor was cleared after 2 stages of Mohs micrographic surgery with excision down to tibialis anterior fascia (Figure 1A). The resultant defect measured 43×33 mm in area and 9 mm in depth (wound size, 12,771 mm3). Reconstructive options were discussed, including random-pattern flap repair and skin graft. Given the patient’s risk of bleeding, the decision was made to forego a flap repair. Additionally, the patient was a heavy smoker and could not comply with the wound care and elevation and ambulation restrictions required for optimal skin graft care. Therefore, a decision was made to proceed with secondary intention healing using a methacrylate polymer powder dressing.

After achieving hemostasis, a novel 10-mg sterile, biologically inert methacrylate polymer powder dressing was poured over the wound in a uniform layer to fill and seal the entire wound surface (Figure 1B). Sterile normal saline 0.1 mL was sprayed onto the powder to activate particle aggregation. No secondary dressing was used, and the patient was permitted to get the dressing wet after 48 hours.

The dressing was changed in a similar fashion 4 weeks after application, following gentle debridement with gauze and normal saline. Eight weeks after surgery, the wound exhibited healthy granulation tissue and measured 5×6 mm in area and 2 mm in depth (wound size, 60 mm3), which represented a 99.5% reduction in wound size (Figure 1C). The dressing was not painful, and there were no reported adverse effects. The patient continued to smoke and ambulate fully throughout this period. No antibiotics were used.

A, A wound on the left pretibial region following Mohs micrographic surgery. B, A methacrylate polymer powder dressing was applied to the wound. C, Eight weeks after surgery, the methacrylate polymer was no longer intact
FIGURE 1. A, A wound on the left pretibial region following Mohs micrographic surgery. B, A methacrylate polymer powder dressing was applied to the wound. C, Eight weeks after surgery, the methacrylate polymer was no longer intact, and moist wound healing was encouraged by daily cleaning with soap and water and application of liquid petroleum jelly. The wound reduced in size by 99.5%.

Methacrylate polymer powder dressings are a novel and sophisticated dressing modality with great promise for the management of surgical wounds on the lower limb. The dressing is a sterile powder consisting of 84.8% poly-2-hydroxyethylmethacrylate, 14.9% poly-2-hydroxypropylmethacrylate, and 0.3% sodium deoxycholate. These hydrophilic polymers have a covalent methacrylate backbone with a hydroxyl aliphatic side chain. When saline or wound exudate contacts the powder, the spheres hydrate and nonreversibly aggregate to form a moist, flexible dressing that conforms to the topography of the wound and seals it (Figure 2).1

A, Methacrylate polymer powder. B, Aggregation of the methacrylate polymer powder after application of normal saline medium.
FIGURE 2. A, Methacrylate polymer powder. B, Aggregation of the methacrylate polymer powder after application of normal saline medium.

Once the spheres have aggregated, they are designed to orient in a honeycomb formation with 4- to 10-nm openings that serve as capillary channels (Figure 3). This porous architecture of the polymer is essential for adequate moisture management. It allows for vapor transpiration at a rate of 12 L/m2 per day, which ensures the capillary flow from the moist wound surface is evenly distributed through the dressing, contributing to its 68% water content. Notably, this approximately three-fifths water composition is similar to the water makeup of human skin. Optimized moisture management is theorized to enhance epithelial migration, stimulate angiogenesis, retain growth factors, promote autolytic debridement, and maintain ideal voltage and oxygen gradients for wound healing. The risk for infection is not increased by the existence of these pores, as their small size does not allow for bacterial migration.1

Mechanism of methacrylate polymer powder
FIGURE 3. Mechanism of methacrylate polymer powder. When saline is added to the methacrylate polymer powder, the particles form an aggregated, organized honeycomb structure with pores 4 to 10 nm in diameter that serves as capillary channels. The small size allows for wound moisture management but does not permit bacterial transmigration. Illustration courtesy of Ni-ka Ford, MS (New York, New York).

This case demonstrates the effectiveness of using a methacrylate polymer powder dressing to promote timely wound healing in a poorly vascularized lower leg surgical wound. The low maintenance, user-friendly dressing was changed at monthly intervals, which spared the patient the inconvenience and pain associated with the repeated application of more conventional primary and secondary dressings. The dressing was well tolerated and resulted in a 99.5% reduction in wound size. Further studies are needed to investigate the utility of this promising technology.

References

1. Fitzgerald RH, Bharara M, Mills JL, et al. Use of a nanoflex powder dressing for wound management following debridement for necrotising fasciitis in the diabetic foot. Int Wound J. 2009;6:133-139.

References

1. Fitzgerald RH, Bharara M, Mills JL, et al. Use of a nanoflex powder dressing for wound management following debridement for necrotising fasciitis in the diabetic foot. Int Wound J. 2009;6:133-139.

Issue
Cutis - 111(1)
Issue
Cutis - 111(1)
Page Number
E5-E7
Page Number
E5-E7
Publications
Publications
Topics
Article Type
Display Headline
Methacrylate Polymer Powder Dressing for a Lower Leg Surgical Defect
Display Headline
Methacrylate Polymer Powder Dressing for a Lower Leg Surgical Defect
Sections
Inside the Article

PRACTICE POINTS

  • Lower leg surgical wounds are difficult to manage, as venous stasis, bacterial colonization, and high tension may contribute to protracted healing.
  • A methacrylate polymer powder dressing is user friendly and facilitates granulation and reduction in size of difficult lower leg wounds.
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

Insights From the 2020-2021 Dermatology Residency Match

Article Type
Changed
Wed, 01/11/2023 - 10:59
Display Headline
Insights From the 2020-2021 Dermatology Residency Match

To the Editor:

Data from the program director survey of the National Resident Matching Program offer key insights into the 2021 dermatology application process.1,2 Examination of data from the 2020 (N=12) and 2021 (N=17) program director survey regarding interviewing applicants revealed that specialty-specific letters of recommendation (LORs), personal prior knowledge of an applicant, and personal statement increased in importance by 17%, 7.4%, and 17%, respectively, whereas away rotations within the department decreased in importance by 44.9% (Table).1,2 Interestingly, for ranking applicants, programs decreased their emphasis on specialty-specific LORs by 25.8% and away rotations within the department by 22.7% and increased emphasis on personal statements by 14.7% and personal prior knowledge of an applicant by 0.8% from 2020 to 2021 (Table).1,2 These findings align with the prior recommendation to limit away rotations; data are contradictory—when comparing factors for interviewing as compared to ranking applicants—for specialty-specific LORs.

Comparison of Interviewing and Ranking Factors for the Selection of Dermatology Residency Applicants

We further compared data from the otolaryngology cycle, which implemented preference signaling by which an applicant can signal their interest in a particular residency program in the 2021 Match, to data from dermatology with no preference signaling. A 90% probability of matching is estimated to require approximately 8 or 9 interviews for dermatology or 12 interviews for otolaryngology for MD senior students in 2020.4 In prior dermatology application cycles, the most highly qualified candidates constituted 7% to 21% of all applicants but were estimated to receive half of all interviews, causing a maldistribution of interviews.5,6

For the 2021 otolaryngology match, the Society of University Otolaryngologists implemented a novel preference signaling system that allowed candidates to show interest in programs by sending 5 preferences, or tokens.7 Recent data reports from the otolaryngology cycle demonstrated at least a 2-fold increase in the rate of receiving an interview invitation for signaled programs compared to the closest nonsignaled program if applicants were provided an additional token.7 Regarding overall applicant competitiveness (ie, dividing participants into quartiles based on their competitiveness), the highest increase in the overall rate of interview invitations (3.5 [total invitations/total applications]) was demonstrated for fourth-quartile (ie, “lowest quartile”) applicants compared with the increase in the overall rate of interview invitations seen in other quartiles (first quartile, an increase of 2.3; second quartile, an increase of 2.6; and third quartile, an increase of 2.4).7 We look forward to seeing the impact of preference signaling on the results of the 2022 dermatology cycle.

Despite changes in the interviewing process to accommodate COVID-19 pandemic safety recommendations, the overall dermatology postgraduate year (PGY) 2 fill rate remained unchanged from 2018 (98.6%) to 2021 (98.7%). Zero PGY-1 positions and 5 PGY-2 positions were unfilled in the 2021 Main Residency Match compared to 1 unfilled PGY-1 position and 4 unfilled PGY-2 positions in 2018.8 The coordinated interview invitation release, holistic review of applications, increased number of rankings, and virtual interviews might have helped offset potential obstacles imparted by inability to complete away rotations, inability to obtain LORs, and conducting interviews virtually.5

A limitation of our analysis is the low response rate of program directors to National Resident Matching Program surveys.

These strategies—holistic application review and coordinated interview release—may be considered in future cycles given their convenience and negligible impact on the dermatology match rate. For example, virtual interviews relieve the financial and time burdens of in-person interviews—approximately $10,000 for each US senior applicant—thus potentially allowing for a more equitable matching process.3 Inversely, in-person interviews allow participants to effectively network and form more meaningful connections while obtaining a better understanding of facilities and surrounding locales. As such, the medical community should continue to come to a consensus on the optimal format to host interviews.

References
  1. Results of the 2021 NRMP Program Director Survey. National Resident Matching Program. August 2021. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2021/11/2021-PD-Survey-Report-for-WWW.pdf
  2. Results of the 2020 NRMP Program Director Survey. National Resident Matching Program. August 2020. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2022/01/2020-PD-Survey.pdf
  3. Rojek NW, Shinkai K, Fett N. Dermatology faculty and residents’ perspectives on the dermatology residency application process: a nationwide survey. J Am Acad Dermatol. 2018;79:157-159. doi:10.1016/j.jaad.2018.01.00
  4. Charting Outcomes in the Match: Senior Students of U.S. MD Medical Schools. National Resident Matching Program. July 2020. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2021/08/Charting-Outcomes-in-the-Match-2020_MD-Senior_final.pdf
  5. Thatiparthi A, Martin A, Liu J, et al. Preliminary outcomes of 2020-2021 dermatology residency application cycle and adverse effects of COVID-19. J Am Acad Dermatol. 2021;84:e263-e264. doi:10.1016/j.jaad.2021.03.034
  6. Hammoud MM, Standiford T, Carmody JB. Potential implications of COVID-19 for the 2020-2021 residency application cycle. JAMA. 2020;324:29-30. doi:10.1001/jama.2020.8911
  7. Interview offer rate with/without ENTSignaling. Society of University Otolaryngologists. Updated July 19, 2022. Accessed December 12, 2022. https://opdo-hns.org/mpage/signaling-updates
  8. Results and Data: 2021 Main Residency Match. National Resident Matching Program. May 2021. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2021/08/MRM-Results_and-Data_2021.pdf
Article PDF
Author and Disclosure Information

Dr. Thatiparthi is from the College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California. Dr. Martin is from the University of California, Riverside School of Medicine, Riverside. Dr. Wu is from the Department of Dermatology, University of Miami Leonard M. Miller School of Medicine, Florida.

Drs. Thatiparthi and Martin report no conflict of interest. Dr. Wu is or has been an investigator, consultant, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly & Company, EPI Health, Galderma, Janssen, LEO Pharma, Mindera, Novartis, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceutical, UCB, and Zerigo Health. He also has received research grants from AbbVie, Amgen, Eli Lilly & Company, Janssen, Novartis, and Pfizer Inc.

Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 ([email protected]).

Issue
Cutis - 110(6)
Publications
Topics
Page Number
E22-E24
Sections
Author and Disclosure Information

Dr. Thatiparthi is from the College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California. Dr. Martin is from the University of California, Riverside School of Medicine, Riverside. Dr. Wu is from the Department of Dermatology, University of Miami Leonard M. Miller School of Medicine, Florida.

Drs. Thatiparthi and Martin report no conflict of interest. Dr. Wu is or has been an investigator, consultant, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly & Company, EPI Health, Galderma, Janssen, LEO Pharma, Mindera, Novartis, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceutical, UCB, and Zerigo Health. He also has received research grants from AbbVie, Amgen, Eli Lilly & Company, Janssen, Novartis, and Pfizer Inc.

Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 ([email protected]).

Author and Disclosure Information

Dr. Thatiparthi is from the College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California. Dr. Martin is from the University of California, Riverside School of Medicine, Riverside. Dr. Wu is from the Department of Dermatology, University of Miami Leonard M. Miller School of Medicine, Florida.

Drs. Thatiparthi and Martin report no conflict of interest. Dr. Wu is or has been an investigator, consultant, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly & Company, EPI Health, Galderma, Janssen, LEO Pharma, Mindera, Novartis, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceutical, UCB, and Zerigo Health. He also has received research grants from AbbVie, Amgen, Eli Lilly & Company, Janssen, Novartis, and Pfizer Inc.

Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 ([email protected]).

Article PDF
Article PDF

To the Editor:

Data from the program director survey of the National Resident Matching Program offer key insights into the 2021 dermatology application process.1,2 Examination of data from the 2020 (N=12) and 2021 (N=17) program director survey regarding interviewing applicants revealed that specialty-specific letters of recommendation (LORs), personal prior knowledge of an applicant, and personal statement increased in importance by 17%, 7.4%, and 17%, respectively, whereas away rotations within the department decreased in importance by 44.9% (Table).1,2 Interestingly, for ranking applicants, programs decreased their emphasis on specialty-specific LORs by 25.8% and away rotations within the department by 22.7% and increased emphasis on personal statements by 14.7% and personal prior knowledge of an applicant by 0.8% from 2020 to 2021 (Table).1,2 These findings align with the prior recommendation to limit away rotations; data are contradictory—when comparing factors for interviewing as compared to ranking applicants—for specialty-specific LORs.

Comparison of Interviewing and Ranking Factors for the Selection of Dermatology Residency Applicants

We further compared data from the otolaryngology cycle, which implemented preference signaling by which an applicant can signal their interest in a particular residency program in the 2021 Match, to data from dermatology with no preference signaling. A 90% probability of matching is estimated to require approximately 8 or 9 interviews for dermatology or 12 interviews for otolaryngology for MD senior students in 2020.4 In prior dermatology application cycles, the most highly qualified candidates constituted 7% to 21% of all applicants but were estimated to receive half of all interviews, causing a maldistribution of interviews.5,6

For the 2021 otolaryngology match, the Society of University Otolaryngologists implemented a novel preference signaling system that allowed candidates to show interest in programs by sending 5 preferences, or tokens.7 Recent data reports from the otolaryngology cycle demonstrated at least a 2-fold increase in the rate of receiving an interview invitation for signaled programs compared to the closest nonsignaled program if applicants were provided an additional token.7 Regarding overall applicant competitiveness (ie, dividing participants into quartiles based on their competitiveness), the highest increase in the overall rate of interview invitations (3.5 [total invitations/total applications]) was demonstrated for fourth-quartile (ie, “lowest quartile”) applicants compared with the increase in the overall rate of interview invitations seen in other quartiles (first quartile, an increase of 2.3; second quartile, an increase of 2.6; and third quartile, an increase of 2.4).7 We look forward to seeing the impact of preference signaling on the results of the 2022 dermatology cycle.

Despite changes in the interviewing process to accommodate COVID-19 pandemic safety recommendations, the overall dermatology postgraduate year (PGY) 2 fill rate remained unchanged from 2018 (98.6%) to 2021 (98.7%). Zero PGY-1 positions and 5 PGY-2 positions were unfilled in the 2021 Main Residency Match compared to 1 unfilled PGY-1 position and 4 unfilled PGY-2 positions in 2018.8 The coordinated interview invitation release, holistic review of applications, increased number of rankings, and virtual interviews might have helped offset potential obstacles imparted by inability to complete away rotations, inability to obtain LORs, and conducting interviews virtually.5

A limitation of our analysis is the low response rate of program directors to National Resident Matching Program surveys.

These strategies—holistic application review and coordinated interview release—may be considered in future cycles given their convenience and negligible impact on the dermatology match rate. For example, virtual interviews relieve the financial and time burdens of in-person interviews—approximately $10,000 for each US senior applicant—thus potentially allowing for a more equitable matching process.3 Inversely, in-person interviews allow participants to effectively network and form more meaningful connections while obtaining a better understanding of facilities and surrounding locales. As such, the medical community should continue to come to a consensus on the optimal format to host interviews.

To the Editor:

Data from the program director survey of the National Resident Matching Program offer key insights into the 2021 dermatology application process.1,2 Examination of data from the 2020 (N=12) and 2021 (N=17) program director survey regarding interviewing applicants revealed that specialty-specific letters of recommendation (LORs), personal prior knowledge of an applicant, and personal statement increased in importance by 17%, 7.4%, and 17%, respectively, whereas away rotations within the department decreased in importance by 44.9% (Table).1,2 Interestingly, for ranking applicants, programs decreased their emphasis on specialty-specific LORs by 25.8% and away rotations within the department by 22.7% and increased emphasis on personal statements by 14.7% and personal prior knowledge of an applicant by 0.8% from 2020 to 2021 (Table).1,2 These findings align with the prior recommendation to limit away rotations; data are contradictory—when comparing factors for interviewing as compared to ranking applicants—for specialty-specific LORs.

Comparison of Interviewing and Ranking Factors for the Selection of Dermatology Residency Applicants

We further compared data from the otolaryngology cycle, which implemented preference signaling by which an applicant can signal their interest in a particular residency program in the 2021 Match, to data from dermatology with no preference signaling. A 90% probability of matching is estimated to require approximately 8 or 9 interviews for dermatology or 12 interviews for otolaryngology for MD senior students in 2020.4 In prior dermatology application cycles, the most highly qualified candidates constituted 7% to 21% of all applicants but were estimated to receive half of all interviews, causing a maldistribution of interviews.5,6

For the 2021 otolaryngology match, the Society of University Otolaryngologists implemented a novel preference signaling system that allowed candidates to show interest in programs by sending 5 preferences, or tokens.7 Recent data reports from the otolaryngology cycle demonstrated at least a 2-fold increase in the rate of receiving an interview invitation for signaled programs compared to the closest nonsignaled program if applicants were provided an additional token.7 Regarding overall applicant competitiveness (ie, dividing participants into quartiles based on their competitiveness), the highest increase in the overall rate of interview invitations (3.5 [total invitations/total applications]) was demonstrated for fourth-quartile (ie, “lowest quartile”) applicants compared with the increase in the overall rate of interview invitations seen in other quartiles (first quartile, an increase of 2.3; second quartile, an increase of 2.6; and third quartile, an increase of 2.4).7 We look forward to seeing the impact of preference signaling on the results of the 2022 dermatology cycle.

Despite changes in the interviewing process to accommodate COVID-19 pandemic safety recommendations, the overall dermatology postgraduate year (PGY) 2 fill rate remained unchanged from 2018 (98.6%) to 2021 (98.7%). Zero PGY-1 positions and 5 PGY-2 positions were unfilled in the 2021 Main Residency Match compared to 1 unfilled PGY-1 position and 4 unfilled PGY-2 positions in 2018.8 The coordinated interview invitation release, holistic review of applications, increased number of rankings, and virtual interviews might have helped offset potential obstacles imparted by inability to complete away rotations, inability to obtain LORs, and conducting interviews virtually.5

A limitation of our analysis is the low response rate of program directors to National Resident Matching Program surveys.

These strategies—holistic application review and coordinated interview release—may be considered in future cycles given their convenience and negligible impact on the dermatology match rate. For example, virtual interviews relieve the financial and time burdens of in-person interviews—approximately $10,000 for each US senior applicant—thus potentially allowing for a more equitable matching process.3 Inversely, in-person interviews allow participants to effectively network and form more meaningful connections while obtaining a better understanding of facilities and surrounding locales. As such, the medical community should continue to come to a consensus on the optimal format to host interviews.

References
  1. Results of the 2021 NRMP Program Director Survey. National Resident Matching Program. August 2021. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2021/11/2021-PD-Survey-Report-for-WWW.pdf
  2. Results of the 2020 NRMP Program Director Survey. National Resident Matching Program. August 2020. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2022/01/2020-PD-Survey.pdf
  3. Rojek NW, Shinkai K, Fett N. Dermatology faculty and residents’ perspectives on the dermatology residency application process: a nationwide survey. J Am Acad Dermatol. 2018;79:157-159. doi:10.1016/j.jaad.2018.01.00
  4. Charting Outcomes in the Match: Senior Students of U.S. MD Medical Schools. National Resident Matching Program. July 2020. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2021/08/Charting-Outcomes-in-the-Match-2020_MD-Senior_final.pdf
  5. Thatiparthi A, Martin A, Liu J, et al. Preliminary outcomes of 2020-2021 dermatology residency application cycle and adverse effects of COVID-19. J Am Acad Dermatol. 2021;84:e263-e264. doi:10.1016/j.jaad.2021.03.034
  6. Hammoud MM, Standiford T, Carmody JB. Potential implications of COVID-19 for the 2020-2021 residency application cycle. JAMA. 2020;324:29-30. doi:10.1001/jama.2020.8911
  7. Interview offer rate with/without ENTSignaling. Society of University Otolaryngologists. Updated July 19, 2022. Accessed December 12, 2022. https://opdo-hns.org/mpage/signaling-updates
  8. Results and Data: 2021 Main Residency Match. National Resident Matching Program. May 2021. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2021/08/MRM-Results_and-Data_2021.pdf
References
  1. Results of the 2021 NRMP Program Director Survey. National Resident Matching Program. August 2021. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2021/11/2021-PD-Survey-Report-for-WWW.pdf
  2. Results of the 2020 NRMP Program Director Survey. National Resident Matching Program. August 2020. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2022/01/2020-PD-Survey.pdf
  3. Rojek NW, Shinkai K, Fett N. Dermatology faculty and residents’ perspectives on the dermatology residency application process: a nationwide survey. J Am Acad Dermatol. 2018;79:157-159. doi:10.1016/j.jaad.2018.01.00
  4. Charting Outcomes in the Match: Senior Students of U.S. MD Medical Schools. National Resident Matching Program. July 2020. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2021/08/Charting-Outcomes-in-the-Match-2020_MD-Senior_final.pdf
  5. Thatiparthi A, Martin A, Liu J, et al. Preliminary outcomes of 2020-2021 dermatology residency application cycle and adverse effects of COVID-19. J Am Acad Dermatol. 2021;84:e263-e264. doi:10.1016/j.jaad.2021.03.034
  6. Hammoud MM, Standiford T, Carmody JB. Potential implications of COVID-19 for the 2020-2021 residency application cycle. JAMA. 2020;324:29-30. doi:10.1001/jama.2020.8911
  7. Interview offer rate with/without ENTSignaling. Society of University Otolaryngologists. Updated July 19, 2022. Accessed December 12, 2022. https://opdo-hns.org/mpage/signaling-updates
  8. Results and Data: 2021 Main Residency Match. National Resident Matching Program. May 2021. Accessed December 6, 2021. https://www.nrmp.org/wp-content/uploads/2021/08/MRM-Results_and-Data_2021.pdf
Issue
Cutis - 110(6)
Issue
Cutis - 110(6)
Page Number
E22-E24
Page Number
E22-E24
Publications
Publications
Topics
Article Type
Display Headline
Insights From the 2020-2021 Dermatology Residency Match
Display Headline
Insights From the 2020-2021 Dermatology Residency Match
Sections
Inside the Article

PRACTICE POINTS

  • Although there have been numerous changes to the dermatology interview process due to the COVID-19 pandemic, the overall fill rate for postgraduate year 2 positions remained unchanged from 2018 (prepandemic) to 2021 (postpandemic).
  • Strategies to accommodate new safety recommendations for interviews may reduce the financial burden (approximately $10,000 for each senior applicant) and time constraints on applicants. These strategies should be considered for implementation in future cycles.
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

Oral Propranolol Used as Adjunct Therapy in Cutaneous Angiosarcoma

Article Type
Changed
Thu, 01/05/2023 - 09:15
Display Headline
Oral Propranolol Used as Adjunct Therapy in Cutaneous Angiosarcoma

To the Editor:

Angiosarcoma is a malignancy of the vascular endothelium that most commonly presents on the skin.1 Patients diagnosed with cutaneous angiosarcoma, which is a rare and aggressive malignancy, have a 5-year survival rate of approximately 30%.2,3 Angiosarcoma can be seen in the setting of chronic lymphedema; radiation therapy; and sporadically in elderly patients, where it is commonly seen on the head and neck. Presentation on the head and neck has been associated with worse outcomes, with a projected overall 10-year survival rate of 13.8%; the survival rate is lower if the tumor is surgically unresectable or larger in size. Metastasis can occur via both lymphatic and hematogenous routes, with pulmonary and hepatic metastases most frequently observed.1 Prognostications of poor outcomes for patients with head and neck cutaneous angiosarcoma via a 5-year survival rate were identified in a meta-analysis and included the following: patient age older than 70 years, larger tumors, tumor location of scalp vs face, nonsurgical treatments, and lack of clear margins on histology.2

Treatment of angiosarcoma historically has encompassed both surgical resection and adjuvant radiation therapy with suboptimal success. Evidence supporting various treatment regimens remains sparse due to the low incidence of the neoplasm. Although surgical resection is the only documented curative treatment, cutaneous angiosarcomas frequently are found to have positive surgical margins and require adjuvant radiation. Use of high-dose radiation (>50 Gy) with application over a wide treatment area such as total scalp irradiation is recommended.4 Although radiation has been found to diminish local recurrence rates, it has not substantially affected rates of distant disease recurrence.1 Cytotoxic chemotherapy has clinical utility in minimizing progression, but standard regimens afford a progression-free survival of only months.3 Adjuvant treatment with paclitaxel has been shown to have improved efficacy in scalp angiosarcoma vs other visceral sites, showing a nonprogression rate of 42% at 4 months after treatment.5 More recently, targeted chemotherapeutics, including the vascular endothelial growth factor inhibitor bevacizumab and tyrosine kinase inhibitor sorafenib, have shown some survival benefit, but it is unclear if these agents are superior to traditional cytotoxic agents.4,6-10 A phase 2 study of paclitaxel administered weekly with or without bevacizumab showed similar progression-free survival and overall survival, albeit at the expense of added toxicity experienced by participants in the combined group.10

The addition of the nonselective β-adrenergic blocker propranolol to the treatment armamentarium, which was pursued due to its utility in the treatment of benign infantile hemangioma and demonstrated ability to limit the expression of adrenergic receptors in angiosarcoma, has gained clinical attention for possible augmentation of cutaneous angiosarcoma therapy.11-14 Propranolol has been shown to reduce metastasis in other neoplasms—both vascular and nonvascular—and may play a role as an adjuvant treatment to current therapies in angiosarcoma.15-20 We report a patient with cutaneous angiosarcoma (T2 classification) with disease-free survival of nearly 6 years without evidence of recurrence in the setting of continuous propranolol use supplementary to chemotherapy and radiation.

Cutaneous angiosarcoma at the time of diagnosis.
FIGURE 1. Cutaneous angiosarcoma at the time of diagnosis. A, An ecchymotic patch showed extensive involvement of right scalp, forehead, and temple. B, Extension of the ecchymotic patch on the left side of the face.

A 78-year-old man with a history of multiple basal cell carcinomas, hypertension, and remote smoking history presented to the dermatology clinic with an enlarging red-brown plaque on the scalp of 2 months’ duration. The lesion had grown rapidly to involve the forehead, right temple, preauricular region, and parietal scalp. At presentation, the tumor measured more than 20 cm in diameter at its greatest point (Figure 1). Physical examination revealed a 6-mm purple nodule within the lesion on the patient’s right parietal scalp. No clinical lymphadenopathy was appreciated at the time of diagnosis. Punch biopsies of the right parietal scalp nodule and right temple patch showed findings consistent with angiosarcoma with diffuse cytoplasmic staining of CD31 in atypical endothelial cells and no staining for human herpesvirus 8 (Figure 2). Concurrent computed tomography of the head showed thickening of the right epidermis, dermis, and deeper scalp tissues, but there was no evidence of skull involvement. Computed tomography of the thorax, abdomen, and pelvis showed no evidence of metastatic disease. After a diagnostic workup, the patient was diagnosed with T2bN0M0 angiosarcoma.

A, A punch biopsy of the right parietal scalp showed cytologically atypical endothelial cells forming slitlike vascular spaces in the dermis (H&E, original magnification ×100). B, Cytoplasmic CD31 staining of endothelial lining of slit-like atypical
FIGURE 2. A, A punch biopsy of the right parietal scalp showed cytologically atypical endothelial cells forming slitlike vascular spaces in the dermis (H&E, original magnification ×100). B, Cytoplasmic CD31 staining of endothelial lining of slit-like atypical vascular spaces (original magnification ×400).

The lesion was determined to be nonresectable due to the extent of the patient’s cutaneous disease. The patient was started on a regimen of paclitaxel, scalp radiation, and oral propranolol. Propranolol 40 mg twice daily was initiated at the time of diagnosis with a plan to continue indefinitely. Starting 1 month after staging, the patient completed 10 weekly cycles of paclitaxel, and he was treated with 60 Gy of scalp radiation in 30 fractions, starting with the second cycle of paclitaxel. He tolerated both well with no reported adverse events. Repeat computed tomography performed 1 month after completion of chemotherapy and radiation showed no evidence of a mass or fluid collection in subcutaneous scalp tissues and no evidence of metastatic disease. This correlated with an observed clinical regression at 1 month and complete clinical response at 5 months with residual hemosiderin and radiation changes. The area of prior disease involvement subsequently evolved from violet to dusky gray in appearance to an eventual complete resolution 26 months after diagnosis, accompanied by atrophic radiation-induced sequelae (Figure 3).

No clinical evidence of disease 8 months after initial diagnosis following treatment with radiation therapy and adjunctive propranolol 40 mg twice daily. Only postinflammatory pigment change remained A B on examination.
FIGURE 3. A and B, No clinical evidence of disease 8 months after initial diagnosis following treatment with radiation therapy and adjunctive propranolol 40 mg twice daily. Only postinflammatory pigment change remained A B on examination.

The patient’s postchemotherapy course was complicated by hospitalization for a suspected malignant pleural effusion. Analysis revealed growing ground-glass opacities and nodules in the right lower lung lobe. A thoracentesis with cytology studies was negative for malignancy. Continued monitoring over 19 months demonstrated eventual resolution of those findings. He experienced notable complication from local radiation therapy to the scalp with chronic cutaneous ulceration refractory to wound care and surgical intervention. The patient did not exhibit additional signs or symptoms concerning for recurrence or metastasis and was followed by dermatology and oncology until he died nearly 5 years after initial diagnosis due to complications from acute hypoxic respiratory failure secondary to COVID-19. The last imaging obtained showed no convincing evidence of metastasis, though spinal imaging within a month of his death showed lesions favored to represent benign angiomatous growths. His survival after diagnosis ultimately reached 57 months without confirmed disease recurrence and cause of death unrelated to malignancy history, which is a markedly long documented survival for this extent of disease.

Cutaneous angiosarcoma is an aggressive yet rare malignancy without effective treatments for prolonging survival or eradicating disease. Cutaneous angiosarcoma of the head and neck has a reported 10-year survival rate of 13.8%.1 Although angiosarcoma in any location holds a bleak prognosis, cutaneous angiosarcoma of the scalp with a T2 classification has a 2-year survival rate of 0%. Moreover, even if remission is achieved, disease is highly recurrent, typically within months with the current standard of care.3,21,22

Emerging evidence for the possible role of β-adrenergic receptor blockade in the treatment of malignant vascular neoplasms is promising. Microarrays from a host of vascular growths have demonstrated expression of β-adrenergic receptors in 77% of sampled angiosarcoma specimens in addition to strong expression in infantile hemangiomas, hemangiomas, hemangioendotheliomas, and vascular malformations.19 Research findings have further verified the validity of this approach with the demonstration of b1-, b2-, and b3- adrenergic receptor expression by angiosarcoma cell lines. Propranolol subsequently was shown to effectively target proliferation of these cells and induce apoptosis in a dose-dependent manner and moreover be synergistic in effect with other chemotherapies.15 Several genes have exhibited differential expression between control tumor cells and propranolol-treated cells. Specifically, target genes including AXL (a receptor tyrosine kinase associated with cell adhesion, proliferation, and apoptosis and found to upregulated in melanoma and leukemia) and ERBB receptor feedback inhibitor 1 (receptor tyrosine kinase, with ERBB family members commonly overexpressed or mutated in the setting malignancy) have been posited as possible explanatory factors in the observed angiosarcoma response to propranolol.23

Several cases describing propranolol use as an adjunctive therapy for angiosarcoma suggest a beneficial role in clinical medicine. One case report described propranolol monotherapy for lesion to our patient, with a resultant reduction in Ki-67 as a measure of proliferative index within 1 week of initiating propranolol therapy.13 Propranolol also has been shown to halt or slow progression of metastatic disease in visceral and metastatic angiosarcomas.12-14 In combination with oral etoposide and cyclophosphamide, maintenance propranolol therapy in 7 cases of advanced cutaneous angiosarcoma resulted in 1 complete response and 3 very good partial responses, with a median progression-free survival of 11 months.11 Larger-scale studies have not been published, but the growing number of case reports and case series warrants further investigation of the utility of propranolol as an adjunct to current therapies in advanced angiosarcoma.

References
  1. Abraham JA, Hornicek FJ, Kaufman AM, et al. Treatment and outcome of 82 patients with angiosarcoma. Ann Surg Oncol. 2007;14:1953-1967.
  2. Shin JY, Roh SG, Lee NH, et al. Predisposing factors for poor prognosis of angiosarcoma of the scalp and face: systematic review and meta-analysis. Head Neck. 2017;39:380-386.
  3. Fury MG, Antonescu CR, Zee KJV, et al. A 14-year retrospective review of angiosarcoma: clinical characteristics, prognostic factors, and treatment outcomes with surgery and chemotherapy. Cancer. 2005;11:241-247.
  4. Dossett LA, Harrington M, Cruse CW, et al. Cutaneous angiosarcoma. Curr Probl Cancer. 2015;39:258-263.
  5. Penel N, Bui BN, Bay JO, et al. Phase II trial of weekly paclitaxel for unresectable angiosarcoma: the ANGIOTAX study. J Clin Oncol. 2008;26:5269-5274.
  6. Agulnik M, Yarber JL, Okuno SH, et al. An open-label, multicenter, phase II study of bevacizumab for the treatment of angiosarcoma and epithelioid hemangioendotheliomas. Ann Oncol. 2013;24:257-263.
  7. Maki RG, D’Adamo DR, Keohan ML, et al. Phase II study of sorafenib in patients with metastatic or recurrent sarcomas. J Clin Oncol. 2009;27:3133-3140.
  8. Ishida Y, Otsuka A, Kabashima K. Cutaneous angiosarcoma: update on biology and latest treatment. Curr Opin Oncol. 2018;30:107-112.
  9. Ray-Coquard I, Italiano A, Bompas E, et al. Sorafenib for patients with advanced angiosarcoma: a phase II trial from the French Sarcoma Group (GSF/GETO). Oncologist. 2012;17:260-266.
  10. Ray-Coquard IL, Domont J, Tresch-Bruneel E, et al. Paclitaxel given once per week with or without bevacizumab in patients with advanced angiosarcoma: a randomized phase II trial. J Clin Oncol. 2015;33:2797-2802.
  11. Pasquier E, Andre N, Street J, et al. Effective management of advanced angiosarcoma by the synergistic combination of propranolol and vinblastine-based metronomic chemotherapy: a bench to bedside study. EBioMedicine. 2016;6:87-95.
  12. Banavali S, Pasquier E, Andre N. Targeted therapy with propranolol and metronomic chemotherapy combination: sustained complete response of a relapsing metastatic angiosarcoma. Ecancermedicalscience. 2015;9:499.
  13. Chow W, Amaya CN, Rains S, et al. Growth attenuation of cutaneous angiosarcoma with propranolol-mediated beta-blockade. JAMA Dermatol. 2015;151:1226-1229.
  14. Daguze J, Saint-Jean M, Peuvrel L, et al. Visceral metastatic angiosarcoma treated effectively with oral cyclophosphamide combined with propranolol. JAAD Case Rep. 2016;2:497-499.
  15. Stiles JM, Amaya C, Rains S, et al. Targeting of beta adrenergic receptors results in therapeutic efficacy against models of hemangioendothelioma and angiosarcoma. PLoS One. 2013;8:e60021.
  16. Chang PY, Chung CH, Chang WC, et al. The effect of propranolol on the prognosis of hepatocellular carcinoma: a nationwide population-based study. PLoS One. 2019;14:e0216828.
  17. De Giorgi V, Grazzini M, Benemei S, et al. Propranolol for off-label treatment of patients with melanoma: results from a cohort study. JAMA Oncol. 2018;4:e172908.
  18. Rico M, Baglioni M, Bondarenko M, et al. Metformin and propranolol combination prevents cancer progression and metastasis in different breast cancer models. Oncotarget. 2017;8:2874-2889.
  19. Chisholm KM, Chang KW, Truong MT, et al. β-Adrenergic receptor expression in vascular tumors. Mod Pathol. 2012;25:1446-1451.
  20. Leaute-Labreze C, Dumas de la Roque E, Hubiche T, et al. Propranolol for severe hemangiomas of infancy. N Engl J Med. 2008;358:2649-2651.
  21. Maddox JC, Evans HL. Angiosarcoma of skin and soft tissue: a study of forty-four cases. Cancer. 1981;48:1907-1921.
  22. Morgan MB, Swann M, Somach S, et al. Cutaneous angiosarcoma: a case series with prognostic correlation. J Am Acad Dermatol. 2004;50:867-874.
  23. Zhou S, Liu P, Jiang W, et al. Identification of potential target genes associated with the effect of propranolol on angiosarcoma via microarray analysis. Oncol Lett. 2017;13:4267-4275.
Article PDF
Author and Disclosure Information

Drs. Nyers and Perry are from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston. Dr. De Jesús is from the Division of Dermatology, Department of Medicine, Ralph H. Johnson Veterans Affairs Medical Center, Charleston.

The authors report no conflict of interest.

Correspondence: Graciela De Jesús, MD, Division of Dermatology, Department of Medicine, Medical Services (111), Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee St, Charleston, SC 29401 ([email protected]).

Issue
Cutis - 110(6)
Publications
Topics
Page Number
E33-E36
Sections
Author and Disclosure Information

Drs. Nyers and Perry are from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston. Dr. De Jesús is from the Division of Dermatology, Department of Medicine, Ralph H. Johnson Veterans Affairs Medical Center, Charleston.

The authors report no conflict of interest.

Correspondence: Graciela De Jesús, MD, Division of Dermatology, Department of Medicine, Medical Services (111), Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee St, Charleston, SC 29401 ([email protected]).

Author and Disclosure Information

Drs. Nyers and Perry are from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston. Dr. De Jesús is from the Division of Dermatology, Department of Medicine, Ralph H. Johnson Veterans Affairs Medical Center, Charleston.

The authors report no conflict of interest.

Correspondence: Graciela De Jesús, MD, Division of Dermatology, Department of Medicine, Medical Services (111), Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee St, Charleston, SC 29401 ([email protected]).

Article PDF
Article PDF

To the Editor:

Angiosarcoma is a malignancy of the vascular endothelium that most commonly presents on the skin.1 Patients diagnosed with cutaneous angiosarcoma, which is a rare and aggressive malignancy, have a 5-year survival rate of approximately 30%.2,3 Angiosarcoma can be seen in the setting of chronic lymphedema; radiation therapy; and sporadically in elderly patients, where it is commonly seen on the head and neck. Presentation on the head and neck has been associated with worse outcomes, with a projected overall 10-year survival rate of 13.8%; the survival rate is lower if the tumor is surgically unresectable or larger in size. Metastasis can occur via both lymphatic and hematogenous routes, with pulmonary and hepatic metastases most frequently observed.1 Prognostications of poor outcomes for patients with head and neck cutaneous angiosarcoma via a 5-year survival rate were identified in a meta-analysis and included the following: patient age older than 70 years, larger tumors, tumor location of scalp vs face, nonsurgical treatments, and lack of clear margins on histology.2

Treatment of angiosarcoma historically has encompassed both surgical resection and adjuvant radiation therapy with suboptimal success. Evidence supporting various treatment regimens remains sparse due to the low incidence of the neoplasm. Although surgical resection is the only documented curative treatment, cutaneous angiosarcomas frequently are found to have positive surgical margins and require adjuvant radiation. Use of high-dose radiation (>50 Gy) with application over a wide treatment area such as total scalp irradiation is recommended.4 Although radiation has been found to diminish local recurrence rates, it has not substantially affected rates of distant disease recurrence.1 Cytotoxic chemotherapy has clinical utility in minimizing progression, but standard regimens afford a progression-free survival of only months.3 Adjuvant treatment with paclitaxel has been shown to have improved efficacy in scalp angiosarcoma vs other visceral sites, showing a nonprogression rate of 42% at 4 months after treatment.5 More recently, targeted chemotherapeutics, including the vascular endothelial growth factor inhibitor bevacizumab and tyrosine kinase inhibitor sorafenib, have shown some survival benefit, but it is unclear if these agents are superior to traditional cytotoxic agents.4,6-10 A phase 2 study of paclitaxel administered weekly with or without bevacizumab showed similar progression-free survival and overall survival, albeit at the expense of added toxicity experienced by participants in the combined group.10

The addition of the nonselective β-adrenergic blocker propranolol to the treatment armamentarium, which was pursued due to its utility in the treatment of benign infantile hemangioma and demonstrated ability to limit the expression of adrenergic receptors in angiosarcoma, has gained clinical attention for possible augmentation of cutaneous angiosarcoma therapy.11-14 Propranolol has been shown to reduce metastasis in other neoplasms—both vascular and nonvascular—and may play a role as an adjuvant treatment to current therapies in angiosarcoma.15-20 We report a patient with cutaneous angiosarcoma (T2 classification) with disease-free survival of nearly 6 years without evidence of recurrence in the setting of continuous propranolol use supplementary to chemotherapy and radiation.

Cutaneous angiosarcoma at the time of diagnosis.
FIGURE 1. Cutaneous angiosarcoma at the time of diagnosis. A, An ecchymotic patch showed extensive involvement of right scalp, forehead, and temple. B, Extension of the ecchymotic patch on the left side of the face.

A 78-year-old man with a history of multiple basal cell carcinomas, hypertension, and remote smoking history presented to the dermatology clinic with an enlarging red-brown plaque on the scalp of 2 months’ duration. The lesion had grown rapidly to involve the forehead, right temple, preauricular region, and parietal scalp. At presentation, the tumor measured more than 20 cm in diameter at its greatest point (Figure 1). Physical examination revealed a 6-mm purple nodule within the lesion on the patient’s right parietal scalp. No clinical lymphadenopathy was appreciated at the time of diagnosis. Punch biopsies of the right parietal scalp nodule and right temple patch showed findings consistent with angiosarcoma with diffuse cytoplasmic staining of CD31 in atypical endothelial cells and no staining for human herpesvirus 8 (Figure 2). Concurrent computed tomography of the head showed thickening of the right epidermis, dermis, and deeper scalp tissues, but there was no evidence of skull involvement. Computed tomography of the thorax, abdomen, and pelvis showed no evidence of metastatic disease. After a diagnostic workup, the patient was diagnosed with T2bN0M0 angiosarcoma.

A, A punch biopsy of the right parietal scalp showed cytologically atypical endothelial cells forming slitlike vascular spaces in the dermis (H&E, original magnification ×100). B, Cytoplasmic CD31 staining of endothelial lining of slit-like atypical
FIGURE 2. A, A punch biopsy of the right parietal scalp showed cytologically atypical endothelial cells forming slitlike vascular spaces in the dermis (H&E, original magnification ×100). B, Cytoplasmic CD31 staining of endothelial lining of slit-like atypical vascular spaces (original magnification ×400).

The lesion was determined to be nonresectable due to the extent of the patient’s cutaneous disease. The patient was started on a regimen of paclitaxel, scalp radiation, and oral propranolol. Propranolol 40 mg twice daily was initiated at the time of diagnosis with a plan to continue indefinitely. Starting 1 month after staging, the patient completed 10 weekly cycles of paclitaxel, and he was treated with 60 Gy of scalp radiation in 30 fractions, starting with the second cycle of paclitaxel. He tolerated both well with no reported adverse events. Repeat computed tomography performed 1 month after completion of chemotherapy and radiation showed no evidence of a mass or fluid collection in subcutaneous scalp tissues and no evidence of metastatic disease. This correlated with an observed clinical regression at 1 month and complete clinical response at 5 months with residual hemosiderin and radiation changes. The area of prior disease involvement subsequently evolved from violet to dusky gray in appearance to an eventual complete resolution 26 months after diagnosis, accompanied by atrophic radiation-induced sequelae (Figure 3).

No clinical evidence of disease 8 months after initial diagnosis following treatment with radiation therapy and adjunctive propranolol 40 mg twice daily. Only postinflammatory pigment change remained A B on examination.
FIGURE 3. A and B, No clinical evidence of disease 8 months after initial diagnosis following treatment with radiation therapy and adjunctive propranolol 40 mg twice daily. Only postinflammatory pigment change remained A B on examination.

The patient’s postchemotherapy course was complicated by hospitalization for a suspected malignant pleural effusion. Analysis revealed growing ground-glass opacities and nodules in the right lower lung lobe. A thoracentesis with cytology studies was negative for malignancy. Continued monitoring over 19 months demonstrated eventual resolution of those findings. He experienced notable complication from local radiation therapy to the scalp with chronic cutaneous ulceration refractory to wound care and surgical intervention. The patient did not exhibit additional signs or symptoms concerning for recurrence or metastasis and was followed by dermatology and oncology until he died nearly 5 years after initial diagnosis due to complications from acute hypoxic respiratory failure secondary to COVID-19. The last imaging obtained showed no convincing evidence of metastasis, though spinal imaging within a month of his death showed lesions favored to represent benign angiomatous growths. His survival after diagnosis ultimately reached 57 months without confirmed disease recurrence and cause of death unrelated to malignancy history, which is a markedly long documented survival for this extent of disease.

Cutaneous angiosarcoma is an aggressive yet rare malignancy without effective treatments for prolonging survival or eradicating disease. Cutaneous angiosarcoma of the head and neck has a reported 10-year survival rate of 13.8%.1 Although angiosarcoma in any location holds a bleak prognosis, cutaneous angiosarcoma of the scalp with a T2 classification has a 2-year survival rate of 0%. Moreover, even if remission is achieved, disease is highly recurrent, typically within months with the current standard of care.3,21,22

Emerging evidence for the possible role of β-adrenergic receptor blockade in the treatment of malignant vascular neoplasms is promising. Microarrays from a host of vascular growths have demonstrated expression of β-adrenergic receptors in 77% of sampled angiosarcoma specimens in addition to strong expression in infantile hemangiomas, hemangiomas, hemangioendotheliomas, and vascular malformations.19 Research findings have further verified the validity of this approach with the demonstration of b1-, b2-, and b3- adrenergic receptor expression by angiosarcoma cell lines. Propranolol subsequently was shown to effectively target proliferation of these cells and induce apoptosis in a dose-dependent manner and moreover be synergistic in effect with other chemotherapies.15 Several genes have exhibited differential expression between control tumor cells and propranolol-treated cells. Specifically, target genes including AXL (a receptor tyrosine kinase associated with cell adhesion, proliferation, and apoptosis and found to upregulated in melanoma and leukemia) and ERBB receptor feedback inhibitor 1 (receptor tyrosine kinase, with ERBB family members commonly overexpressed or mutated in the setting malignancy) have been posited as possible explanatory factors in the observed angiosarcoma response to propranolol.23

Several cases describing propranolol use as an adjunctive therapy for angiosarcoma suggest a beneficial role in clinical medicine. One case report described propranolol monotherapy for lesion to our patient, with a resultant reduction in Ki-67 as a measure of proliferative index within 1 week of initiating propranolol therapy.13 Propranolol also has been shown to halt or slow progression of metastatic disease in visceral and metastatic angiosarcomas.12-14 In combination with oral etoposide and cyclophosphamide, maintenance propranolol therapy in 7 cases of advanced cutaneous angiosarcoma resulted in 1 complete response and 3 very good partial responses, with a median progression-free survival of 11 months.11 Larger-scale studies have not been published, but the growing number of case reports and case series warrants further investigation of the utility of propranolol as an adjunct to current therapies in advanced angiosarcoma.

To the Editor:

Angiosarcoma is a malignancy of the vascular endothelium that most commonly presents on the skin.1 Patients diagnosed with cutaneous angiosarcoma, which is a rare and aggressive malignancy, have a 5-year survival rate of approximately 30%.2,3 Angiosarcoma can be seen in the setting of chronic lymphedema; radiation therapy; and sporadically in elderly patients, where it is commonly seen on the head and neck. Presentation on the head and neck has been associated with worse outcomes, with a projected overall 10-year survival rate of 13.8%; the survival rate is lower if the tumor is surgically unresectable or larger in size. Metastasis can occur via both lymphatic and hematogenous routes, with pulmonary and hepatic metastases most frequently observed.1 Prognostications of poor outcomes for patients with head and neck cutaneous angiosarcoma via a 5-year survival rate were identified in a meta-analysis and included the following: patient age older than 70 years, larger tumors, tumor location of scalp vs face, nonsurgical treatments, and lack of clear margins on histology.2

Treatment of angiosarcoma historically has encompassed both surgical resection and adjuvant radiation therapy with suboptimal success. Evidence supporting various treatment regimens remains sparse due to the low incidence of the neoplasm. Although surgical resection is the only documented curative treatment, cutaneous angiosarcomas frequently are found to have positive surgical margins and require adjuvant radiation. Use of high-dose radiation (>50 Gy) with application over a wide treatment area such as total scalp irradiation is recommended.4 Although radiation has been found to diminish local recurrence rates, it has not substantially affected rates of distant disease recurrence.1 Cytotoxic chemotherapy has clinical utility in minimizing progression, but standard regimens afford a progression-free survival of only months.3 Adjuvant treatment with paclitaxel has been shown to have improved efficacy in scalp angiosarcoma vs other visceral sites, showing a nonprogression rate of 42% at 4 months after treatment.5 More recently, targeted chemotherapeutics, including the vascular endothelial growth factor inhibitor bevacizumab and tyrosine kinase inhibitor sorafenib, have shown some survival benefit, but it is unclear if these agents are superior to traditional cytotoxic agents.4,6-10 A phase 2 study of paclitaxel administered weekly with or without bevacizumab showed similar progression-free survival and overall survival, albeit at the expense of added toxicity experienced by participants in the combined group.10

The addition of the nonselective β-adrenergic blocker propranolol to the treatment armamentarium, which was pursued due to its utility in the treatment of benign infantile hemangioma and demonstrated ability to limit the expression of adrenergic receptors in angiosarcoma, has gained clinical attention for possible augmentation of cutaneous angiosarcoma therapy.11-14 Propranolol has been shown to reduce metastasis in other neoplasms—both vascular and nonvascular—and may play a role as an adjuvant treatment to current therapies in angiosarcoma.15-20 We report a patient with cutaneous angiosarcoma (T2 classification) with disease-free survival of nearly 6 years without evidence of recurrence in the setting of continuous propranolol use supplementary to chemotherapy and radiation.

Cutaneous angiosarcoma at the time of diagnosis.
FIGURE 1. Cutaneous angiosarcoma at the time of diagnosis. A, An ecchymotic patch showed extensive involvement of right scalp, forehead, and temple. B, Extension of the ecchymotic patch on the left side of the face.

A 78-year-old man with a history of multiple basal cell carcinomas, hypertension, and remote smoking history presented to the dermatology clinic with an enlarging red-brown plaque on the scalp of 2 months’ duration. The lesion had grown rapidly to involve the forehead, right temple, preauricular region, and parietal scalp. At presentation, the tumor measured more than 20 cm in diameter at its greatest point (Figure 1). Physical examination revealed a 6-mm purple nodule within the lesion on the patient’s right parietal scalp. No clinical lymphadenopathy was appreciated at the time of diagnosis. Punch biopsies of the right parietal scalp nodule and right temple patch showed findings consistent with angiosarcoma with diffuse cytoplasmic staining of CD31 in atypical endothelial cells and no staining for human herpesvirus 8 (Figure 2). Concurrent computed tomography of the head showed thickening of the right epidermis, dermis, and deeper scalp tissues, but there was no evidence of skull involvement. Computed tomography of the thorax, abdomen, and pelvis showed no evidence of metastatic disease. After a diagnostic workup, the patient was diagnosed with T2bN0M0 angiosarcoma.

A, A punch biopsy of the right parietal scalp showed cytologically atypical endothelial cells forming slitlike vascular spaces in the dermis (H&E, original magnification ×100). B, Cytoplasmic CD31 staining of endothelial lining of slit-like atypical
FIGURE 2. A, A punch biopsy of the right parietal scalp showed cytologically atypical endothelial cells forming slitlike vascular spaces in the dermis (H&E, original magnification ×100). B, Cytoplasmic CD31 staining of endothelial lining of slit-like atypical vascular spaces (original magnification ×400).

The lesion was determined to be nonresectable due to the extent of the patient’s cutaneous disease. The patient was started on a regimen of paclitaxel, scalp radiation, and oral propranolol. Propranolol 40 mg twice daily was initiated at the time of diagnosis with a plan to continue indefinitely. Starting 1 month after staging, the patient completed 10 weekly cycles of paclitaxel, and he was treated with 60 Gy of scalp radiation in 30 fractions, starting with the second cycle of paclitaxel. He tolerated both well with no reported adverse events. Repeat computed tomography performed 1 month after completion of chemotherapy and radiation showed no evidence of a mass or fluid collection in subcutaneous scalp tissues and no evidence of metastatic disease. This correlated with an observed clinical regression at 1 month and complete clinical response at 5 months with residual hemosiderin and radiation changes. The area of prior disease involvement subsequently evolved from violet to dusky gray in appearance to an eventual complete resolution 26 months after diagnosis, accompanied by atrophic radiation-induced sequelae (Figure 3).

No clinical evidence of disease 8 months after initial diagnosis following treatment with radiation therapy and adjunctive propranolol 40 mg twice daily. Only postinflammatory pigment change remained A B on examination.
FIGURE 3. A and B, No clinical evidence of disease 8 months after initial diagnosis following treatment with radiation therapy and adjunctive propranolol 40 mg twice daily. Only postinflammatory pigment change remained A B on examination.

The patient’s postchemotherapy course was complicated by hospitalization for a suspected malignant pleural effusion. Analysis revealed growing ground-glass opacities and nodules in the right lower lung lobe. A thoracentesis with cytology studies was negative for malignancy. Continued monitoring over 19 months demonstrated eventual resolution of those findings. He experienced notable complication from local radiation therapy to the scalp with chronic cutaneous ulceration refractory to wound care and surgical intervention. The patient did not exhibit additional signs or symptoms concerning for recurrence or metastasis and was followed by dermatology and oncology until he died nearly 5 years after initial diagnosis due to complications from acute hypoxic respiratory failure secondary to COVID-19. The last imaging obtained showed no convincing evidence of metastasis, though spinal imaging within a month of his death showed lesions favored to represent benign angiomatous growths. His survival after diagnosis ultimately reached 57 months without confirmed disease recurrence and cause of death unrelated to malignancy history, which is a markedly long documented survival for this extent of disease.

Cutaneous angiosarcoma is an aggressive yet rare malignancy without effective treatments for prolonging survival or eradicating disease. Cutaneous angiosarcoma of the head and neck has a reported 10-year survival rate of 13.8%.1 Although angiosarcoma in any location holds a bleak prognosis, cutaneous angiosarcoma of the scalp with a T2 classification has a 2-year survival rate of 0%. Moreover, even if remission is achieved, disease is highly recurrent, typically within months with the current standard of care.3,21,22

Emerging evidence for the possible role of β-adrenergic receptor blockade in the treatment of malignant vascular neoplasms is promising. Microarrays from a host of vascular growths have demonstrated expression of β-adrenergic receptors in 77% of sampled angiosarcoma specimens in addition to strong expression in infantile hemangiomas, hemangiomas, hemangioendotheliomas, and vascular malformations.19 Research findings have further verified the validity of this approach with the demonstration of b1-, b2-, and b3- adrenergic receptor expression by angiosarcoma cell lines. Propranolol subsequently was shown to effectively target proliferation of these cells and induce apoptosis in a dose-dependent manner and moreover be synergistic in effect with other chemotherapies.15 Several genes have exhibited differential expression between control tumor cells and propranolol-treated cells. Specifically, target genes including AXL (a receptor tyrosine kinase associated with cell adhesion, proliferation, and apoptosis and found to upregulated in melanoma and leukemia) and ERBB receptor feedback inhibitor 1 (receptor tyrosine kinase, with ERBB family members commonly overexpressed or mutated in the setting malignancy) have been posited as possible explanatory factors in the observed angiosarcoma response to propranolol.23

Several cases describing propranolol use as an adjunctive therapy for angiosarcoma suggest a beneficial role in clinical medicine. One case report described propranolol monotherapy for lesion to our patient, with a resultant reduction in Ki-67 as a measure of proliferative index within 1 week of initiating propranolol therapy.13 Propranolol also has been shown to halt or slow progression of metastatic disease in visceral and metastatic angiosarcomas.12-14 In combination with oral etoposide and cyclophosphamide, maintenance propranolol therapy in 7 cases of advanced cutaneous angiosarcoma resulted in 1 complete response and 3 very good partial responses, with a median progression-free survival of 11 months.11 Larger-scale studies have not been published, but the growing number of case reports and case series warrants further investigation of the utility of propranolol as an adjunct to current therapies in advanced angiosarcoma.

References
  1. Abraham JA, Hornicek FJ, Kaufman AM, et al. Treatment and outcome of 82 patients with angiosarcoma. Ann Surg Oncol. 2007;14:1953-1967.
  2. Shin JY, Roh SG, Lee NH, et al. Predisposing factors for poor prognosis of angiosarcoma of the scalp and face: systematic review and meta-analysis. Head Neck. 2017;39:380-386.
  3. Fury MG, Antonescu CR, Zee KJV, et al. A 14-year retrospective review of angiosarcoma: clinical characteristics, prognostic factors, and treatment outcomes with surgery and chemotherapy. Cancer. 2005;11:241-247.
  4. Dossett LA, Harrington M, Cruse CW, et al. Cutaneous angiosarcoma. Curr Probl Cancer. 2015;39:258-263.
  5. Penel N, Bui BN, Bay JO, et al. Phase II trial of weekly paclitaxel for unresectable angiosarcoma: the ANGIOTAX study. J Clin Oncol. 2008;26:5269-5274.
  6. Agulnik M, Yarber JL, Okuno SH, et al. An open-label, multicenter, phase II study of bevacizumab for the treatment of angiosarcoma and epithelioid hemangioendotheliomas. Ann Oncol. 2013;24:257-263.
  7. Maki RG, D’Adamo DR, Keohan ML, et al. Phase II study of sorafenib in patients with metastatic or recurrent sarcomas. J Clin Oncol. 2009;27:3133-3140.
  8. Ishida Y, Otsuka A, Kabashima K. Cutaneous angiosarcoma: update on biology and latest treatment. Curr Opin Oncol. 2018;30:107-112.
  9. Ray-Coquard I, Italiano A, Bompas E, et al. Sorafenib for patients with advanced angiosarcoma: a phase II trial from the French Sarcoma Group (GSF/GETO). Oncologist. 2012;17:260-266.
  10. Ray-Coquard IL, Domont J, Tresch-Bruneel E, et al. Paclitaxel given once per week with or without bevacizumab in patients with advanced angiosarcoma: a randomized phase II trial. J Clin Oncol. 2015;33:2797-2802.
  11. Pasquier E, Andre N, Street J, et al. Effective management of advanced angiosarcoma by the synergistic combination of propranolol and vinblastine-based metronomic chemotherapy: a bench to bedside study. EBioMedicine. 2016;6:87-95.
  12. Banavali S, Pasquier E, Andre N. Targeted therapy with propranolol and metronomic chemotherapy combination: sustained complete response of a relapsing metastatic angiosarcoma. Ecancermedicalscience. 2015;9:499.
  13. Chow W, Amaya CN, Rains S, et al. Growth attenuation of cutaneous angiosarcoma with propranolol-mediated beta-blockade. JAMA Dermatol. 2015;151:1226-1229.
  14. Daguze J, Saint-Jean M, Peuvrel L, et al. Visceral metastatic angiosarcoma treated effectively with oral cyclophosphamide combined with propranolol. JAAD Case Rep. 2016;2:497-499.
  15. Stiles JM, Amaya C, Rains S, et al. Targeting of beta adrenergic receptors results in therapeutic efficacy against models of hemangioendothelioma and angiosarcoma. PLoS One. 2013;8:e60021.
  16. Chang PY, Chung CH, Chang WC, et al. The effect of propranolol on the prognosis of hepatocellular carcinoma: a nationwide population-based study. PLoS One. 2019;14:e0216828.
  17. De Giorgi V, Grazzini M, Benemei S, et al. Propranolol for off-label treatment of patients with melanoma: results from a cohort study. JAMA Oncol. 2018;4:e172908.
  18. Rico M, Baglioni M, Bondarenko M, et al. Metformin and propranolol combination prevents cancer progression and metastasis in different breast cancer models. Oncotarget. 2017;8:2874-2889.
  19. Chisholm KM, Chang KW, Truong MT, et al. β-Adrenergic receptor expression in vascular tumors. Mod Pathol. 2012;25:1446-1451.
  20. Leaute-Labreze C, Dumas de la Roque E, Hubiche T, et al. Propranolol for severe hemangiomas of infancy. N Engl J Med. 2008;358:2649-2651.
  21. Maddox JC, Evans HL. Angiosarcoma of skin and soft tissue: a study of forty-four cases. Cancer. 1981;48:1907-1921.
  22. Morgan MB, Swann M, Somach S, et al. Cutaneous angiosarcoma: a case series with prognostic correlation. J Am Acad Dermatol. 2004;50:867-874.
  23. Zhou S, Liu P, Jiang W, et al. Identification of potential target genes associated with the effect of propranolol on angiosarcoma via microarray analysis. Oncol Lett. 2017;13:4267-4275.
References
  1. Abraham JA, Hornicek FJ, Kaufman AM, et al. Treatment and outcome of 82 patients with angiosarcoma. Ann Surg Oncol. 2007;14:1953-1967.
  2. Shin JY, Roh SG, Lee NH, et al. Predisposing factors for poor prognosis of angiosarcoma of the scalp and face: systematic review and meta-analysis. Head Neck. 2017;39:380-386.
  3. Fury MG, Antonescu CR, Zee KJV, et al. A 14-year retrospective review of angiosarcoma: clinical characteristics, prognostic factors, and treatment outcomes with surgery and chemotherapy. Cancer. 2005;11:241-247.
  4. Dossett LA, Harrington M, Cruse CW, et al. Cutaneous angiosarcoma. Curr Probl Cancer. 2015;39:258-263.
  5. Penel N, Bui BN, Bay JO, et al. Phase II trial of weekly paclitaxel for unresectable angiosarcoma: the ANGIOTAX study. J Clin Oncol. 2008;26:5269-5274.
  6. Agulnik M, Yarber JL, Okuno SH, et al. An open-label, multicenter, phase II study of bevacizumab for the treatment of angiosarcoma and epithelioid hemangioendotheliomas. Ann Oncol. 2013;24:257-263.
  7. Maki RG, D’Adamo DR, Keohan ML, et al. Phase II study of sorafenib in patients with metastatic or recurrent sarcomas. J Clin Oncol. 2009;27:3133-3140.
  8. Ishida Y, Otsuka A, Kabashima K. Cutaneous angiosarcoma: update on biology and latest treatment. Curr Opin Oncol. 2018;30:107-112.
  9. Ray-Coquard I, Italiano A, Bompas E, et al. Sorafenib for patients with advanced angiosarcoma: a phase II trial from the French Sarcoma Group (GSF/GETO). Oncologist. 2012;17:260-266.
  10. Ray-Coquard IL, Domont J, Tresch-Bruneel E, et al. Paclitaxel given once per week with or without bevacizumab in patients with advanced angiosarcoma: a randomized phase II trial. J Clin Oncol. 2015;33:2797-2802.
  11. Pasquier E, Andre N, Street J, et al. Effective management of advanced angiosarcoma by the synergistic combination of propranolol and vinblastine-based metronomic chemotherapy: a bench to bedside study. EBioMedicine. 2016;6:87-95.
  12. Banavali S, Pasquier E, Andre N. Targeted therapy with propranolol and metronomic chemotherapy combination: sustained complete response of a relapsing metastatic angiosarcoma. Ecancermedicalscience. 2015;9:499.
  13. Chow W, Amaya CN, Rains S, et al. Growth attenuation of cutaneous angiosarcoma with propranolol-mediated beta-blockade. JAMA Dermatol. 2015;151:1226-1229.
  14. Daguze J, Saint-Jean M, Peuvrel L, et al. Visceral metastatic angiosarcoma treated effectively with oral cyclophosphamide combined with propranolol. JAAD Case Rep. 2016;2:497-499.
  15. Stiles JM, Amaya C, Rains S, et al. Targeting of beta adrenergic receptors results in therapeutic efficacy against models of hemangioendothelioma and angiosarcoma. PLoS One. 2013;8:e60021.
  16. Chang PY, Chung CH, Chang WC, et al. The effect of propranolol on the prognosis of hepatocellular carcinoma: a nationwide population-based study. PLoS One. 2019;14:e0216828.
  17. De Giorgi V, Grazzini M, Benemei S, et al. Propranolol for off-label treatment of patients with melanoma: results from a cohort study. JAMA Oncol. 2018;4:e172908.
  18. Rico M, Baglioni M, Bondarenko M, et al. Metformin and propranolol combination prevents cancer progression and metastasis in different breast cancer models. Oncotarget. 2017;8:2874-2889.
  19. Chisholm KM, Chang KW, Truong MT, et al. β-Adrenergic receptor expression in vascular tumors. Mod Pathol. 2012;25:1446-1451.
  20. Leaute-Labreze C, Dumas de la Roque E, Hubiche T, et al. Propranolol for severe hemangiomas of infancy. N Engl J Med. 2008;358:2649-2651.
  21. Maddox JC, Evans HL. Angiosarcoma of skin and soft tissue: a study of forty-four cases. Cancer. 1981;48:1907-1921.
  22. Morgan MB, Swann M, Somach S, et al. Cutaneous angiosarcoma: a case series with prognostic correlation. J Am Acad Dermatol. 2004;50:867-874.
  23. Zhou S, Liu P, Jiang W, et al. Identification of potential target genes associated with the effect of propranolol on angiosarcoma via microarray analysis. Oncol Lett. 2017;13:4267-4275.
Issue
Cutis - 110(6)
Issue
Cutis - 110(6)
Page Number
E33-E36
Page Number
E33-E36
Publications
Publications
Topics
Article Type
Display Headline
Oral Propranolol Used as Adjunct Therapy in Cutaneous Angiosarcoma
Display Headline
Oral Propranolol Used as Adjunct Therapy in Cutaneous Angiosarcoma
Sections
Inside the Article

PRACTICE POINTS

  • In one classic presentation, cutaneous angiosarcoma characteristically appears as a bruiselike patch on the head and neck of an elderly gentleman.
  • Although cutaneous angiosarcoma typically portends a poor prognosis at the time of diagnosis, adjunctive oral propranolol may be a promising and relatively benign therapy, posited to afford benefit in a manner similar to its efficacy in the treatment of infantile hemangiomas.
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

Use of Dupilumab in Severe, Multifactorial, Chronic Itch for Geriatric Patients

Article Type
Changed
Thu, 01/05/2023 - 09:11
Display Headline
Use of Dupilumab in Severe, Multifactorial, Chronic Itch for Geriatric Patients

To the Editor:

Today’s geriatric population is the fastest growing in history. The National Institutes of Health predicts there will be over 1.5 billion individuals aged 65 years and older by the year 2050: 17% of the world’s population.1 Pruritus—either acute or chronic (>6 weeks)—is defined as a sensory perception that leads to an intense desire to scratch.2 Chronic pruritus is an increasing health concern that impacts quality of life within the geriatric population. Elderly patients have various risk factors for developing chronic itch, including aging skin, polypharmacy, and increased systemic comorbidities.3-7

Although the therapeutic armamentarium for chronic itch continues to grow, health care providers often are hesitant to prescribe medications for geriatric patients because of comorbidities and potential drug-drug interactions. Novel biologic therapies now provide alternatives for this complex population. Dupilumab is a fully humanized, monoclonal antibody approved for treatment-resistant atopic dermatitis. This biologic prevents helper T-cell (TH2) signaling, IL-4 and IL-13 release, and subsequent effector cell (eg, mast cell, eosinophil) activity.8-10 The combined efficacy and safety of this medication has changed the treatment landscape of resistant atopic dermatitis. We present the use of dupilumab in a geriatric patient with severe and recalcitrant itch resistant to numerous topical and oral medications.

An 81-year-old man presented to the clinic with a long history of generalized pruritic rash. His medical history was significant for insulin-dependent type 2 diabetes mellitus (T2DM), hypertension, and renal cancer following a right nephrectomy. Laboratory results approximately 14 months prior to the visit revealed a blood urea nitrogen level of 31 mg/dL (reference range, 7–20 mg/dL), creatinine level of 2.20 mg/dL (reference range, 0.7–1.3 mg/dL), and glomerular filtration rate of 29 mL/min (reference range, 90–120 mL/min). Physical examination revealed numerous pink excoriated papules on the face, neck, trunk, and extremities. Lichenified plaques were present on both arms and legs. The patient received the diagnosis of severe atopic dermatitis with greater than 10% body surface area involvement. The investigator global assessment score was 4/4, indicating severe disease burden, and biopsy results reported spongiotic dermatitis. He proceeded to trial various topical corticosteroids, including hydrocortisone ointment 2.5%, betamethasone valerate ointment 0.01%, fluocinonide ointment 0.05%, and mupirocin ointment without benefit. Three subsequent courses of oral steroids failed to provide durable relief. At this point, the peak pruritus numerical rating scale (NRS) score was 7/10, indicating severe pruritus, with a negative impact on the patient’s quality of life and sleep.

Therapy was switched to tacrolimus acetonide ointment 0.1%, betamethasone dipropionate ointment 0.05%, and triamcinolone acetonide ointment 0.1%. Eleven days later, the patient denied experiencing any response to the topical regimen and sought alternative therapy for the itch and associated poor sleep; the NRS score was 10/10, indicating very severe pruritus. Prednisone 20 mg and doxepin 10 mg were initiated for symptom management until the intended transition to dupilumab. The patient began dupilumab with a loading dose of 600 mg, then 300 mg every other week thereafter. At 2- and 4-month follow-up, the patient reported notable relief in symptoms. The rash had improved, and the NRS score decreased from 10/10 to 3/10. He endorsed improved sleep and quality of life.

Pruritus may arise from a series of age-related mechanisms such as structural and chemical changes within the epidermis, underlying neuropathy, medication side effects, infection, malignancy, thyroid dysregulation, liver disease, and chronic kidney disease (CKD).5,6,11 Identifying the underlying etiology often is difficult and involves a complete history and physical examination as well as an appropriate contextualized laboratory workup.

Our patient’s comorbid T2DM and renal disease may have contributed to the pruritus. Type 2 diabetes mellitus can cause diabetic neuropathy, a sequela known to lead to various complications, including pruritus. One study identified a 4-fold increase in pruritus in those with diabetic polyneuropathy compared with age-matched nondiabetics.12,13 An additional study found that pruritus was present in 70% of patients with small fiber neuropathy.14 We needed to consider the role of our patient’s insulin-dependent T2DM and potential underlying neuropathy when addressing the pruritic symptoms.

Furthermore, our patient’s stage IV CKD and elevated urea level also may factor into the pruritus. The pathophysiology of CKD-associated pruritus (also referred to as uremic pruritus) remains poorly understood. Suggested mechanisms include immune-mediated neural inflammation and erroneous nociceptive-receptor activity.15,16 Although uremic pruritus is appreciated primarily in late dialysis-dependent disease, research shows that a notable portion of those with lesser disease, similar to our patient, also experience a significant itch burden.17 Diminishing pruritus is difficult and often aided by management of the underlying renal disease.18

In addition to disease management, symptomatic treatment incorporates the use of emollients, corticosteroids, and antihistamines. Unfortunately, the clinical response in the elderly population to such regimens often is poor.19 Dupilumab is an optimistic therapeutic option for chronic pruritus. By inhibiting the IL-4α receptor found on helper T cells, this biologic inhibits TH2 differentiation and subsequent inflammatory activity. One report identified an optimistic response to dupilumab in the management of uremic pruritus.20 The remarkable improvement and absence of adverse effects in our patient confirmed the utility and safety of dupilumab in complex cases such as elderly patients with multiple comorbidities. Such relief may result from inhibition of proinflammatory cytokine activity as well as decreased afferent spinal cord itch stimuli.10 The positive results from this case cast a favorable outlook on the treatment of chronic itch in the complex geriatric population.

References
  1. World’s older population grows dramatically. News release. National Institute on Aging. Published March 28, 2016. Accessed December 23, 2022. http://www.nih.gov/news-events/news-releases/worlds-older-population-grows-dramatically
  2. Grundmann S, Ständer S. Chronic pruritus: clinics and treatment. Ann Dermatol. 2011;23:1-11.
  3. Berger TG, Shive M, Harper GM. Pruritus in the older patient: a clinical review. JAMA. 2013;310:2443-2450. doi:10.1001/jama.2013.282023
  4. Valdes-Rodriguez, R, Mollanazar NK, González-Muro J, et al. Itch prevalence and characteristics in a Hispanic geriatric population: a comprehensive study using a standardized itch questionnaire. Acta Derm Venereol. 2015;95:417-421. doi:10.2340/00015555-1968
  5. Li J, Tang H, Hu X, et al. Aquaporin-3 gene and protein expression in sun-protected human skin decreases with skin ageing. Australas J Dermatol. 2010;51:106-112.
  6. Choi EH, Man MQ, Xu P, et al. Stratum corneum acidification is impaired in moderately aged human and murine skin. J Invest Dermatol. 2007;127:2847-2856.
  7. Fenske NA, Lober CW. Structural and functional changes of normal aging skin. J Am Acad Dermatol. 1986;15(4 pt 1):571-585.
  8. Paller AS, Kabashima K, Bieber T. Therapeutic pipeline for atopic dermatitis: end of the drought? J Allergy Clin Immunol. 2017;140:633-643. doi:10.1016/j.jaci.2017.07.006
  9. Kabashima K. New concept of the pathogenesis of atopic dermatitis: interplay among the barrier, allergy, and pruritus as a trinity. J Dermatol Sci. 2013;70:3-11.
  10. Feld M, Garcia R, Buddenkotte J, et al. The pruritus- and TH2-associated cytokine IL-31 promotes growth of sensory nerves. J Allergy Clin Immunol. 2016;138:500-508.
  11. Valdes-Rodriguez R, Stull C, Yosipovitch G. Chronic pruritus in the elderly: pathophysiology, diagnosis and management. Drugs Aging. 2015;32:201-215. doi:10.1007/s40266-015-0246-0
  12. Misery L, Brenaut E, Le Garrec R, et al. Neuropathic pruritus. Nat Rev Neurol. 2014;10:408-416.
  13. Yamaoka H, Sasaki H, Yamasaki H, et al. Truncal pruritus of unknown origin may be a symptom of diabetic polyneuropathy. Diabetes Care. 2010;33:150-155.
  14. Brenaut E, Marcorelles P, Genestet S, et al. Pruritus: an underrecognized symptom of small-fiber neuropathies. J Am Acad Dermatol. 2015;72:328-332.
  15. Adigun M, Badu LA, Berner NM, et al. Uremic pruritus review. US Pharm. 2015;40:HS12-HS15.
  16. Simonsen E, Komenda P, Lerner B, et al. Treatment of uremic pruritus: a systematic review. Am J Kidney Dis. 2017;70:638-655.
  17. Carstens E, Akiyama T, eds. Itch: Mechanisms and Treatment. CRC Press/Taylor & Francis; 2014.
  18. Shirazian S, Aina O, Park Y, et al. Chronic kidney disease-associated pruritus: impact on quality of life and current management challenges. Int J Nephrol Renovasc Dis. 2017;10:11-26.
  19. Brummer GC, Wang LT, Sontheimer RD. A possible role for dupilumab (Dupixent) in the management of idiopathic chronic eczematous eruption of aging. Dermatol Online J. 2018;24:13030/qt55z1f6xh.
  20. Silverberg JI, Brieva J. A successful case of dupilumab treatment for severe uremic pruritus. JAAD Case Rep. 2019;5:339-341.
Article PDF
Author and Disclosure Information

Dr. Pousti is from the Department of Dermatology, University of California San Diego, La Jolla. Dr. Valdes-Rodriguez is from the Department of Dermatology, University of Florida, Gainesville.

The authors report no conflict of interest.

Correspondence: Bobak T. Pousti, MD, Department of Dermatology, University of California San Diego, 8899 University Center Ln, La Jolla, CA 92122 ([email protected]).

Issue
Cutis - 110(6)
Publications
Topics
Page Number
E31-E32
Sections
Author and Disclosure Information

Dr. Pousti is from the Department of Dermatology, University of California San Diego, La Jolla. Dr. Valdes-Rodriguez is from the Department of Dermatology, University of Florida, Gainesville.

The authors report no conflict of interest.

Correspondence: Bobak T. Pousti, MD, Department of Dermatology, University of California San Diego, 8899 University Center Ln, La Jolla, CA 92122 ([email protected]).

Author and Disclosure Information

Dr. Pousti is from the Department of Dermatology, University of California San Diego, La Jolla. Dr. Valdes-Rodriguez is from the Department of Dermatology, University of Florida, Gainesville.

The authors report no conflict of interest.

Correspondence: Bobak T. Pousti, MD, Department of Dermatology, University of California San Diego, 8899 University Center Ln, La Jolla, CA 92122 ([email protected]).

Article PDF
Article PDF

To the Editor:

Today’s geriatric population is the fastest growing in history. The National Institutes of Health predicts there will be over 1.5 billion individuals aged 65 years and older by the year 2050: 17% of the world’s population.1 Pruritus—either acute or chronic (>6 weeks)—is defined as a sensory perception that leads to an intense desire to scratch.2 Chronic pruritus is an increasing health concern that impacts quality of life within the geriatric population. Elderly patients have various risk factors for developing chronic itch, including aging skin, polypharmacy, and increased systemic comorbidities.3-7

Although the therapeutic armamentarium for chronic itch continues to grow, health care providers often are hesitant to prescribe medications for geriatric patients because of comorbidities and potential drug-drug interactions. Novel biologic therapies now provide alternatives for this complex population. Dupilumab is a fully humanized, monoclonal antibody approved for treatment-resistant atopic dermatitis. This biologic prevents helper T-cell (TH2) signaling, IL-4 and IL-13 release, and subsequent effector cell (eg, mast cell, eosinophil) activity.8-10 The combined efficacy and safety of this medication has changed the treatment landscape of resistant atopic dermatitis. We present the use of dupilumab in a geriatric patient with severe and recalcitrant itch resistant to numerous topical and oral medications.

An 81-year-old man presented to the clinic with a long history of generalized pruritic rash. His medical history was significant for insulin-dependent type 2 diabetes mellitus (T2DM), hypertension, and renal cancer following a right nephrectomy. Laboratory results approximately 14 months prior to the visit revealed a blood urea nitrogen level of 31 mg/dL (reference range, 7–20 mg/dL), creatinine level of 2.20 mg/dL (reference range, 0.7–1.3 mg/dL), and glomerular filtration rate of 29 mL/min (reference range, 90–120 mL/min). Physical examination revealed numerous pink excoriated papules on the face, neck, trunk, and extremities. Lichenified plaques were present on both arms and legs. The patient received the diagnosis of severe atopic dermatitis with greater than 10% body surface area involvement. The investigator global assessment score was 4/4, indicating severe disease burden, and biopsy results reported spongiotic dermatitis. He proceeded to trial various topical corticosteroids, including hydrocortisone ointment 2.5%, betamethasone valerate ointment 0.01%, fluocinonide ointment 0.05%, and mupirocin ointment without benefit. Three subsequent courses of oral steroids failed to provide durable relief. At this point, the peak pruritus numerical rating scale (NRS) score was 7/10, indicating severe pruritus, with a negative impact on the patient’s quality of life and sleep.

Therapy was switched to tacrolimus acetonide ointment 0.1%, betamethasone dipropionate ointment 0.05%, and triamcinolone acetonide ointment 0.1%. Eleven days later, the patient denied experiencing any response to the topical regimen and sought alternative therapy for the itch and associated poor sleep; the NRS score was 10/10, indicating very severe pruritus. Prednisone 20 mg and doxepin 10 mg were initiated for symptom management until the intended transition to dupilumab. The patient began dupilumab with a loading dose of 600 mg, then 300 mg every other week thereafter. At 2- and 4-month follow-up, the patient reported notable relief in symptoms. The rash had improved, and the NRS score decreased from 10/10 to 3/10. He endorsed improved sleep and quality of life.

Pruritus may arise from a series of age-related mechanisms such as structural and chemical changes within the epidermis, underlying neuropathy, medication side effects, infection, malignancy, thyroid dysregulation, liver disease, and chronic kidney disease (CKD).5,6,11 Identifying the underlying etiology often is difficult and involves a complete history and physical examination as well as an appropriate contextualized laboratory workup.

Our patient’s comorbid T2DM and renal disease may have contributed to the pruritus. Type 2 diabetes mellitus can cause diabetic neuropathy, a sequela known to lead to various complications, including pruritus. One study identified a 4-fold increase in pruritus in those with diabetic polyneuropathy compared with age-matched nondiabetics.12,13 An additional study found that pruritus was present in 70% of patients with small fiber neuropathy.14 We needed to consider the role of our patient’s insulin-dependent T2DM and potential underlying neuropathy when addressing the pruritic symptoms.

Furthermore, our patient’s stage IV CKD and elevated urea level also may factor into the pruritus. The pathophysiology of CKD-associated pruritus (also referred to as uremic pruritus) remains poorly understood. Suggested mechanisms include immune-mediated neural inflammation and erroneous nociceptive-receptor activity.15,16 Although uremic pruritus is appreciated primarily in late dialysis-dependent disease, research shows that a notable portion of those with lesser disease, similar to our patient, also experience a significant itch burden.17 Diminishing pruritus is difficult and often aided by management of the underlying renal disease.18

In addition to disease management, symptomatic treatment incorporates the use of emollients, corticosteroids, and antihistamines. Unfortunately, the clinical response in the elderly population to such regimens often is poor.19 Dupilumab is an optimistic therapeutic option for chronic pruritus. By inhibiting the IL-4α receptor found on helper T cells, this biologic inhibits TH2 differentiation and subsequent inflammatory activity. One report identified an optimistic response to dupilumab in the management of uremic pruritus.20 The remarkable improvement and absence of adverse effects in our patient confirmed the utility and safety of dupilumab in complex cases such as elderly patients with multiple comorbidities. Such relief may result from inhibition of proinflammatory cytokine activity as well as decreased afferent spinal cord itch stimuli.10 The positive results from this case cast a favorable outlook on the treatment of chronic itch in the complex geriatric population.

To the Editor:

Today’s geriatric population is the fastest growing in history. The National Institutes of Health predicts there will be over 1.5 billion individuals aged 65 years and older by the year 2050: 17% of the world’s population.1 Pruritus—either acute or chronic (>6 weeks)—is defined as a sensory perception that leads to an intense desire to scratch.2 Chronic pruritus is an increasing health concern that impacts quality of life within the geriatric population. Elderly patients have various risk factors for developing chronic itch, including aging skin, polypharmacy, and increased systemic comorbidities.3-7

Although the therapeutic armamentarium for chronic itch continues to grow, health care providers often are hesitant to prescribe medications for geriatric patients because of comorbidities and potential drug-drug interactions. Novel biologic therapies now provide alternatives for this complex population. Dupilumab is a fully humanized, monoclonal antibody approved for treatment-resistant atopic dermatitis. This biologic prevents helper T-cell (TH2) signaling, IL-4 and IL-13 release, and subsequent effector cell (eg, mast cell, eosinophil) activity.8-10 The combined efficacy and safety of this medication has changed the treatment landscape of resistant atopic dermatitis. We present the use of dupilumab in a geriatric patient with severe and recalcitrant itch resistant to numerous topical and oral medications.

An 81-year-old man presented to the clinic with a long history of generalized pruritic rash. His medical history was significant for insulin-dependent type 2 diabetes mellitus (T2DM), hypertension, and renal cancer following a right nephrectomy. Laboratory results approximately 14 months prior to the visit revealed a blood urea nitrogen level of 31 mg/dL (reference range, 7–20 mg/dL), creatinine level of 2.20 mg/dL (reference range, 0.7–1.3 mg/dL), and glomerular filtration rate of 29 mL/min (reference range, 90–120 mL/min). Physical examination revealed numerous pink excoriated papules on the face, neck, trunk, and extremities. Lichenified plaques were present on both arms and legs. The patient received the diagnosis of severe atopic dermatitis with greater than 10% body surface area involvement. The investigator global assessment score was 4/4, indicating severe disease burden, and biopsy results reported spongiotic dermatitis. He proceeded to trial various topical corticosteroids, including hydrocortisone ointment 2.5%, betamethasone valerate ointment 0.01%, fluocinonide ointment 0.05%, and mupirocin ointment without benefit. Three subsequent courses of oral steroids failed to provide durable relief. At this point, the peak pruritus numerical rating scale (NRS) score was 7/10, indicating severe pruritus, with a negative impact on the patient’s quality of life and sleep.

Therapy was switched to tacrolimus acetonide ointment 0.1%, betamethasone dipropionate ointment 0.05%, and triamcinolone acetonide ointment 0.1%. Eleven days later, the patient denied experiencing any response to the topical regimen and sought alternative therapy for the itch and associated poor sleep; the NRS score was 10/10, indicating very severe pruritus. Prednisone 20 mg and doxepin 10 mg were initiated for symptom management until the intended transition to dupilumab. The patient began dupilumab with a loading dose of 600 mg, then 300 mg every other week thereafter. At 2- and 4-month follow-up, the patient reported notable relief in symptoms. The rash had improved, and the NRS score decreased from 10/10 to 3/10. He endorsed improved sleep and quality of life.

Pruritus may arise from a series of age-related mechanisms such as structural and chemical changes within the epidermis, underlying neuropathy, medication side effects, infection, malignancy, thyroid dysregulation, liver disease, and chronic kidney disease (CKD).5,6,11 Identifying the underlying etiology often is difficult and involves a complete history and physical examination as well as an appropriate contextualized laboratory workup.

Our patient’s comorbid T2DM and renal disease may have contributed to the pruritus. Type 2 diabetes mellitus can cause diabetic neuropathy, a sequela known to lead to various complications, including pruritus. One study identified a 4-fold increase in pruritus in those with diabetic polyneuropathy compared with age-matched nondiabetics.12,13 An additional study found that pruritus was present in 70% of patients with small fiber neuropathy.14 We needed to consider the role of our patient’s insulin-dependent T2DM and potential underlying neuropathy when addressing the pruritic symptoms.

Furthermore, our patient’s stage IV CKD and elevated urea level also may factor into the pruritus. The pathophysiology of CKD-associated pruritus (also referred to as uremic pruritus) remains poorly understood. Suggested mechanisms include immune-mediated neural inflammation and erroneous nociceptive-receptor activity.15,16 Although uremic pruritus is appreciated primarily in late dialysis-dependent disease, research shows that a notable portion of those with lesser disease, similar to our patient, also experience a significant itch burden.17 Diminishing pruritus is difficult and often aided by management of the underlying renal disease.18

In addition to disease management, symptomatic treatment incorporates the use of emollients, corticosteroids, and antihistamines. Unfortunately, the clinical response in the elderly population to such regimens often is poor.19 Dupilumab is an optimistic therapeutic option for chronic pruritus. By inhibiting the IL-4α receptor found on helper T cells, this biologic inhibits TH2 differentiation and subsequent inflammatory activity. One report identified an optimistic response to dupilumab in the management of uremic pruritus.20 The remarkable improvement and absence of adverse effects in our patient confirmed the utility and safety of dupilumab in complex cases such as elderly patients with multiple comorbidities. Such relief may result from inhibition of proinflammatory cytokine activity as well as decreased afferent spinal cord itch stimuli.10 The positive results from this case cast a favorable outlook on the treatment of chronic itch in the complex geriatric population.

References
  1. World’s older population grows dramatically. News release. National Institute on Aging. Published March 28, 2016. Accessed December 23, 2022. http://www.nih.gov/news-events/news-releases/worlds-older-population-grows-dramatically
  2. Grundmann S, Ständer S. Chronic pruritus: clinics and treatment. Ann Dermatol. 2011;23:1-11.
  3. Berger TG, Shive M, Harper GM. Pruritus in the older patient: a clinical review. JAMA. 2013;310:2443-2450. doi:10.1001/jama.2013.282023
  4. Valdes-Rodriguez, R, Mollanazar NK, González-Muro J, et al. Itch prevalence and characteristics in a Hispanic geriatric population: a comprehensive study using a standardized itch questionnaire. Acta Derm Venereol. 2015;95:417-421. doi:10.2340/00015555-1968
  5. Li J, Tang H, Hu X, et al. Aquaporin-3 gene and protein expression in sun-protected human skin decreases with skin ageing. Australas J Dermatol. 2010;51:106-112.
  6. Choi EH, Man MQ, Xu P, et al. Stratum corneum acidification is impaired in moderately aged human and murine skin. J Invest Dermatol. 2007;127:2847-2856.
  7. Fenske NA, Lober CW. Structural and functional changes of normal aging skin. J Am Acad Dermatol. 1986;15(4 pt 1):571-585.
  8. Paller AS, Kabashima K, Bieber T. Therapeutic pipeline for atopic dermatitis: end of the drought? J Allergy Clin Immunol. 2017;140:633-643. doi:10.1016/j.jaci.2017.07.006
  9. Kabashima K. New concept of the pathogenesis of atopic dermatitis: interplay among the barrier, allergy, and pruritus as a trinity. J Dermatol Sci. 2013;70:3-11.
  10. Feld M, Garcia R, Buddenkotte J, et al. The pruritus- and TH2-associated cytokine IL-31 promotes growth of sensory nerves. J Allergy Clin Immunol. 2016;138:500-508.
  11. Valdes-Rodriguez R, Stull C, Yosipovitch G. Chronic pruritus in the elderly: pathophysiology, diagnosis and management. Drugs Aging. 2015;32:201-215. doi:10.1007/s40266-015-0246-0
  12. Misery L, Brenaut E, Le Garrec R, et al. Neuropathic pruritus. Nat Rev Neurol. 2014;10:408-416.
  13. Yamaoka H, Sasaki H, Yamasaki H, et al. Truncal pruritus of unknown origin may be a symptom of diabetic polyneuropathy. Diabetes Care. 2010;33:150-155.
  14. Brenaut E, Marcorelles P, Genestet S, et al. Pruritus: an underrecognized symptom of small-fiber neuropathies. J Am Acad Dermatol. 2015;72:328-332.
  15. Adigun M, Badu LA, Berner NM, et al. Uremic pruritus review. US Pharm. 2015;40:HS12-HS15.
  16. Simonsen E, Komenda P, Lerner B, et al. Treatment of uremic pruritus: a systematic review. Am J Kidney Dis. 2017;70:638-655.
  17. Carstens E, Akiyama T, eds. Itch: Mechanisms and Treatment. CRC Press/Taylor & Francis; 2014.
  18. Shirazian S, Aina O, Park Y, et al. Chronic kidney disease-associated pruritus: impact on quality of life and current management challenges. Int J Nephrol Renovasc Dis. 2017;10:11-26.
  19. Brummer GC, Wang LT, Sontheimer RD. A possible role for dupilumab (Dupixent) in the management of idiopathic chronic eczematous eruption of aging. Dermatol Online J. 2018;24:13030/qt55z1f6xh.
  20. Silverberg JI, Brieva J. A successful case of dupilumab treatment for severe uremic pruritus. JAAD Case Rep. 2019;5:339-341.
References
  1. World’s older population grows dramatically. News release. National Institute on Aging. Published March 28, 2016. Accessed December 23, 2022. http://www.nih.gov/news-events/news-releases/worlds-older-population-grows-dramatically
  2. Grundmann S, Ständer S. Chronic pruritus: clinics and treatment. Ann Dermatol. 2011;23:1-11.
  3. Berger TG, Shive M, Harper GM. Pruritus in the older patient: a clinical review. JAMA. 2013;310:2443-2450. doi:10.1001/jama.2013.282023
  4. Valdes-Rodriguez, R, Mollanazar NK, González-Muro J, et al. Itch prevalence and characteristics in a Hispanic geriatric population: a comprehensive study using a standardized itch questionnaire. Acta Derm Venereol. 2015;95:417-421. doi:10.2340/00015555-1968
  5. Li J, Tang H, Hu X, et al. Aquaporin-3 gene and protein expression in sun-protected human skin decreases with skin ageing. Australas J Dermatol. 2010;51:106-112.
  6. Choi EH, Man MQ, Xu P, et al. Stratum corneum acidification is impaired in moderately aged human and murine skin. J Invest Dermatol. 2007;127:2847-2856.
  7. Fenske NA, Lober CW. Structural and functional changes of normal aging skin. J Am Acad Dermatol. 1986;15(4 pt 1):571-585.
  8. Paller AS, Kabashima K, Bieber T. Therapeutic pipeline for atopic dermatitis: end of the drought? J Allergy Clin Immunol. 2017;140:633-643. doi:10.1016/j.jaci.2017.07.006
  9. Kabashima K. New concept of the pathogenesis of atopic dermatitis: interplay among the barrier, allergy, and pruritus as a trinity. J Dermatol Sci. 2013;70:3-11.
  10. Feld M, Garcia R, Buddenkotte J, et al. The pruritus- and TH2-associated cytokine IL-31 promotes growth of sensory nerves. J Allergy Clin Immunol. 2016;138:500-508.
  11. Valdes-Rodriguez R, Stull C, Yosipovitch G. Chronic pruritus in the elderly: pathophysiology, diagnosis and management. Drugs Aging. 2015;32:201-215. doi:10.1007/s40266-015-0246-0
  12. Misery L, Brenaut E, Le Garrec R, et al. Neuropathic pruritus. Nat Rev Neurol. 2014;10:408-416.
  13. Yamaoka H, Sasaki H, Yamasaki H, et al. Truncal pruritus of unknown origin may be a symptom of diabetic polyneuropathy. Diabetes Care. 2010;33:150-155.
  14. Brenaut E, Marcorelles P, Genestet S, et al. Pruritus: an underrecognized symptom of small-fiber neuropathies. J Am Acad Dermatol. 2015;72:328-332.
  15. Adigun M, Badu LA, Berner NM, et al. Uremic pruritus review. US Pharm. 2015;40:HS12-HS15.
  16. Simonsen E, Komenda P, Lerner B, et al. Treatment of uremic pruritus: a systematic review. Am J Kidney Dis. 2017;70:638-655.
  17. Carstens E, Akiyama T, eds. Itch: Mechanisms and Treatment. CRC Press/Taylor & Francis; 2014.
  18. Shirazian S, Aina O, Park Y, et al. Chronic kidney disease-associated pruritus: impact on quality of life and current management challenges. Int J Nephrol Renovasc Dis. 2017;10:11-26.
  19. Brummer GC, Wang LT, Sontheimer RD. A possible role for dupilumab (Dupixent) in the management of idiopathic chronic eczematous eruption of aging. Dermatol Online J. 2018;24:13030/qt55z1f6xh.
  20. Silverberg JI, Brieva J. A successful case of dupilumab treatment for severe uremic pruritus. JAAD Case Rep. 2019;5:339-341.
Issue
Cutis - 110(6)
Issue
Cutis - 110(6)
Page Number
E31-E32
Page Number
E31-E32
Publications
Publications
Topics
Article Type
Display Headline
Use of Dupilumab in Severe, Multifactorial, Chronic Itch for Geriatric Patients
Display Headline
Use of Dupilumab in Severe, Multifactorial, Chronic Itch for Geriatric Patients
Sections
Inside the Article

PRACTICE POINTS

  • A series of age-related mechanisms within the epidermis, underlying neuropathy, medication side effects, infection, malignancy, thyroid dysregulation, liver disease, and chronic kidney disease may contribute to pruritus in elderly patients.
  • Patients with mild kidney disease may still experience a recalcitrant and notable itch burden.
  • Dupilumab is efficacious and safe in the management of chronic pruritus, even in complex cases such as elderly patients with multiple comorbidities.
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

Innovations in Dermatology Fall Abstract Compendium

Article Type
Changed
Tue, 01/03/2023 - 15:04
Sponsor
Supported by a educational grants from: La Roche-Posay (a L'Oreal Company) and …
Issue
Cutis - 110(6)
Publications
Sections
Sponsor
Supported by a educational grants from: La Roche-Posay (a L'Oreal Company) and …
Sponsor
Supported by a educational grants from: La Roche-Posay (a L'Oreal Company) and …
Issue
Cutis - 110(6)
Issue
Cutis - 110(6)
Publications
Publications
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 01/03/2023 - 14:45
Un-Gate On Date
Tue, 01/03/2023 - 14:45
Use ProPublica
CFC Schedule Remove Status
Tue, 01/03/2023 - 14:45
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

Rapidly Growing Nodule Within a Previously Radiated Area of the Scalp

Article Type
Changed
Thu, 01/05/2023 - 13:04
Display Headline
Rapidly Growing Nodule Within a Previously Radiated Area of the Scalp

The Diagnosis: Pseudoangiomatous Squamous Cell Carcinoma

Pseudoangiomatous squamous cell carcinoma (PSCC), a variant of acantholytic squamous cell carcinoma (SCC), is a rare epithelial neoplasm that can mimic angiosarcoma.1 Clinically, PSCC presents as a white-gray ulcer or nodular pink tumor on sun-exposed areas, typically on the head and neck. Due to its increased potential for metastasis, this variant of SCC is considered particularly aggressive. Histologically, PSCC shows nests of acantholytic atypical keratinocytes arranged in anastomosing arrays that form pseudovascular or pseudoglandular structures.2 Acantholytic spaces frequently are filled with erythrocytes. Immunohistochemically, PSCC tumor cells express classic squamous markers such as cytokeratin (CK) 5 and p63 but not vascular markers such as CD31, CD34, and von Willebrand factor.3 In our patient, histopathology of the lesion revealed invasive nests, lobules, and interconnected columns of well-differentiated squamous tumor cells that emanated from the base of the epidermis. The tumor exhibited acantholysis forming ectatic and slitlike spaces, some of which contained erythrocytes. The neoplastic cells, including those lining pseudovascular spaces, positively stained for CK5 (Figure 1A) and nuclear p63 but lacked reactivity to CD31 (Figure 1B) and CD34, corroborating squamous and not vascular differentiation. Current treatment guidelines include Mohs micrographic surgery, excisional surgery, or radiation.4 Our patient’s lesion was completely removed by Mohs micrographic surgery. Three months later, there was no evidence of recurrence.

Pseudovascular (pseudoangiomatous) squamous cell carcinoma.
FIGURE 1. Pseudovascular (pseudoangiomatous) squamous cell carcinoma. A, The tumor cells exhibited strong cytoplasmic immunostaining to cytokeratin 5 (original magnification ×20). B, The vascular marker CD31 labeled background vasculature but not tumor cells (original magnification ×20).

Angiosarcoma is an aggressive neoplasm associated with a poor prognosis and 5-year survival rate of 30% to 40%. The etiology of angiosarcoma still is unclear, but identified risk factors include prior radiation therapy, lymphedema (Stewart-Treves syndrome), and genetic predisposition.5 In the skin, angiosarcoma often occurs in the head and neck region, accounting for 60% of cutaneous cases.5,6 Early in the disease, most patients present with a bruiselike lesion on the scalp or forehead, often delaying the diagnosis.6 As the cancer progresses, tissue infiltration, edema, and hemorrhage contribute to the formation of violaceous nodules, which eventually prompt for biopsy. Angiosarcoma spans a broad histologic spectrum depending on the cytology of malignant cells (eg, spindle, small round, epithelioid) and their capacity for vasoformation. Welldifferentiated angiosarcoma shows retiform slitlike spaces in between collagen bundles that are lined by hyperchromatic hobnailing endothelial cells (Figure 2).7 Epithelioid angiosarcoma can be mistaken for SCC.8 Immunohistochemically, angiosarcoma stains positively for CD31, CD34, ETS-related gene 1, D2-40, and factor VIII.9 In our patient, the neoplasm was negative for vascular markers CD31 and CD34.

Angiosarcoma
FIGURE 2. Angiosarcoma. Nodular deposition of spindled cells forming poorly defined vascular spaces in the superficial dermis as well as fascicular and infiltrative growth in the mid to deep dermis are present (H&E, original magnification ×3). Atypical cells show crowding, stacking, scattered mitoses as well as nuclear hyperchromasia (H&E, original magnification ×20 [inset]).

Bacillary angiomatosis (BA), caused by Bartonella henselae, is a rare disease that first was identified in HIV patients with diminished CD4+ T-cell counts. In the skin, BA often manifests as centrally ulcerated, single or clustered, reddish-purple nodules.10 Histologically, it is characterized by highly vascularized, histiocyterich infiltrates with admixed neutrophils and plasma cells (Figure 3). Capillaries often proliferate in a lobular fashion.11 Atypical cytology with areas of necrosis may mimic angiosarcoma.12 The pathognomonic feature of BA is the presence of enlarged histiocytes with pink-purplish cytoplasm corresponding to intracytoplasmic aggregates of bacteria, which can be revealed by Warthin-Starry or Grocott-Gomori methenamine-silver staining. Immunohistochemically, proliferative benign capillaries are highlighted by CD34 and CD31, and histiocytes are decorated by CD68.12 This diagnosis was excluded based on the patient’s history, clinical presentation, and positive staining for CK5 and p63.

Bacillary angiomatosis
FIGURE 3. Bacillary angiomatosis. The dermis contains a nodular infiltrate of histiocytes, lymphocytes, neutrophils, and vascular spaces with erythrocytes (H&E, original magnification ×3). Pink-red intracellular aggregates of gram-negative Bartonella henselae can be seen (arrow) (H&E, original magnification ×20 [inset]).

Squamoid eccrine ductal carcinoma is an exceedingly rare subtype of eccrine carcinoma that mimics SCC both clinically and histologically.13 It most often occurs on the head and neck of elderly patients. This neoplasm can look similar to SCC and its variants, including PSCC. Histologically, squamoid eccrine ductal carcinoma exhibits a biphasic growth pattern.14 Well-differentiated squamous dysplasia transitions to carcinoma with eccrine duct formation as the tumor percolates deep into the dermis (Figure 4). As a result, superficial skin biopsies often lead to an incorrect diagnosis.15 Unlike SCC, the risk for locoregional and widespread metastasis is elevated. Identifying ducts in the deep aspect of the tumor is critical, thus immunohistochemical staining for carcinoembryonic antigen and epithelial membrane antigen is paramount for the diagnosis.15 Pseudoangiomatous SCC will stain negative for carcinoembryonic antigen, as was the case in our patient.

Squamoid eccrine ductal carcinoma
FIGURE 4. Squamoid eccrine ductal carcinoma. Cytologically atypical squamoid epithelium arrayed in irregularly shaped nests, cords, and as individual cells can be seen. Within the tumor nests are multiple, variably ectatic ductal structures joined by small intracytoplasmic microductules (H&E, original magnification ×3).

Pseudoepitheliomatous hyperplasia is a benign histologic reaction that can result from trauma, chronic inflammation (ie, pyoderma gangrenosum), tattoo placement, underlying neoplasia or fungal infection, or a spider bite reaction.14,15 It most commonly is seen as a well-demarcated nodule or plaque associated with scaling or crusting. Papules vary in size from less than 1 cm to several centimeters. Histologically, it is defined by an acanthotic proliferation of the adnexal epithelium and epidermis (Figure 5).16,17 Irregular strands, cords, and nests of squamoid cells can extend into the dermis.18 It can closely mimic SCC, but there are a few key differences. Pseudoepitheliomatous hyperplasia will not display atypical mitotic figures or atypical nuclei and will never invade lymphatics or vascular systems.19 Pseudoepitheliomatous hyperplasia shows identical histology to well-differentiated SCC, and thus clinicopathologic correlation and mindful histologic evaluation are crucial. The presence of an increased influx of neutrophils and histiocytes should prompt for microbial stains or deeper sectioning. A superficial biopsy should be followed by a deep biopsy. In our patient, microorganismal stains were negative.

Pseudoepitheliomatous hyperplasia secondary to phaeohyphomycosis
FIGURE 5. Pseudoepitheliomatous hyperplasia secondary to phaeohyphomycosis. Irregular squamous epithelial hyperplasia in the background of focal suppurative inflammation can be seen (H&E, original magnification ×3). Numerous melanin-pigmented hyphae and scattered yeasts (arrow) are evident (H&E, original magnification ×60 [inset]). Reference bar indicates 500 μm.

References
  1. Kiyohara T, Miyamoto M, Shijimaya T, et al. Pseudovascular squamous cell carcinoma: a review of the published work and reassessment of prognosis. J Dermatol. 2018;45:1448-1451.
  2. Nagore E, Sánchez-Motilla JM, Pérez-Vallés A, et al. Pseudovascular squamous cell carcinoma of the skin. Clin Exp Dermatol. 2000;25:206-208.
  3. Han X, Lin X, Shao X. Pseudovascular adenoid squamous cell carcinoma of the tongue: a case report and literature review. Int J Clin Exp Pathol. 2020;13:1086-1089.
  4. Singh S, Bisht N, Purkayastha A, et al. Acantholytic squamous cell carcinoma of the scalp in an elderly patient treated with radical radiotherapy. J Cancer Res Pract. 2018;5:165-168.
  5. Cao J, Wang J, He C, et al. Angiosarcoma: a review of diagnosis and current treatment. Am J Cancer Res. 2019;9:2303-2313.
  6. Buehler D, Rice SR, Moody JS, et al. Angiosarcoma outcomes and prognostic factors: a 25-year single institution experience. Am J Clin Oncol. 2014;37:473-479.
  7. Ronen S, Ivan D, Torres-Cabala CA, et al. Post‐radiation vascular lesions of the breast. J Cutan Pathol. 2019;46:52-58.
  8. Shilpa K, Leelavathy B, Gorur D, et al. Early-onset epithelioid angiosarcoma: diagnostic enigma, a rare case report. Indian J Dermatopathol Diagn Dermatol. 2019;6:36-38.
  9. Gaballah AH, Jensen CT, Palmquist S, et al. Angiosarcoma: clinical and imaging features from head to toe [published online May 4, 2017]. Br J Radiol. 2017;90:20170039. doi:10.1259/bjr.20170039
  10. Hoffman CF, Papadopoulos D, Palmer DM, et al. A case report of bacillary angiomatosis in a patient infected with human immunodeficiency virus. Cutis. 2002;69:175-178.
  11. Biwer E, Uerlich M, Wimheuer R, et al. Bacillary angiomatosis: an important differential diagnosis in patients with HIV. Am J Dermatopathol. 1994;16:110.
  12. Medeiros LJ, Miranda RN. Bacillary angiomatosis. In: Medeiros LJ, Miranda RN, eds. Diagnostic Pathology: Lymph Nodes and Extranodal Lymphomas. 2nd ed. Elsevier; 2018:58-63.
  13. van der Horst MP, Garcia-Herrera A, Markiewicz D, et al. Squamoid eccrine ductal carcinoma: a clinicopathologic study of 30 cases. Am J Surg Pathol. 2016;40:755-760.
  14. Mckissack S, Wohltmann W, Dalton S, et al. Squamoid eccrine ductal carcinoma: an aggressive mimicker of squamous cell carcinoma. Am J Dermatopathol. 2019;41:140-143.
  15. Wollina U. Pyoderma gangrenosum—a review. Orphanet J Rare Dis. 2007;2:19
  16. Chow P, Goddard L, Greenway H, et al. Squamoid eccrine ductal carcinoma: the Scripps experience. Dermatol Surg. 2021;47:1115-1117.
  17. Zayour M, Lazova R. Pseudoepitheliomatous hyperplasia: a review. Am J Dermatopathol. 2011;33:112-122; quiz 123-126.
  18. Lynch JM. Understanding pseudoepitheliomatous hyperplasia. Pathol Case Rev. 2004;9:36-45.
  19. Goel R, Wallace ML. Pseudoepitheliomatous hyperplasia secondary to cutaneous aspergillus. Am J Dermatopathol. 2001;23:224-226.
Article PDF
Author and Disclosure Information

Mr. Thompson and Ms. Mattia are from Florida State University College of Medicine, Tallahassee. Drs. Dolson, Schaffer, and Green are from Dermatology Associates of Tallahassee.

The authors report no conflict of interest.

Correspondence: William Harris Green, MD, Dermatology Associates of Tallahassee, 1707 Riggins Rd, Tallahassee, FL 32308 ([email protected]).

Issue
Cutis - 111(1)
Publications
Topics
Page Number
40,46-48
Sections
Author and Disclosure Information

Mr. Thompson and Ms. Mattia are from Florida State University College of Medicine, Tallahassee. Drs. Dolson, Schaffer, and Green are from Dermatology Associates of Tallahassee.

The authors report no conflict of interest.

Correspondence: William Harris Green, MD, Dermatology Associates of Tallahassee, 1707 Riggins Rd, Tallahassee, FL 32308 ([email protected]).

Author and Disclosure Information

Mr. Thompson and Ms. Mattia are from Florida State University College of Medicine, Tallahassee. Drs. Dolson, Schaffer, and Green are from Dermatology Associates of Tallahassee.

The authors report no conflict of interest.

Correspondence: William Harris Green, MD, Dermatology Associates of Tallahassee, 1707 Riggins Rd, Tallahassee, FL 32308 ([email protected]).

Article PDF
Article PDF
Related Articles

The Diagnosis: Pseudoangiomatous Squamous Cell Carcinoma

Pseudoangiomatous squamous cell carcinoma (PSCC), a variant of acantholytic squamous cell carcinoma (SCC), is a rare epithelial neoplasm that can mimic angiosarcoma.1 Clinically, PSCC presents as a white-gray ulcer or nodular pink tumor on sun-exposed areas, typically on the head and neck. Due to its increased potential for metastasis, this variant of SCC is considered particularly aggressive. Histologically, PSCC shows nests of acantholytic atypical keratinocytes arranged in anastomosing arrays that form pseudovascular or pseudoglandular structures.2 Acantholytic spaces frequently are filled with erythrocytes. Immunohistochemically, PSCC tumor cells express classic squamous markers such as cytokeratin (CK) 5 and p63 but not vascular markers such as CD31, CD34, and von Willebrand factor.3 In our patient, histopathology of the lesion revealed invasive nests, lobules, and interconnected columns of well-differentiated squamous tumor cells that emanated from the base of the epidermis. The tumor exhibited acantholysis forming ectatic and slitlike spaces, some of which contained erythrocytes. The neoplastic cells, including those lining pseudovascular spaces, positively stained for CK5 (Figure 1A) and nuclear p63 but lacked reactivity to CD31 (Figure 1B) and CD34, corroborating squamous and not vascular differentiation. Current treatment guidelines include Mohs micrographic surgery, excisional surgery, or radiation.4 Our patient’s lesion was completely removed by Mohs micrographic surgery. Three months later, there was no evidence of recurrence.

Pseudovascular (pseudoangiomatous) squamous cell carcinoma.
FIGURE 1. Pseudovascular (pseudoangiomatous) squamous cell carcinoma. A, The tumor cells exhibited strong cytoplasmic immunostaining to cytokeratin 5 (original magnification ×20). B, The vascular marker CD31 labeled background vasculature but not tumor cells (original magnification ×20).

Angiosarcoma is an aggressive neoplasm associated with a poor prognosis and 5-year survival rate of 30% to 40%. The etiology of angiosarcoma still is unclear, but identified risk factors include prior radiation therapy, lymphedema (Stewart-Treves syndrome), and genetic predisposition.5 In the skin, angiosarcoma often occurs in the head and neck region, accounting for 60% of cutaneous cases.5,6 Early in the disease, most patients present with a bruiselike lesion on the scalp or forehead, often delaying the diagnosis.6 As the cancer progresses, tissue infiltration, edema, and hemorrhage contribute to the formation of violaceous nodules, which eventually prompt for biopsy. Angiosarcoma spans a broad histologic spectrum depending on the cytology of malignant cells (eg, spindle, small round, epithelioid) and their capacity for vasoformation. Welldifferentiated angiosarcoma shows retiform slitlike spaces in between collagen bundles that are lined by hyperchromatic hobnailing endothelial cells (Figure 2).7 Epithelioid angiosarcoma can be mistaken for SCC.8 Immunohistochemically, angiosarcoma stains positively for CD31, CD34, ETS-related gene 1, D2-40, and factor VIII.9 In our patient, the neoplasm was negative for vascular markers CD31 and CD34.

Angiosarcoma
FIGURE 2. Angiosarcoma. Nodular deposition of spindled cells forming poorly defined vascular spaces in the superficial dermis as well as fascicular and infiltrative growth in the mid to deep dermis are present (H&E, original magnification ×3). Atypical cells show crowding, stacking, scattered mitoses as well as nuclear hyperchromasia (H&E, original magnification ×20 [inset]).

Bacillary angiomatosis (BA), caused by Bartonella henselae, is a rare disease that first was identified in HIV patients with diminished CD4+ T-cell counts. In the skin, BA often manifests as centrally ulcerated, single or clustered, reddish-purple nodules.10 Histologically, it is characterized by highly vascularized, histiocyterich infiltrates with admixed neutrophils and plasma cells (Figure 3). Capillaries often proliferate in a lobular fashion.11 Atypical cytology with areas of necrosis may mimic angiosarcoma.12 The pathognomonic feature of BA is the presence of enlarged histiocytes with pink-purplish cytoplasm corresponding to intracytoplasmic aggregates of bacteria, which can be revealed by Warthin-Starry or Grocott-Gomori methenamine-silver staining. Immunohistochemically, proliferative benign capillaries are highlighted by CD34 and CD31, and histiocytes are decorated by CD68.12 This diagnosis was excluded based on the patient’s history, clinical presentation, and positive staining for CK5 and p63.

Bacillary angiomatosis
FIGURE 3. Bacillary angiomatosis. The dermis contains a nodular infiltrate of histiocytes, lymphocytes, neutrophils, and vascular spaces with erythrocytes (H&E, original magnification ×3). Pink-red intracellular aggregates of gram-negative Bartonella henselae can be seen (arrow) (H&E, original magnification ×20 [inset]).

Squamoid eccrine ductal carcinoma is an exceedingly rare subtype of eccrine carcinoma that mimics SCC both clinically and histologically.13 It most often occurs on the head and neck of elderly patients. This neoplasm can look similar to SCC and its variants, including PSCC. Histologically, squamoid eccrine ductal carcinoma exhibits a biphasic growth pattern.14 Well-differentiated squamous dysplasia transitions to carcinoma with eccrine duct formation as the tumor percolates deep into the dermis (Figure 4). As a result, superficial skin biopsies often lead to an incorrect diagnosis.15 Unlike SCC, the risk for locoregional and widespread metastasis is elevated. Identifying ducts in the deep aspect of the tumor is critical, thus immunohistochemical staining for carcinoembryonic antigen and epithelial membrane antigen is paramount for the diagnosis.15 Pseudoangiomatous SCC will stain negative for carcinoembryonic antigen, as was the case in our patient.

Squamoid eccrine ductal carcinoma
FIGURE 4. Squamoid eccrine ductal carcinoma. Cytologically atypical squamoid epithelium arrayed in irregularly shaped nests, cords, and as individual cells can be seen. Within the tumor nests are multiple, variably ectatic ductal structures joined by small intracytoplasmic microductules (H&E, original magnification ×3).

Pseudoepitheliomatous hyperplasia is a benign histologic reaction that can result from trauma, chronic inflammation (ie, pyoderma gangrenosum), tattoo placement, underlying neoplasia or fungal infection, or a spider bite reaction.14,15 It most commonly is seen as a well-demarcated nodule or plaque associated with scaling or crusting. Papules vary in size from less than 1 cm to several centimeters. Histologically, it is defined by an acanthotic proliferation of the adnexal epithelium and epidermis (Figure 5).16,17 Irregular strands, cords, and nests of squamoid cells can extend into the dermis.18 It can closely mimic SCC, but there are a few key differences. Pseudoepitheliomatous hyperplasia will not display atypical mitotic figures or atypical nuclei and will never invade lymphatics or vascular systems.19 Pseudoepitheliomatous hyperplasia shows identical histology to well-differentiated SCC, and thus clinicopathologic correlation and mindful histologic evaluation are crucial. The presence of an increased influx of neutrophils and histiocytes should prompt for microbial stains or deeper sectioning. A superficial biopsy should be followed by a deep biopsy. In our patient, microorganismal stains were negative.

Pseudoepitheliomatous hyperplasia secondary to phaeohyphomycosis
FIGURE 5. Pseudoepitheliomatous hyperplasia secondary to phaeohyphomycosis. Irregular squamous epithelial hyperplasia in the background of focal suppurative inflammation can be seen (H&E, original magnification ×3). Numerous melanin-pigmented hyphae and scattered yeasts (arrow) are evident (H&E, original magnification ×60 [inset]). Reference bar indicates 500 μm.

The Diagnosis: Pseudoangiomatous Squamous Cell Carcinoma

Pseudoangiomatous squamous cell carcinoma (PSCC), a variant of acantholytic squamous cell carcinoma (SCC), is a rare epithelial neoplasm that can mimic angiosarcoma.1 Clinically, PSCC presents as a white-gray ulcer or nodular pink tumor on sun-exposed areas, typically on the head and neck. Due to its increased potential for metastasis, this variant of SCC is considered particularly aggressive. Histologically, PSCC shows nests of acantholytic atypical keratinocytes arranged in anastomosing arrays that form pseudovascular or pseudoglandular structures.2 Acantholytic spaces frequently are filled with erythrocytes. Immunohistochemically, PSCC tumor cells express classic squamous markers such as cytokeratin (CK) 5 and p63 but not vascular markers such as CD31, CD34, and von Willebrand factor.3 In our patient, histopathology of the lesion revealed invasive nests, lobules, and interconnected columns of well-differentiated squamous tumor cells that emanated from the base of the epidermis. The tumor exhibited acantholysis forming ectatic and slitlike spaces, some of which contained erythrocytes. The neoplastic cells, including those lining pseudovascular spaces, positively stained for CK5 (Figure 1A) and nuclear p63 but lacked reactivity to CD31 (Figure 1B) and CD34, corroborating squamous and not vascular differentiation. Current treatment guidelines include Mohs micrographic surgery, excisional surgery, or radiation.4 Our patient’s lesion was completely removed by Mohs micrographic surgery. Three months later, there was no evidence of recurrence.

Pseudovascular (pseudoangiomatous) squamous cell carcinoma.
FIGURE 1. Pseudovascular (pseudoangiomatous) squamous cell carcinoma. A, The tumor cells exhibited strong cytoplasmic immunostaining to cytokeratin 5 (original magnification ×20). B, The vascular marker CD31 labeled background vasculature but not tumor cells (original magnification ×20).

Angiosarcoma is an aggressive neoplasm associated with a poor prognosis and 5-year survival rate of 30% to 40%. The etiology of angiosarcoma still is unclear, but identified risk factors include prior radiation therapy, lymphedema (Stewart-Treves syndrome), and genetic predisposition.5 In the skin, angiosarcoma often occurs in the head and neck region, accounting for 60% of cutaneous cases.5,6 Early in the disease, most patients present with a bruiselike lesion on the scalp or forehead, often delaying the diagnosis.6 As the cancer progresses, tissue infiltration, edema, and hemorrhage contribute to the formation of violaceous nodules, which eventually prompt for biopsy. Angiosarcoma spans a broad histologic spectrum depending on the cytology of malignant cells (eg, spindle, small round, epithelioid) and their capacity for vasoformation. Welldifferentiated angiosarcoma shows retiform slitlike spaces in between collagen bundles that are lined by hyperchromatic hobnailing endothelial cells (Figure 2).7 Epithelioid angiosarcoma can be mistaken for SCC.8 Immunohistochemically, angiosarcoma stains positively for CD31, CD34, ETS-related gene 1, D2-40, and factor VIII.9 In our patient, the neoplasm was negative for vascular markers CD31 and CD34.

Angiosarcoma
FIGURE 2. Angiosarcoma. Nodular deposition of spindled cells forming poorly defined vascular spaces in the superficial dermis as well as fascicular and infiltrative growth in the mid to deep dermis are present (H&E, original magnification ×3). Atypical cells show crowding, stacking, scattered mitoses as well as nuclear hyperchromasia (H&E, original magnification ×20 [inset]).

Bacillary angiomatosis (BA), caused by Bartonella henselae, is a rare disease that first was identified in HIV patients with diminished CD4+ T-cell counts. In the skin, BA often manifests as centrally ulcerated, single or clustered, reddish-purple nodules.10 Histologically, it is characterized by highly vascularized, histiocyterich infiltrates with admixed neutrophils and plasma cells (Figure 3). Capillaries often proliferate in a lobular fashion.11 Atypical cytology with areas of necrosis may mimic angiosarcoma.12 The pathognomonic feature of BA is the presence of enlarged histiocytes with pink-purplish cytoplasm corresponding to intracytoplasmic aggregates of bacteria, which can be revealed by Warthin-Starry or Grocott-Gomori methenamine-silver staining. Immunohistochemically, proliferative benign capillaries are highlighted by CD34 and CD31, and histiocytes are decorated by CD68.12 This diagnosis was excluded based on the patient’s history, clinical presentation, and positive staining for CK5 and p63.

Bacillary angiomatosis
FIGURE 3. Bacillary angiomatosis. The dermis contains a nodular infiltrate of histiocytes, lymphocytes, neutrophils, and vascular spaces with erythrocytes (H&E, original magnification ×3). Pink-red intracellular aggregates of gram-negative Bartonella henselae can be seen (arrow) (H&E, original magnification ×20 [inset]).

Squamoid eccrine ductal carcinoma is an exceedingly rare subtype of eccrine carcinoma that mimics SCC both clinically and histologically.13 It most often occurs on the head and neck of elderly patients. This neoplasm can look similar to SCC and its variants, including PSCC. Histologically, squamoid eccrine ductal carcinoma exhibits a biphasic growth pattern.14 Well-differentiated squamous dysplasia transitions to carcinoma with eccrine duct formation as the tumor percolates deep into the dermis (Figure 4). As a result, superficial skin biopsies often lead to an incorrect diagnosis.15 Unlike SCC, the risk for locoregional and widespread metastasis is elevated. Identifying ducts in the deep aspect of the tumor is critical, thus immunohistochemical staining for carcinoembryonic antigen and epithelial membrane antigen is paramount for the diagnosis.15 Pseudoangiomatous SCC will stain negative for carcinoembryonic antigen, as was the case in our patient.

Squamoid eccrine ductal carcinoma
FIGURE 4. Squamoid eccrine ductal carcinoma. Cytologically atypical squamoid epithelium arrayed in irregularly shaped nests, cords, and as individual cells can be seen. Within the tumor nests are multiple, variably ectatic ductal structures joined by small intracytoplasmic microductules (H&E, original magnification ×3).

Pseudoepitheliomatous hyperplasia is a benign histologic reaction that can result from trauma, chronic inflammation (ie, pyoderma gangrenosum), tattoo placement, underlying neoplasia or fungal infection, or a spider bite reaction.14,15 It most commonly is seen as a well-demarcated nodule or plaque associated with scaling or crusting. Papules vary in size from less than 1 cm to several centimeters. Histologically, it is defined by an acanthotic proliferation of the adnexal epithelium and epidermis (Figure 5).16,17 Irregular strands, cords, and nests of squamoid cells can extend into the dermis.18 It can closely mimic SCC, but there are a few key differences. Pseudoepitheliomatous hyperplasia will not display atypical mitotic figures or atypical nuclei and will never invade lymphatics or vascular systems.19 Pseudoepitheliomatous hyperplasia shows identical histology to well-differentiated SCC, and thus clinicopathologic correlation and mindful histologic evaluation are crucial. The presence of an increased influx of neutrophils and histiocytes should prompt for microbial stains or deeper sectioning. A superficial biopsy should be followed by a deep biopsy. In our patient, microorganismal stains were negative.

Pseudoepitheliomatous hyperplasia secondary to phaeohyphomycosis
FIGURE 5. Pseudoepitheliomatous hyperplasia secondary to phaeohyphomycosis. Irregular squamous epithelial hyperplasia in the background of focal suppurative inflammation can be seen (H&E, original magnification ×3). Numerous melanin-pigmented hyphae and scattered yeasts (arrow) are evident (H&E, original magnification ×60 [inset]). Reference bar indicates 500 μm.

References
  1. Kiyohara T, Miyamoto M, Shijimaya T, et al. Pseudovascular squamous cell carcinoma: a review of the published work and reassessment of prognosis. J Dermatol. 2018;45:1448-1451.
  2. Nagore E, Sánchez-Motilla JM, Pérez-Vallés A, et al. Pseudovascular squamous cell carcinoma of the skin. Clin Exp Dermatol. 2000;25:206-208.
  3. Han X, Lin X, Shao X. Pseudovascular adenoid squamous cell carcinoma of the tongue: a case report and literature review. Int J Clin Exp Pathol. 2020;13:1086-1089.
  4. Singh S, Bisht N, Purkayastha A, et al. Acantholytic squamous cell carcinoma of the scalp in an elderly patient treated with radical radiotherapy. J Cancer Res Pract. 2018;5:165-168.
  5. Cao J, Wang J, He C, et al. Angiosarcoma: a review of diagnosis and current treatment. Am J Cancer Res. 2019;9:2303-2313.
  6. Buehler D, Rice SR, Moody JS, et al. Angiosarcoma outcomes and prognostic factors: a 25-year single institution experience. Am J Clin Oncol. 2014;37:473-479.
  7. Ronen S, Ivan D, Torres-Cabala CA, et al. Post‐radiation vascular lesions of the breast. J Cutan Pathol. 2019;46:52-58.
  8. Shilpa K, Leelavathy B, Gorur D, et al. Early-onset epithelioid angiosarcoma: diagnostic enigma, a rare case report. Indian J Dermatopathol Diagn Dermatol. 2019;6:36-38.
  9. Gaballah AH, Jensen CT, Palmquist S, et al. Angiosarcoma: clinical and imaging features from head to toe [published online May 4, 2017]. Br J Radiol. 2017;90:20170039. doi:10.1259/bjr.20170039
  10. Hoffman CF, Papadopoulos D, Palmer DM, et al. A case report of bacillary angiomatosis in a patient infected with human immunodeficiency virus. Cutis. 2002;69:175-178.
  11. Biwer E, Uerlich M, Wimheuer R, et al. Bacillary angiomatosis: an important differential diagnosis in patients with HIV. Am J Dermatopathol. 1994;16:110.
  12. Medeiros LJ, Miranda RN. Bacillary angiomatosis. In: Medeiros LJ, Miranda RN, eds. Diagnostic Pathology: Lymph Nodes and Extranodal Lymphomas. 2nd ed. Elsevier; 2018:58-63.
  13. van der Horst MP, Garcia-Herrera A, Markiewicz D, et al. Squamoid eccrine ductal carcinoma: a clinicopathologic study of 30 cases. Am J Surg Pathol. 2016;40:755-760.
  14. Mckissack S, Wohltmann W, Dalton S, et al. Squamoid eccrine ductal carcinoma: an aggressive mimicker of squamous cell carcinoma. Am J Dermatopathol. 2019;41:140-143.
  15. Wollina U. Pyoderma gangrenosum—a review. Orphanet J Rare Dis. 2007;2:19
  16. Chow P, Goddard L, Greenway H, et al. Squamoid eccrine ductal carcinoma: the Scripps experience. Dermatol Surg. 2021;47:1115-1117.
  17. Zayour M, Lazova R. Pseudoepitheliomatous hyperplasia: a review. Am J Dermatopathol. 2011;33:112-122; quiz 123-126.
  18. Lynch JM. Understanding pseudoepitheliomatous hyperplasia. Pathol Case Rev. 2004;9:36-45.
  19. Goel R, Wallace ML. Pseudoepitheliomatous hyperplasia secondary to cutaneous aspergillus. Am J Dermatopathol. 2001;23:224-226.
References
  1. Kiyohara T, Miyamoto M, Shijimaya T, et al. Pseudovascular squamous cell carcinoma: a review of the published work and reassessment of prognosis. J Dermatol. 2018;45:1448-1451.
  2. Nagore E, Sánchez-Motilla JM, Pérez-Vallés A, et al. Pseudovascular squamous cell carcinoma of the skin. Clin Exp Dermatol. 2000;25:206-208.
  3. Han X, Lin X, Shao X. Pseudovascular adenoid squamous cell carcinoma of the tongue: a case report and literature review. Int J Clin Exp Pathol. 2020;13:1086-1089.
  4. Singh S, Bisht N, Purkayastha A, et al. Acantholytic squamous cell carcinoma of the scalp in an elderly patient treated with radical radiotherapy. J Cancer Res Pract. 2018;5:165-168.
  5. Cao J, Wang J, He C, et al. Angiosarcoma: a review of diagnosis and current treatment. Am J Cancer Res. 2019;9:2303-2313.
  6. Buehler D, Rice SR, Moody JS, et al. Angiosarcoma outcomes and prognostic factors: a 25-year single institution experience. Am J Clin Oncol. 2014;37:473-479.
  7. Ronen S, Ivan D, Torres-Cabala CA, et al. Post‐radiation vascular lesions of the breast. J Cutan Pathol. 2019;46:52-58.
  8. Shilpa K, Leelavathy B, Gorur D, et al. Early-onset epithelioid angiosarcoma: diagnostic enigma, a rare case report. Indian J Dermatopathol Diagn Dermatol. 2019;6:36-38.
  9. Gaballah AH, Jensen CT, Palmquist S, et al. Angiosarcoma: clinical and imaging features from head to toe [published online May 4, 2017]. Br J Radiol. 2017;90:20170039. doi:10.1259/bjr.20170039
  10. Hoffman CF, Papadopoulos D, Palmer DM, et al. A case report of bacillary angiomatosis in a patient infected with human immunodeficiency virus. Cutis. 2002;69:175-178.
  11. Biwer E, Uerlich M, Wimheuer R, et al. Bacillary angiomatosis: an important differential diagnosis in patients with HIV. Am J Dermatopathol. 1994;16:110.
  12. Medeiros LJ, Miranda RN. Bacillary angiomatosis. In: Medeiros LJ, Miranda RN, eds. Diagnostic Pathology: Lymph Nodes and Extranodal Lymphomas. 2nd ed. Elsevier; 2018:58-63.
  13. van der Horst MP, Garcia-Herrera A, Markiewicz D, et al. Squamoid eccrine ductal carcinoma: a clinicopathologic study of 30 cases. Am J Surg Pathol. 2016;40:755-760.
  14. Mckissack S, Wohltmann W, Dalton S, et al. Squamoid eccrine ductal carcinoma: an aggressive mimicker of squamous cell carcinoma. Am J Dermatopathol. 2019;41:140-143.
  15. Wollina U. Pyoderma gangrenosum—a review. Orphanet J Rare Dis. 2007;2:19
  16. Chow P, Goddard L, Greenway H, et al. Squamoid eccrine ductal carcinoma: the Scripps experience. Dermatol Surg. 2021;47:1115-1117.
  17. Zayour M, Lazova R. Pseudoepitheliomatous hyperplasia: a review. Am J Dermatopathol. 2011;33:112-122; quiz 123-126.
  18. Lynch JM. Understanding pseudoepitheliomatous hyperplasia. Pathol Case Rev. 2004;9:36-45.
  19. Goel R, Wallace ML. Pseudoepitheliomatous hyperplasia secondary to cutaneous aspergillus. Am J Dermatopathol. 2001;23:224-226.
Issue
Cutis - 111(1)
Issue
Cutis - 111(1)
Page Number
40,46-48
Page Number
40,46-48
Publications
Publications
Topics
Article Type
Display Headline
Rapidly Growing Nodule Within a Previously Radiated Area of the Scalp
Display Headline
Rapidly Growing Nodule Within a Previously Radiated Area of the Scalp
Sections
Questionnaire Body

An 84-year-old man with a history of nonmelanoma skin cancer presented to our clinic with a 1.6×1.5-cm exophytic lesion on the left posterior parietal scalp. The lesion nearly doubled in size over the last 4 months. The patient received radiation therapy in this area for the treatment of basal cell carcinoma 7 years prior to presentation. A shave biopsy was performed.

Rapidly growing nodule within a previously radiated area of the scalp

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 01/03/2023 - 09:45
Un-Gate On Date
Tue, 01/03/2023 - 09:45
Use ProPublica
CFC Schedule Remove Status
Tue, 01/03/2023 - 09:45
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

Rituximab Treatment and Improvement of Health-Related Quality of Life in Patients With Pemphigus

Article Type
Changed
Thu, 01/05/2023 - 12:51
Display Headline
Rituximab Treatment and Improvement of Health-Related Quality of Life in Patients With Pemphigus

Pemphigus is a group of autoimmune blistering diseases characterized by the development of painful and flaccid blisters on the skin and/or mucous membranes. Pemphigus vulgaris (PV) and pemphigus foliaceus (PF) are 2 major subtypes and can be distinguished by the location of blister formation or the specificity of autoantibodies directed against different desmogleins.1,2 Although rare, pemphigus is considered a serious and life-threatening condition with a great impact on quality of life (QOL) due to disease symptoms (eg, painful lesions, physical appearance of skin lesions) as well as treatment complications (eg, adverse drug effects, cost of treatment).3-6 Moreover, the physical and psychological effects can lead to marked functional morbidity and work-related disability during patients’ productive years.7 Therefore, affected individuals usually have a remarkably compromised health-related quality of life (HRQOL).8 Effective treatments may considerably improve the QOL of patients with pemphigus.6

Despite the available treatment options, finding the best regimen for pemphigus remains a challenge. Corticosteroids are assumed to be the main treatment, though they have considerable side effects.9,10 Adjuvant therapies are used to suppress or modulate immune responses, leading to remission with the least possible need for corticosteroids. Finding an optimal steroid-sparing agent has been the aim of research, and biologic agents seem to be the best option.8 Rituximab (RTX), an anti-CD20 monoclonal antibody, has shown great promise in several studies of its clinical efficacy and has become a first-line treatment in new guidelines.11-14 Rituximab treatment has been associated with notable improvement in physician-assessed outcome measures with a favorable safety profile in patients with pemphigus.11-15 However, it is important to assess response to treatment from a patient’s perspective through the use of outcome-assessment measures that encompass patient-reported outcomes to reflect the complete patient experience and establish the overall impact of RTX as well as its likelihood of acceptance by patients with pemphigus.

In our study, we compared clinical outcomes and HRQOL through the use of disease-specific measures as well as comprehensive generic health status measures among patients with PV and PF who received RTX treatment 3 months earlier and those who received RTX in the last 2 weeks. The clinical relevance of the patient-reported outcomes is discussed.

MATERIALS AND METHODS

Study Design

We conducted a single-center cross-sectional study of 96 patients with pemphigus aged 18 to 65 years of either sex who were willing to participate in this study. Patients with a confirmed diagnosis of PV or PF who received RTX 3 months earlier or in the last 2 weeks were enrolled in the study. Patients were identified using Dermatry.ir, an archiving software that contains patients’ medical data. Exclusion criteria included lack of sufficient knowledge of the concepts of the questionnaires as well as age younger than 16 years. The study was conducted from October 2019 to April 2020 by the Autoimmune Bullous Disease Research Center at Razi Hospital in Tehran, Iran, which is the main dermatology-specific center and teaching hospital of Iran. The study protocol was approved by the relevant ethics committee.

Patients were categorized into 2 groups: (1) those who received RTX 3 months earlier (3M group); and (2) those who received RTX in the last 2 weeks (R group).

After an explanation of the study to participants, informed written consent was signed by each patient, and their personal data (eg, age, sex, education, marital status, smoking status), as well as clinical data (eg, type of pemphigus, duration of disease, site of onset, prednisolone dosage, presence of Nikolsky sign, anti-DSG1 and anti-DSG3 values, Pemphigus Disease Area Index [PDAI] score, RTX treatment protocol); any known comorbidities such as hypertension, diabetes mellitus, or morbid obesity; and any chronic pulmonary, cardiac, endocrinologic, renal, or hepatic condition, were collected and recorded in a predefined Case Record.

Patient-Reported Outcome Measures

The effect of RTX on QOL in patients with pemphigus was assessed using 2 HRQOL instruments: (1) a general health status indicator, the 36-Item Short Form Survey (SF-36), and (2) a validated, Persian version of a dermatology-specific questionnaire, Dermatology Life Quality Index (DLQI). The questionnaires were completed by each patient or by an assistant if needed.

 

 

The SF-36 is a widely used 36-item questionnaire measuring functional health and well-being across 8 domains—mental health, pain, physical function, role emotional, role physical, social functioning, vitality, and general health perception—with scores for each ranging from 0 to 100. The physical component scores (PCSs) and mental component scores (MCSs) were derived from these 8 subscales, each ranging from 0 to 400, with higher scores indicating better health status.6

The DLQI, one of the most frequently used QOL measures in dermatology, contains 10 questions, each referring to the prior week and classified in the following 6 subscales: symptoms and feelings, daily activities, leisure, personal relationships, work and school, and treatment.16 The total score ranges from 0 (no impact) to 30 (very high impact), with a higher score indicating a lower QOL (eTable 1). The minimal clinically important difference (MCD) for the DLQI was considered to be 2- to 5-point changes in prior studies.17,18 In this study, we used an MCD of a 5-point change or more between study groups.

Classification of Patients Based on DLQI Questionnaire

Moreover, the patient general assessment (PGA) of disease severity was identified using a 3-point scale (1=mild, 2=moderate, 3=severe).

Statistical Analysis

Data were analyzed using SPSS Statistics version 23. P≤.05 was considered significant. Mean and SD were calculated for descriptive data. The t test, Fisher exact test, analysis of variance, multiple regression analysis, and logistic regression analysis were used to identify the relationship between variables.

RESULTS

Patient Characteristics

A total of 96 patients were enrolled in this study. The mean (SD) age of participants was 41.42 (15.1) years (range, 18–58 years). Of 96 patients whose data were included, 55 (57.29%) patients had received RTX 3 months earlier (3M group) and 41 (42.71%) received RTX in the last 2 weeks (R group). A summary of study patient characteristics in each group is provided in eTable 2. There was no significant difference between the 2 groups in terms of age, sex, type of pemphigus, marital status, education, positive Nikolsky sign, smoking status, existence of comorbidities, site of lesions, and RTX treatment protocol. However, a significant difference was found for duration of disease (P=.0124) and mean prednisolone dosage (P=.001) as well as severity of disease measured by PDAI score (P=.003) and anti-DSG1 (P=.003) and anti-DSG3 (P=.021) values.

Summary of Patient Demographic and Clinical Data

Patient-Reported Outcomes

Physical and mental component scores are summarized in eTable 3. Generally, SF-36 scores were improved with RTX treatment in all dimensions except for mental health, though these differences were not statistically significant, with the greatest mean improvement in the role physical index (75.45 in the 3M group vs 53.04 in the R group; P=.009). Mean SF-36 PCS and MCS scores were higher in the 3M group vs the R group, though the difference in MCS score did not reach the level of significance (eTable 3).

Patient-Reported Outcomes

Mean DLQI scores in the R and 3M groups were 12.31 and 6.96, respectively, indicating a considerable burden on HRQOL in both groups. However, a statistically significant difference between these values was seen that also was clinically meaningful, indicating a significant improvement of QOL in patients receiving RTX 3 months earlier (P=.005)(eTable 3).

 

 

The PGA scores indicated that patients in the 3M group were significantly more likely to report less severe disease vs the R group (P=.008)(eTable 3).

Multivariate Analysis—Effect of the patient characteristics and some disease features on indices of QOL was evaluated using the multiple linear regression model. eTable 4 shows the P values of those analyses.

Comparison of QOL Indices Between 2 Study Groups (R and 3M) for Study Variants

COMMENT

Pemphigus is a chronic disabling disease with notable QOL impairment due to disease burden as well as the need for long-term use of immunosuppressive agents during the disease course. To study the effect of RTX on QOL of patients with pemphigus, we compared 2 sets of patients. Prior studies have shown that clinically significant effects of RTX take 4 to 12 weeks to appear.19,20 Therefore, we selected patients who received RTX 3 months earlier to measure their HRQOL indices and compare them with patients who had received RTX in the last 2 weeks as a control group to investigate the effect of RTX intrinsically, as this was the focus of this study.

In our study, one of the research tools was the DLQI. Healthy patients typically have an average score of 0.5.21 The mean DLQI score of the patients in R group was 12.31, which was similar to prior analysis8 and reflects a substantial burden of disease comparable to atopic dermatitis and psoriasis.21,22 In patients in the 3M group, the mean DLQI score was lower than the R group (6.96 vs 12.31), indicating a significant (P=.005) and clinically meaningful improvement in QOL of patients due to the dramatic therapeutic effect of RTX. However, this score indicated a moderate effect on HRQOL, even in the context of clinical improvement due to RTX treatment, which may reflect that the short duration of treatment in the 3M group was a limitation of this study. Although the 12-week treatment duration was comparable with other studies19,20 and major differences in objective measures of treatment efficacy were found in PDAI as well as anti-DSG1 and anti-DSG3 values, longer treatment duration may be needed for a more comprehensive assessment of the benefit of RTX on HRQOL indices in patients with pemphigus.

Based on results of the SF-36 questionnaire, PCS and MCS scores were not substantially impaired in the R group considering the fact that a mean score of 50 has been articulated as a normative value for all scales.23 These data demonstrated the importance of using a dermatologic-specific instrument such as the DLQI instead of a general questionnaire to assess QOL in patients with pemphigus. However, better indices were reported with RTX treatment in the 3 SF-36 domains—role physical (P=.009), role emotional (P=.03), and general health perception (P=.03)—with the role physical showing the greatest magnitude of mean change (75.45 in the 3M group vs 53.04 in the R group). Notably, PCS was impaired to a greater extent than MCS in patients in the R group and showed a greater magnitude of improvement after 3 months of treatment. These results could be explained by the fact that MCS can be largely changed in diseases with a direct effect on the central nervous system.23

Our results also revealed that the dose of corticosteroid correlated to HRQOL of patients with pemphigus who recently received RTX therapy. Indeed, it is more likely that patients on lower-dose prednisolone have a higher QOL, especially on physical function and social function dimensions of SF-36. This finding is highly expectable by less severe disease due to RTX treatment and also lower potential dose-dependent adverse effects of long-term steroid therapy.

One of the most striking findings of this study was the correlation of location of lesions to QOL indices. We found that the mucocutaneous phenotype was significantly correlated to greater improvement in role emotional, role physical, and social functioning scores due to RTX treatment compared with cutaneous or mucosal types (P=.02, P=.025, and P=.017, respectively). Although mucosal involvement of the disease can be the most burdensome feature because of its large impact on essential activities such as eating and speaking, cutaneous lesions with unpleasant appearance and undesirable symptoms may have a similar impact on QOL. Therefore, having both mucosal and cutaneous lesions causes a worsened QOL and decreased treatment efficacy vs having only one area involved. This may explain the greater improvement in some QOL indices with RTX treatment.

Limitations—Given the cross-sectional design of this study in which patients were observed at a single time point during their treatment course, it is not possible to establish a clear cause-effect relationship between variables. Moreover, we did not evaluate the impact of RTX or prednisolone adverse effects on QOL. Therefore, further prospective studies with longer treatment durations may help to validate our findings. In addition, MCDs for DLQI and SF-36 in pemphigus need to be determined and validated in future studies.

CONCLUSION

The results of our study demonstrated that patients with pemphigus may benefit from taking RTX, not only in terms of clinical improvement of their disease measured by objective indices such as PDAI and anti-DSG1 and anti-DSG3 values but also in several domains that are important to patients, including physical and mental health status (SF-36), HRQOL (DLQI), and overall disease severity (PGA). Rituximab administration in patients with pemphigus can lead to rapid and significant improvement in HRQOL as well as patient- and physician-assessed measures. Its favorable safety profile along with its impact on patients’ daily lives and mental health makes RTX a suitable treatment option for patients with pemphigus. Moreover, we recommend taking QOL indices into account while evaluating the efficacy of new medications to improve our insight into the patient experience and provide better patient adherence to treatment, which is an important issue for optimal control of chronic disorders.

References
  1. Hammers CM, Stanley JR. Mechanisms of disease: pemphigus and bullous pemphigoid. Ann Rev Pathol. 2016;11:175-197.
  2. Kasperkiewicz M, Ellebrecht CT, Takahashi H, et al. Pemphigus. Nat Rev Dis Primers. 2017;3:17026.
  3. Mayrshofer F, Hertl M, Sinkgraven R, et al. Significant decrease in quality of life in patients with pemphigus vulgaris, result from the German Bullous Skin Disease (BSD) Study Group. J Dtsch Dermatol Ges. 2005;3:431-435.
  4. Terrab Z, Benckikhi H, Maaroufi A, et al. Quality of life and pemphigus. Ann Dermatol Venereol. 2005;132:321-328.
  5. Tabolli S, Mozzetta A, Antinone V, et al. The health impact of pemphigus vulgaris and pemphigus foliaceus assessed using the Medical Outcomes Study 36-item short form health survey questionnaire. Br J Dermatol. 2008;158:1029-1034.
  6. Paradisi A, Sampogna F, Di Pietro, C, et al. Quality-of-life assessment in patients with pemphigus using a minimum set of evaluation tools. J Am Acad Dermatol. 2009;60:261-269.
  7. Heelan K, Hitzig SL, Knowles S, et al. Loss of work productivity and quality of life in patients with autoimmune bullous dermatoses. J Cutan Med Surg. 2015;19:546-554.
  8. Ghodsi SZ, Chams-Davatchi C, Daneshpazhooh M, et al. Quality of life and psychological status of patients with pemphigus vulgaris using Dermatology Life Quality Index and General Health Questionnaires. J Dermatol. 2012;39:141-144.
  9. Schäcke H, Döcke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther. 2002;96:2343.
  10. Mohammad-Javad N, Parvaneh H, Maryam G, et al. Randomized trial of tacrolimus 0.1% ointment versus triamcinolone acetonide 0.1% paste in the treatment of oral pemphigus vulgaris. Iranian J Dermatol. 2012;15:42-46.
  11. Lunardon L, Tsai KJ, Propert KJ, et al. Adjuvant rituximab therapy of pemphigus: a single-center experience with 31 patients. Arch Dermatol. 2012;148:1031-1036.
  12. Colliou N, Picard D, Caillot F, et al. Long-term remissions of severe pemphigus after rituximab therapy are associated with prolonged failure of desmoglein B cell response. Sci Transl Med. 2013;5:175ra30.
  13. Heelan K, Al-Mohammedi F, Smith MJ, et al. Durable remission of pemphigus with a fixed-dose rituximab protocol. JAMA Dermatol. 2014;150:703-708.
  14. Joly P, Maho-Vaillant M, Prost-Squarcioni C, et al. First-line rituximab combined with short-term prednisone versus prednisone alone for the treatment of pemphigus (Ritux3): a prospective, multicentre, parallel-group, open-label randomised trial. Lancet. 2017;389:2031-2040
  15. Aryanian Z, Balighi K, Daneshpazhooh M, et al. Rituximab exhibits a better safety profile when used as a first line of treatment for pemphigus vulgaris: a retrospective study. Int Immunopharmacol. 2021;96:107755.
  16. Aghai S, Sodaifi M, Jafari P, et al. DLQI scores in vitiligo: reliability and validity of the Persian version. BMC Dermatol. 2004;4:8.
  17. Schünemann HJ, Akl EA, Guyatt GH. Interpreting the results of patient reported outcome measures in clinical trials: the clinician’s perspective. Health Qual Life Outcomes. 2006;4:62.
  18. Quality of life questionnaires. Cardiff University website. Accessed December 16, 2022. http://sites.cardiff.ac.uk/dermatology/quality-oflife/dermatology-quality-of-life-index-dlqi/dlqi-instructions-foruse-and-scoring/
  19. Kanwar AJ, Tsuruta D, Vinay K, et al. Efficacy and safety of rituximab treatment in Indian pemphigus patients. J Eur Acad Dermatol Venereol. 2013;27:E17-E23.
  20. Ingen-Housz-Oro S, Valeyrie-Allanore L, Cosnes A, et al. First-line treatment of pemphigus vulgaris with a combination of rituximab and high-potency topical corticosteroids. JAMA Dermatol. 2015;151:200-203.
  21. Finlay AY, Khan GK. Dermatology Life Quality Index (DLQI): a simple practical measure for routine clinical use. Clin Exp Dermatol. 1994;19:210-216.
  22. Aghaei S, Moradi A, Ardekani GS. Impact of psoriasis on quality of life in Iran. Indian J Dermatol Venereol Leprol. 2009;75:220.
  23. Ware JE Jr, Sherbourne CD. The MOS 36-item Short-Form Health Survey (SF-36). 1. conceptual framework and item selection. Med Care. 1992;30:473-483.
Article PDF
Author and Disclosure Information

From the Autoimmune Bullous Disease Research Center, Razi Hospital, Tehran University of Medical Sciences, Iran. Dr. Aryanian also is from the Department of Dermatology, Babol University of Medical Sciences, Iran. Drs. Balighi, Nassimi, and Etesami also are from the Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences. Dr. Goodarzi also is from the Department of Dermatology, School of Medicine, Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran.

The authors report no conflict of interest.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Parvaneh Hatami, MD, Autoimmune Bullous Diseases Research Center, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran 1199663911 ([email protected]).

Issue
Cutis - 111(1)
Publications
Topics
Page Number
53-56,E1-E4
Sections
Author and Disclosure Information

From the Autoimmune Bullous Disease Research Center, Razi Hospital, Tehran University of Medical Sciences, Iran. Dr. Aryanian also is from the Department of Dermatology, Babol University of Medical Sciences, Iran. Drs. Balighi, Nassimi, and Etesami also are from the Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences. Dr. Goodarzi also is from the Department of Dermatology, School of Medicine, Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran.

The authors report no conflict of interest.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Parvaneh Hatami, MD, Autoimmune Bullous Diseases Research Center, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran 1199663911 ([email protected]).

Author and Disclosure Information

From the Autoimmune Bullous Disease Research Center, Razi Hospital, Tehran University of Medical Sciences, Iran. Dr. Aryanian also is from the Department of Dermatology, Babol University of Medical Sciences, Iran. Drs. Balighi, Nassimi, and Etesami also are from the Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences. Dr. Goodarzi also is from the Department of Dermatology, School of Medicine, Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran.

The authors report no conflict of interest.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Parvaneh Hatami, MD, Autoimmune Bullous Diseases Research Center, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran 1199663911 ([email protected]).

Article PDF
Article PDF

Pemphigus is a group of autoimmune blistering diseases characterized by the development of painful and flaccid blisters on the skin and/or mucous membranes. Pemphigus vulgaris (PV) and pemphigus foliaceus (PF) are 2 major subtypes and can be distinguished by the location of blister formation or the specificity of autoantibodies directed against different desmogleins.1,2 Although rare, pemphigus is considered a serious and life-threatening condition with a great impact on quality of life (QOL) due to disease symptoms (eg, painful lesions, physical appearance of skin lesions) as well as treatment complications (eg, adverse drug effects, cost of treatment).3-6 Moreover, the physical and psychological effects can lead to marked functional morbidity and work-related disability during patients’ productive years.7 Therefore, affected individuals usually have a remarkably compromised health-related quality of life (HRQOL).8 Effective treatments may considerably improve the QOL of patients with pemphigus.6

Despite the available treatment options, finding the best regimen for pemphigus remains a challenge. Corticosteroids are assumed to be the main treatment, though they have considerable side effects.9,10 Adjuvant therapies are used to suppress or modulate immune responses, leading to remission with the least possible need for corticosteroids. Finding an optimal steroid-sparing agent has been the aim of research, and biologic agents seem to be the best option.8 Rituximab (RTX), an anti-CD20 monoclonal antibody, has shown great promise in several studies of its clinical efficacy and has become a first-line treatment in new guidelines.11-14 Rituximab treatment has been associated with notable improvement in physician-assessed outcome measures with a favorable safety profile in patients with pemphigus.11-15 However, it is important to assess response to treatment from a patient’s perspective through the use of outcome-assessment measures that encompass patient-reported outcomes to reflect the complete patient experience and establish the overall impact of RTX as well as its likelihood of acceptance by patients with pemphigus.

In our study, we compared clinical outcomes and HRQOL through the use of disease-specific measures as well as comprehensive generic health status measures among patients with PV and PF who received RTX treatment 3 months earlier and those who received RTX in the last 2 weeks. The clinical relevance of the patient-reported outcomes is discussed.

MATERIALS AND METHODS

Study Design

We conducted a single-center cross-sectional study of 96 patients with pemphigus aged 18 to 65 years of either sex who were willing to participate in this study. Patients with a confirmed diagnosis of PV or PF who received RTX 3 months earlier or in the last 2 weeks were enrolled in the study. Patients were identified using Dermatry.ir, an archiving software that contains patients’ medical data. Exclusion criteria included lack of sufficient knowledge of the concepts of the questionnaires as well as age younger than 16 years. The study was conducted from October 2019 to April 2020 by the Autoimmune Bullous Disease Research Center at Razi Hospital in Tehran, Iran, which is the main dermatology-specific center and teaching hospital of Iran. The study protocol was approved by the relevant ethics committee.

Patients were categorized into 2 groups: (1) those who received RTX 3 months earlier (3M group); and (2) those who received RTX in the last 2 weeks (R group).

After an explanation of the study to participants, informed written consent was signed by each patient, and their personal data (eg, age, sex, education, marital status, smoking status), as well as clinical data (eg, type of pemphigus, duration of disease, site of onset, prednisolone dosage, presence of Nikolsky sign, anti-DSG1 and anti-DSG3 values, Pemphigus Disease Area Index [PDAI] score, RTX treatment protocol); any known comorbidities such as hypertension, diabetes mellitus, or morbid obesity; and any chronic pulmonary, cardiac, endocrinologic, renal, or hepatic condition, were collected and recorded in a predefined Case Record.

Patient-Reported Outcome Measures

The effect of RTX on QOL in patients with pemphigus was assessed using 2 HRQOL instruments: (1) a general health status indicator, the 36-Item Short Form Survey (SF-36), and (2) a validated, Persian version of a dermatology-specific questionnaire, Dermatology Life Quality Index (DLQI). The questionnaires were completed by each patient or by an assistant if needed.

 

 

The SF-36 is a widely used 36-item questionnaire measuring functional health and well-being across 8 domains—mental health, pain, physical function, role emotional, role physical, social functioning, vitality, and general health perception—with scores for each ranging from 0 to 100. The physical component scores (PCSs) and mental component scores (MCSs) were derived from these 8 subscales, each ranging from 0 to 400, with higher scores indicating better health status.6

The DLQI, one of the most frequently used QOL measures in dermatology, contains 10 questions, each referring to the prior week and classified in the following 6 subscales: symptoms and feelings, daily activities, leisure, personal relationships, work and school, and treatment.16 The total score ranges from 0 (no impact) to 30 (very high impact), with a higher score indicating a lower QOL (eTable 1). The minimal clinically important difference (MCD) for the DLQI was considered to be 2- to 5-point changes in prior studies.17,18 In this study, we used an MCD of a 5-point change or more between study groups.

Classification of Patients Based on DLQI Questionnaire

Moreover, the patient general assessment (PGA) of disease severity was identified using a 3-point scale (1=mild, 2=moderate, 3=severe).

Statistical Analysis

Data were analyzed using SPSS Statistics version 23. P≤.05 was considered significant. Mean and SD were calculated for descriptive data. The t test, Fisher exact test, analysis of variance, multiple regression analysis, and logistic regression analysis were used to identify the relationship between variables.

RESULTS

Patient Characteristics

A total of 96 patients were enrolled in this study. The mean (SD) age of participants was 41.42 (15.1) years (range, 18–58 years). Of 96 patients whose data were included, 55 (57.29%) patients had received RTX 3 months earlier (3M group) and 41 (42.71%) received RTX in the last 2 weeks (R group). A summary of study patient characteristics in each group is provided in eTable 2. There was no significant difference between the 2 groups in terms of age, sex, type of pemphigus, marital status, education, positive Nikolsky sign, smoking status, existence of comorbidities, site of lesions, and RTX treatment protocol. However, a significant difference was found for duration of disease (P=.0124) and mean prednisolone dosage (P=.001) as well as severity of disease measured by PDAI score (P=.003) and anti-DSG1 (P=.003) and anti-DSG3 (P=.021) values.

Summary of Patient Demographic and Clinical Data

Patient-Reported Outcomes

Physical and mental component scores are summarized in eTable 3. Generally, SF-36 scores were improved with RTX treatment in all dimensions except for mental health, though these differences were not statistically significant, with the greatest mean improvement in the role physical index (75.45 in the 3M group vs 53.04 in the R group; P=.009). Mean SF-36 PCS and MCS scores were higher in the 3M group vs the R group, though the difference in MCS score did not reach the level of significance (eTable 3).

Patient-Reported Outcomes

Mean DLQI scores in the R and 3M groups were 12.31 and 6.96, respectively, indicating a considerable burden on HRQOL in both groups. However, a statistically significant difference between these values was seen that also was clinically meaningful, indicating a significant improvement of QOL in patients receiving RTX 3 months earlier (P=.005)(eTable 3).

 

 

The PGA scores indicated that patients in the 3M group were significantly more likely to report less severe disease vs the R group (P=.008)(eTable 3).

Multivariate Analysis—Effect of the patient characteristics and some disease features on indices of QOL was evaluated using the multiple linear regression model. eTable 4 shows the P values of those analyses.

Comparison of QOL Indices Between 2 Study Groups (R and 3M) for Study Variants

COMMENT

Pemphigus is a chronic disabling disease with notable QOL impairment due to disease burden as well as the need for long-term use of immunosuppressive agents during the disease course. To study the effect of RTX on QOL of patients with pemphigus, we compared 2 sets of patients. Prior studies have shown that clinically significant effects of RTX take 4 to 12 weeks to appear.19,20 Therefore, we selected patients who received RTX 3 months earlier to measure their HRQOL indices and compare them with patients who had received RTX in the last 2 weeks as a control group to investigate the effect of RTX intrinsically, as this was the focus of this study.

In our study, one of the research tools was the DLQI. Healthy patients typically have an average score of 0.5.21 The mean DLQI score of the patients in R group was 12.31, which was similar to prior analysis8 and reflects a substantial burden of disease comparable to atopic dermatitis and psoriasis.21,22 In patients in the 3M group, the mean DLQI score was lower than the R group (6.96 vs 12.31), indicating a significant (P=.005) and clinically meaningful improvement in QOL of patients due to the dramatic therapeutic effect of RTX. However, this score indicated a moderate effect on HRQOL, even in the context of clinical improvement due to RTX treatment, which may reflect that the short duration of treatment in the 3M group was a limitation of this study. Although the 12-week treatment duration was comparable with other studies19,20 and major differences in objective measures of treatment efficacy were found in PDAI as well as anti-DSG1 and anti-DSG3 values, longer treatment duration may be needed for a more comprehensive assessment of the benefit of RTX on HRQOL indices in patients with pemphigus.

Based on results of the SF-36 questionnaire, PCS and MCS scores were not substantially impaired in the R group considering the fact that a mean score of 50 has been articulated as a normative value for all scales.23 These data demonstrated the importance of using a dermatologic-specific instrument such as the DLQI instead of a general questionnaire to assess QOL in patients with pemphigus. However, better indices were reported with RTX treatment in the 3 SF-36 domains—role physical (P=.009), role emotional (P=.03), and general health perception (P=.03)—with the role physical showing the greatest magnitude of mean change (75.45 in the 3M group vs 53.04 in the R group). Notably, PCS was impaired to a greater extent than MCS in patients in the R group and showed a greater magnitude of improvement after 3 months of treatment. These results could be explained by the fact that MCS can be largely changed in diseases with a direct effect on the central nervous system.23

Our results also revealed that the dose of corticosteroid correlated to HRQOL of patients with pemphigus who recently received RTX therapy. Indeed, it is more likely that patients on lower-dose prednisolone have a higher QOL, especially on physical function and social function dimensions of SF-36. This finding is highly expectable by less severe disease due to RTX treatment and also lower potential dose-dependent adverse effects of long-term steroid therapy.

One of the most striking findings of this study was the correlation of location of lesions to QOL indices. We found that the mucocutaneous phenotype was significantly correlated to greater improvement in role emotional, role physical, and social functioning scores due to RTX treatment compared with cutaneous or mucosal types (P=.02, P=.025, and P=.017, respectively). Although mucosal involvement of the disease can be the most burdensome feature because of its large impact on essential activities such as eating and speaking, cutaneous lesions with unpleasant appearance and undesirable symptoms may have a similar impact on QOL. Therefore, having both mucosal and cutaneous lesions causes a worsened QOL and decreased treatment efficacy vs having only one area involved. This may explain the greater improvement in some QOL indices with RTX treatment.

Limitations—Given the cross-sectional design of this study in which patients were observed at a single time point during their treatment course, it is not possible to establish a clear cause-effect relationship between variables. Moreover, we did not evaluate the impact of RTX or prednisolone adverse effects on QOL. Therefore, further prospective studies with longer treatment durations may help to validate our findings. In addition, MCDs for DLQI and SF-36 in pemphigus need to be determined and validated in future studies.

CONCLUSION

The results of our study demonstrated that patients with pemphigus may benefit from taking RTX, not only in terms of clinical improvement of their disease measured by objective indices such as PDAI and anti-DSG1 and anti-DSG3 values but also in several domains that are important to patients, including physical and mental health status (SF-36), HRQOL (DLQI), and overall disease severity (PGA). Rituximab administration in patients with pemphigus can lead to rapid and significant improvement in HRQOL as well as patient- and physician-assessed measures. Its favorable safety profile along with its impact on patients’ daily lives and mental health makes RTX a suitable treatment option for patients with pemphigus. Moreover, we recommend taking QOL indices into account while evaluating the efficacy of new medications to improve our insight into the patient experience and provide better patient adherence to treatment, which is an important issue for optimal control of chronic disorders.

Pemphigus is a group of autoimmune blistering diseases characterized by the development of painful and flaccid blisters on the skin and/or mucous membranes. Pemphigus vulgaris (PV) and pemphigus foliaceus (PF) are 2 major subtypes and can be distinguished by the location of blister formation or the specificity of autoantibodies directed against different desmogleins.1,2 Although rare, pemphigus is considered a serious and life-threatening condition with a great impact on quality of life (QOL) due to disease symptoms (eg, painful lesions, physical appearance of skin lesions) as well as treatment complications (eg, adverse drug effects, cost of treatment).3-6 Moreover, the physical and psychological effects can lead to marked functional morbidity and work-related disability during patients’ productive years.7 Therefore, affected individuals usually have a remarkably compromised health-related quality of life (HRQOL).8 Effective treatments may considerably improve the QOL of patients with pemphigus.6

Despite the available treatment options, finding the best regimen for pemphigus remains a challenge. Corticosteroids are assumed to be the main treatment, though they have considerable side effects.9,10 Adjuvant therapies are used to suppress or modulate immune responses, leading to remission with the least possible need for corticosteroids. Finding an optimal steroid-sparing agent has been the aim of research, and biologic agents seem to be the best option.8 Rituximab (RTX), an anti-CD20 monoclonal antibody, has shown great promise in several studies of its clinical efficacy and has become a first-line treatment in new guidelines.11-14 Rituximab treatment has been associated with notable improvement in physician-assessed outcome measures with a favorable safety profile in patients with pemphigus.11-15 However, it is important to assess response to treatment from a patient’s perspective through the use of outcome-assessment measures that encompass patient-reported outcomes to reflect the complete patient experience and establish the overall impact of RTX as well as its likelihood of acceptance by patients with pemphigus.

In our study, we compared clinical outcomes and HRQOL through the use of disease-specific measures as well as comprehensive generic health status measures among patients with PV and PF who received RTX treatment 3 months earlier and those who received RTX in the last 2 weeks. The clinical relevance of the patient-reported outcomes is discussed.

MATERIALS AND METHODS

Study Design

We conducted a single-center cross-sectional study of 96 patients with pemphigus aged 18 to 65 years of either sex who were willing to participate in this study. Patients with a confirmed diagnosis of PV or PF who received RTX 3 months earlier or in the last 2 weeks were enrolled in the study. Patients were identified using Dermatry.ir, an archiving software that contains patients’ medical data. Exclusion criteria included lack of sufficient knowledge of the concepts of the questionnaires as well as age younger than 16 years. The study was conducted from October 2019 to April 2020 by the Autoimmune Bullous Disease Research Center at Razi Hospital in Tehran, Iran, which is the main dermatology-specific center and teaching hospital of Iran. The study protocol was approved by the relevant ethics committee.

Patients were categorized into 2 groups: (1) those who received RTX 3 months earlier (3M group); and (2) those who received RTX in the last 2 weeks (R group).

After an explanation of the study to participants, informed written consent was signed by each patient, and their personal data (eg, age, sex, education, marital status, smoking status), as well as clinical data (eg, type of pemphigus, duration of disease, site of onset, prednisolone dosage, presence of Nikolsky sign, anti-DSG1 and anti-DSG3 values, Pemphigus Disease Area Index [PDAI] score, RTX treatment protocol); any known comorbidities such as hypertension, diabetes mellitus, or morbid obesity; and any chronic pulmonary, cardiac, endocrinologic, renal, or hepatic condition, were collected and recorded in a predefined Case Record.

Patient-Reported Outcome Measures

The effect of RTX on QOL in patients with pemphigus was assessed using 2 HRQOL instruments: (1) a general health status indicator, the 36-Item Short Form Survey (SF-36), and (2) a validated, Persian version of a dermatology-specific questionnaire, Dermatology Life Quality Index (DLQI). The questionnaires were completed by each patient or by an assistant if needed.

 

 

The SF-36 is a widely used 36-item questionnaire measuring functional health and well-being across 8 domains—mental health, pain, physical function, role emotional, role physical, social functioning, vitality, and general health perception—with scores for each ranging from 0 to 100. The physical component scores (PCSs) and mental component scores (MCSs) were derived from these 8 subscales, each ranging from 0 to 400, with higher scores indicating better health status.6

The DLQI, one of the most frequently used QOL measures in dermatology, contains 10 questions, each referring to the prior week and classified in the following 6 subscales: symptoms and feelings, daily activities, leisure, personal relationships, work and school, and treatment.16 The total score ranges from 0 (no impact) to 30 (very high impact), with a higher score indicating a lower QOL (eTable 1). The minimal clinically important difference (MCD) for the DLQI was considered to be 2- to 5-point changes in prior studies.17,18 In this study, we used an MCD of a 5-point change or more between study groups.

Classification of Patients Based on DLQI Questionnaire

Moreover, the patient general assessment (PGA) of disease severity was identified using a 3-point scale (1=mild, 2=moderate, 3=severe).

Statistical Analysis

Data were analyzed using SPSS Statistics version 23. P≤.05 was considered significant. Mean and SD were calculated for descriptive data. The t test, Fisher exact test, analysis of variance, multiple regression analysis, and logistic regression analysis were used to identify the relationship between variables.

RESULTS

Patient Characteristics

A total of 96 patients were enrolled in this study. The mean (SD) age of participants was 41.42 (15.1) years (range, 18–58 years). Of 96 patients whose data were included, 55 (57.29%) patients had received RTX 3 months earlier (3M group) and 41 (42.71%) received RTX in the last 2 weeks (R group). A summary of study patient characteristics in each group is provided in eTable 2. There was no significant difference between the 2 groups in terms of age, sex, type of pemphigus, marital status, education, positive Nikolsky sign, smoking status, existence of comorbidities, site of lesions, and RTX treatment protocol. However, a significant difference was found for duration of disease (P=.0124) and mean prednisolone dosage (P=.001) as well as severity of disease measured by PDAI score (P=.003) and anti-DSG1 (P=.003) and anti-DSG3 (P=.021) values.

Summary of Patient Demographic and Clinical Data

Patient-Reported Outcomes

Physical and mental component scores are summarized in eTable 3. Generally, SF-36 scores were improved with RTX treatment in all dimensions except for mental health, though these differences were not statistically significant, with the greatest mean improvement in the role physical index (75.45 in the 3M group vs 53.04 in the R group; P=.009). Mean SF-36 PCS and MCS scores were higher in the 3M group vs the R group, though the difference in MCS score did not reach the level of significance (eTable 3).

Patient-Reported Outcomes

Mean DLQI scores in the R and 3M groups were 12.31 and 6.96, respectively, indicating a considerable burden on HRQOL in both groups. However, a statistically significant difference between these values was seen that also was clinically meaningful, indicating a significant improvement of QOL in patients receiving RTX 3 months earlier (P=.005)(eTable 3).

 

 

The PGA scores indicated that patients in the 3M group were significantly more likely to report less severe disease vs the R group (P=.008)(eTable 3).

Multivariate Analysis—Effect of the patient characteristics and some disease features on indices of QOL was evaluated using the multiple linear regression model. eTable 4 shows the P values of those analyses.

Comparison of QOL Indices Between 2 Study Groups (R and 3M) for Study Variants

COMMENT

Pemphigus is a chronic disabling disease with notable QOL impairment due to disease burden as well as the need for long-term use of immunosuppressive agents during the disease course. To study the effect of RTX on QOL of patients with pemphigus, we compared 2 sets of patients. Prior studies have shown that clinically significant effects of RTX take 4 to 12 weeks to appear.19,20 Therefore, we selected patients who received RTX 3 months earlier to measure their HRQOL indices and compare them with patients who had received RTX in the last 2 weeks as a control group to investigate the effect of RTX intrinsically, as this was the focus of this study.

In our study, one of the research tools was the DLQI. Healthy patients typically have an average score of 0.5.21 The mean DLQI score of the patients in R group was 12.31, which was similar to prior analysis8 and reflects a substantial burden of disease comparable to atopic dermatitis and psoriasis.21,22 In patients in the 3M group, the mean DLQI score was lower than the R group (6.96 vs 12.31), indicating a significant (P=.005) and clinically meaningful improvement in QOL of patients due to the dramatic therapeutic effect of RTX. However, this score indicated a moderate effect on HRQOL, even in the context of clinical improvement due to RTX treatment, which may reflect that the short duration of treatment in the 3M group was a limitation of this study. Although the 12-week treatment duration was comparable with other studies19,20 and major differences in objective measures of treatment efficacy were found in PDAI as well as anti-DSG1 and anti-DSG3 values, longer treatment duration may be needed for a more comprehensive assessment of the benefit of RTX on HRQOL indices in patients with pemphigus.

Based on results of the SF-36 questionnaire, PCS and MCS scores were not substantially impaired in the R group considering the fact that a mean score of 50 has been articulated as a normative value for all scales.23 These data demonstrated the importance of using a dermatologic-specific instrument such as the DLQI instead of a general questionnaire to assess QOL in patients with pemphigus. However, better indices were reported with RTX treatment in the 3 SF-36 domains—role physical (P=.009), role emotional (P=.03), and general health perception (P=.03)—with the role physical showing the greatest magnitude of mean change (75.45 in the 3M group vs 53.04 in the R group). Notably, PCS was impaired to a greater extent than MCS in patients in the R group and showed a greater magnitude of improvement after 3 months of treatment. These results could be explained by the fact that MCS can be largely changed in diseases with a direct effect on the central nervous system.23

Our results also revealed that the dose of corticosteroid correlated to HRQOL of patients with pemphigus who recently received RTX therapy. Indeed, it is more likely that patients on lower-dose prednisolone have a higher QOL, especially on physical function and social function dimensions of SF-36. This finding is highly expectable by less severe disease due to RTX treatment and also lower potential dose-dependent adverse effects of long-term steroid therapy.

One of the most striking findings of this study was the correlation of location of lesions to QOL indices. We found that the mucocutaneous phenotype was significantly correlated to greater improvement in role emotional, role physical, and social functioning scores due to RTX treatment compared with cutaneous or mucosal types (P=.02, P=.025, and P=.017, respectively). Although mucosal involvement of the disease can be the most burdensome feature because of its large impact on essential activities such as eating and speaking, cutaneous lesions with unpleasant appearance and undesirable symptoms may have a similar impact on QOL. Therefore, having both mucosal and cutaneous lesions causes a worsened QOL and decreased treatment efficacy vs having only one area involved. This may explain the greater improvement in some QOL indices with RTX treatment.

Limitations—Given the cross-sectional design of this study in which patients were observed at a single time point during their treatment course, it is not possible to establish a clear cause-effect relationship between variables. Moreover, we did not evaluate the impact of RTX or prednisolone adverse effects on QOL. Therefore, further prospective studies with longer treatment durations may help to validate our findings. In addition, MCDs for DLQI and SF-36 in pemphigus need to be determined and validated in future studies.

CONCLUSION

The results of our study demonstrated that patients with pemphigus may benefit from taking RTX, not only in terms of clinical improvement of their disease measured by objective indices such as PDAI and anti-DSG1 and anti-DSG3 values but also in several domains that are important to patients, including physical and mental health status (SF-36), HRQOL (DLQI), and overall disease severity (PGA). Rituximab administration in patients with pemphigus can lead to rapid and significant improvement in HRQOL as well as patient- and physician-assessed measures. Its favorable safety profile along with its impact on patients’ daily lives and mental health makes RTX a suitable treatment option for patients with pemphigus. Moreover, we recommend taking QOL indices into account while evaluating the efficacy of new medications to improve our insight into the patient experience and provide better patient adherence to treatment, which is an important issue for optimal control of chronic disorders.

References
  1. Hammers CM, Stanley JR. Mechanisms of disease: pemphigus and bullous pemphigoid. Ann Rev Pathol. 2016;11:175-197.
  2. Kasperkiewicz M, Ellebrecht CT, Takahashi H, et al. Pemphigus. Nat Rev Dis Primers. 2017;3:17026.
  3. Mayrshofer F, Hertl M, Sinkgraven R, et al. Significant decrease in quality of life in patients with pemphigus vulgaris, result from the German Bullous Skin Disease (BSD) Study Group. J Dtsch Dermatol Ges. 2005;3:431-435.
  4. Terrab Z, Benckikhi H, Maaroufi A, et al. Quality of life and pemphigus. Ann Dermatol Venereol. 2005;132:321-328.
  5. Tabolli S, Mozzetta A, Antinone V, et al. The health impact of pemphigus vulgaris and pemphigus foliaceus assessed using the Medical Outcomes Study 36-item short form health survey questionnaire. Br J Dermatol. 2008;158:1029-1034.
  6. Paradisi A, Sampogna F, Di Pietro, C, et al. Quality-of-life assessment in patients with pemphigus using a minimum set of evaluation tools. J Am Acad Dermatol. 2009;60:261-269.
  7. Heelan K, Hitzig SL, Knowles S, et al. Loss of work productivity and quality of life in patients with autoimmune bullous dermatoses. J Cutan Med Surg. 2015;19:546-554.
  8. Ghodsi SZ, Chams-Davatchi C, Daneshpazhooh M, et al. Quality of life and psychological status of patients with pemphigus vulgaris using Dermatology Life Quality Index and General Health Questionnaires. J Dermatol. 2012;39:141-144.
  9. Schäcke H, Döcke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther. 2002;96:2343.
  10. Mohammad-Javad N, Parvaneh H, Maryam G, et al. Randomized trial of tacrolimus 0.1% ointment versus triamcinolone acetonide 0.1% paste in the treatment of oral pemphigus vulgaris. Iranian J Dermatol. 2012;15:42-46.
  11. Lunardon L, Tsai KJ, Propert KJ, et al. Adjuvant rituximab therapy of pemphigus: a single-center experience with 31 patients. Arch Dermatol. 2012;148:1031-1036.
  12. Colliou N, Picard D, Caillot F, et al. Long-term remissions of severe pemphigus after rituximab therapy are associated with prolonged failure of desmoglein B cell response. Sci Transl Med. 2013;5:175ra30.
  13. Heelan K, Al-Mohammedi F, Smith MJ, et al. Durable remission of pemphigus with a fixed-dose rituximab protocol. JAMA Dermatol. 2014;150:703-708.
  14. Joly P, Maho-Vaillant M, Prost-Squarcioni C, et al. First-line rituximab combined with short-term prednisone versus prednisone alone for the treatment of pemphigus (Ritux3): a prospective, multicentre, parallel-group, open-label randomised trial. Lancet. 2017;389:2031-2040
  15. Aryanian Z, Balighi K, Daneshpazhooh M, et al. Rituximab exhibits a better safety profile when used as a first line of treatment for pemphigus vulgaris: a retrospective study. Int Immunopharmacol. 2021;96:107755.
  16. Aghai S, Sodaifi M, Jafari P, et al. DLQI scores in vitiligo: reliability and validity of the Persian version. BMC Dermatol. 2004;4:8.
  17. Schünemann HJ, Akl EA, Guyatt GH. Interpreting the results of patient reported outcome measures in clinical trials: the clinician’s perspective. Health Qual Life Outcomes. 2006;4:62.
  18. Quality of life questionnaires. Cardiff University website. Accessed December 16, 2022. http://sites.cardiff.ac.uk/dermatology/quality-oflife/dermatology-quality-of-life-index-dlqi/dlqi-instructions-foruse-and-scoring/
  19. Kanwar AJ, Tsuruta D, Vinay K, et al. Efficacy and safety of rituximab treatment in Indian pemphigus patients. J Eur Acad Dermatol Venereol. 2013;27:E17-E23.
  20. Ingen-Housz-Oro S, Valeyrie-Allanore L, Cosnes A, et al. First-line treatment of pemphigus vulgaris with a combination of rituximab and high-potency topical corticosteroids. JAMA Dermatol. 2015;151:200-203.
  21. Finlay AY, Khan GK. Dermatology Life Quality Index (DLQI): a simple practical measure for routine clinical use. Clin Exp Dermatol. 1994;19:210-216.
  22. Aghaei S, Moradi A, Ardekani GS. Impact of psoriasis on quality of life in Iran. Indian J Dermatol Venereol Leprol. 2009;75:220.
  23. Ware JE Jr, Sherbourne CD. The MOS 36-item Short-Form Health Survey (SF-36). 1. conceptual framework and item selection. Med Care. 1992;30:473-483.
References
  1. Hammers CM, Stanley JR. Mechanisms of disease: pemphigus and bullous pemphigoid. Ann Rev Pathol. 2016;11:175-197.
  2. Kasperkiewicz M, Ellebrecht CT, Takahashi H, et al. Pemphigus. Nat Rev Dis Primers. 2017;3:17026.
  3. Mayrshofer F, Hertl M, Sinkgraven R, et al. Significant decrease in quality of life in patients with pemphigus vulgaris, result from the German Bullous Skin Disease (BSD) Study Group. J Dtsch Dermatol Ges. 2005;3:431-435.
  4. Terrab Z, Benckikhi H, Maaroufi A, et al. Quality of life and pemphigus. Ann Dermatol Venereol. 2005;132:321-328.
  5. Tabolli S, Mozzetta A, Antinone V, et al. The health impact of pemphigus vulgaris and pemphigus foliaceus assessed using the Medical Outcomes Study 36-item short form health survey questionnaire. Br J Dermatol. 2008;158:1029-1034.
  6. Paradisi A, Sampogna F, Di Pietro, C, et al. Quality-of-life assessment in patients with pemphigus using a minimum set of evaluation tools. J Am Acad Dermatol. 2009;60:261-269.
  7. Heelan K, Hitzig SL, Knowles S, et al. Loss of work productivity and quality of life in patients with autoimmune bullous dermatoses. J Cutan Med Surg. 2015;19:546-554.
  8. Ghodsi SZ, Chams-Davatchi C, Daneshpazhooh M, et al. Quality of life and psychological status of patients with pemphigus vulgaris using Dermatology Life Quality Index and General Health Questionnaires. J Dermatol. 2012;39:141-144.
  9. Schäcke H, Döcke WD, Asadullah K. Mechanisms involved in the side effects of glucocorticoids. Pharmacol Ther. 2002;96:2343.
  10. Mohammad-Javad N, Parvaneh H, Maryam G, et al. Randomized trial of tacrolimus 0.1% ointment versus triamcinolone acetonide 0.1% paste in the treatment of oral pemphigus vulgaris. Iranian J Dermatol. 2012;15:42-46.
  11. Lunardon L, Tsai KJ, Propert KJ, et al. Adjuvant rituximab therapy of pemphigus: a single-center experience with 31 patients. Arch Dermatol. 2012;148:1031-1036.
  12. Colliou N, Picard D, Caillot F, et al. Long-term remissions of severe pemphigus after rituximab therapy are associated with prolonged failure of desmoglein B cell response. Sci Transl Med. 2013;5:175ra30.
  13. Heelan K, Al-Mohammedi F, Smith MJ, et al. Durable remission of pemphigus with a fixed-dose rituximab protocol. JAMA Dermatol. 2014;150:703-708.
  14. Joly P, Maho-Vaillant M, Prost-Squarcioni C, et al. First-line rituximab combined with short-term prednisone versus prednisone alone for the treatment of pemphigus (Ritux3): a prospective, multicentre, parallel-group, open-label randomised trial. Lancet. 2017;389:2031-2040
  15. Aryanian Z, Balighi K, Daneshpazhooh M, et al. Rituximab exhibits a better safety profile when used as a first line of treatment for pemphigus vulgaris: a retrospective study. Int Immunopharmacol. 2021;96:107755.
  16. Aghai S, Sodaifi M, Jafari P, et al. DLQI scores in vitiligo: reliability and validity of the Persian version. BMC Dermatol. 2004;4:8.
  17. Schünemann HJ, Akl EA, Guyatt GH. Interpreting the results of patient reported outcome measures in clinical trials: the clinician’s perspective. Health Qual Life Outcomes. 2006;4:62.
  18. Quality of life questionnaires. Cardiff University website. Accessed December 16, 2022. http://sites.cardiff.ac.uk/dermatology/quality-oflife/dermatology-quality-of-life-index-dlqi/dlqi-instructions-foruse-and-scoring/
  19. Kanwar AJ, Tsuruta D, Vinay K, et al. Efficacy and safety of rituximab treatment in Indian pemphigus patients. J Eur Acad Dermatol Venereol. 2013;27:E17-E23.
  20. Ingen-Housz-Oro S, Valeyrie-Allanore L, Cosnes A, et al. First-line treatment of pemphigus vulgaris with a combination of rituximab and high-potency topical corticosteroids. JAMA Dermatol. 2015;151:200-203.
  21. Finlay AY, Khan GK. Dermatology Life Quality Index (DLQI): a simple practical measure for routine clinical use. Clin Exp Dermatol. 1994;19:210-216.
  22. Aghaei S, Moradi A, Ardekani GS. Impact of psoriasis on quality of life in Iran. Indian J Dermatol Venereol Leprol. 2009;75:220.
  23. Ware JE Jr, Sherbourne CD. The MOS 36-item Short-Form Health Survey (SF-36). 1. conceptual framework and item selection. Med Care. 1992;30:473-483.
Issue
Cutis - 111(1)
Issue
Cutis - 111(1)
Page Number
53-56,E1-E4
Page Number
53-56,E1-E4
Publications
Publications
Topics
Article Type
Display Headline
Rituximab Treatment and Improvement of Health-Related Quality of Life in Patients With Pemphigus
Display Headline
Rituximab Treatment and Improvement of Health-Related Quality of Life in Patients With Pemphigus
Sections
Inside the Article

PRACTICE POINTS

  • Pemphigus is an autoimmune blistering disease that can negatively affect patients’ lives.
  • Assessing the impact of treatment from a patient’s perspective using outcome assessment measures is important and relevant in trials of new pemphigus treatments including rituximab.
  • Rituximab administration in pemphigus patients led to rapid and notable improvement in health-related quality of life and patient-assessed measures.
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

Cutaneous Manifestations in Hereditary Alpha Tryptasemia

Article Type
Changed
Thu, 01/05/2023 - 12:47
Display Headline
Cutaneous Manifestations in Hereditary Alpha Tryptasemia

Hereditary alpha tryptasemia (HaT), an autosomal-dominant disorder of tryptase overproduction, was first described in 2014 by Lyons et al.1 It has been associated with multiple dermatologic, allergic, gastrointestinal (GI) tract, neuropsychiatric, respiratory, autonomic, and connective tissue abnormalities. These multisystem concerns may include cutaneous flushing, chronic pruritus, urticaria, GI tract symptoms, arthralgia, and autonomic dysfunction.2 The diverse symptoms and the recent discovery of HaT make recognition of this disorder challenging. Currently, it also is believed that HaT is associated with an elevated risk for anaphylaxis and is a biomarker for severe symptoms in disorders with increased mast cell burden such as mastocytosis.3-5

Given the potential cutaneous manifestations and the fact that dermatologic symptoms may be the initial presentation of HaT, awareness and recognition of this condition by dermatologists are essential for diagnosis and treatment. This review summarizes the cutaneous presentations consistent with HaT and discusses various conditions that share overlapping dermatologic symptoms with HaT.

Background on HaT

Mast cells are known to secrete several vasoactive mediators including tryptase and histamine when activated by foreign substances, similar to IgE-mediated hypersensitivity reactions. In their baseline state, mast cells continuously secrete immature forms of tryptases called protryptases.6 These protryptases come in 2 forms: α and β. Although mature tryptase is acutely elevatedin anaphylaxis, persistently elevated total serum tryptase levels frequently are regarded as indicative of a systemic mast cell disorder such as systemic mastocytosis (SM).3 Despite the wide-ranging phenotype of HaT, all individuals with the disorder have an elevated basal serum tryptase level (>8 ng/mL). Hereditary alpha tryptasemia has been identified as another possible cause of persistently elevated levels.2,6

Genetics and Epidemiology of HaT—The humantryptase locus at chromosome 16p13.3 is composed of 4 paralog genes: TPSG1, TPSB2, TPSAB1, and TPSD1.4 Only TPSAB1 encodes for α-tryptase, while both TPSB2 and TPSAB1 encode for β-tryptase.4 Hereditary alpha tryptasemia is an autosomal-dominant disorder resulting from a copy number increase in the α-tryptase encoding sequence within the TPSAB1 gene. Despite the wide-ranging phenotype of HaT, all individuals identified with the disorder have a basal serum tryptase level greater than 8 ng/mL, with mean (SD) levels of 15 (5) ng/mL and 24 (6) ng/mL with gene duplication and triplication, respectively (reference range, 0–11.4 ng/mL).2,6 Hereditary alpha tryptasemia likely is common and largely undiagnosed, with a recently estimated prevalence of 5% in the United Kingdom7 and 5.6% in a cohort of 125 individuals from Italy, Slovenia, and the United States.5

Implications of Increased α-tryptase Levels—After an inciting stimulus, the active portions of α-protryptase and β-protryptase are secreted as tetramers by activated mast cells via degranulation. In vitro, β-tryptase homotetramers have been found to play a role in anaphylaxis, while α-homotetramers are nearly inactive.8,9 Recently, however, it has been discovered that α2β2 tetramers also can form and do so in a higher ratio in individuals with increased α-tryptase–encoding gene copies, such as those with HaT.8 These heterotetramers exhibit unique properties compared with the homotetramers and may stimulate epidermal growth factor–like module-containing mucinlike hormone receptor 2 and protease-activated receptor 2 (PAR2). Epidermal growth factor–like module-containing mucinlike hormone receptor 2 activation likely contributes to vibratory urticaria in patients, while activation of PAR2 may have a range of clinical effects, including worsening asthma, inflammatory bowel disease, pruritus, and the exacerbation of dermal inflammation and hyperalgesia.8,10 Thus, α- and β-tryptase tetramers can be considered mediators that may influence the severity of disorders in which mast cells are naturally prevalent and likely contribute to the phenotype of those with HaT.7 Furthermore, these characteristics have been shown to potentially increase in severity with increasing tryptase levels and with increased TPSAB1 duplications.1,2 In contrast, more than 25% of the population is deficient in α-tryptase without known deleterious effects.5

Cutaneous Manifestations of HaT

A case series reported by Lyons et al1 in 2014 detailed persistent elevated basal serum tryptase levels in 9 families with an autosomal-dominant pattern of inheritance. In this cohort, 31 of 33 (94%) affected individuals had a history of atopic dermatitis (AD), and 26 of 33 (79%) affected individuals reported symptoms consistent with mast cell degranulation, including urticaria; flushing; and/or crampy abdominal pain unprovoked or triggered by heat, exercise, vibration, stress, certain foods, or minor physical stimulation.1 A later report by Lyons et al2 in 2016 identified the TPSAB1 α-tryptase–encoding sequence copy number increase as the causative entity for HaT by examining a group of 96 patients from 35 families with frequent recurrent cutaneous flushing and pruritus, sometimes associated with urticaria and sleep disruption. Flushing and pruritus were found in 45% (33/73) of those with a TPSAB1 duplication and 80% (12/15) of those with a triplication (P=.022), suggesting a gene dose effect regarding α-tryptase encoding sequence copy number and these symptoms.2

A 2019 study further explored the clinical finding of urticaria in patients with HaT by specifically examining if vibration-induced urticaria was affected by TPSAB1 gene dosage.8 A cohort of 56 volunteers—35 healthy and 21 with HaT—underwent tryptase genotyping and cutaneous vibratory challenge. The presence of TPSAB1 was significantly correlated with induction of vibration-induced urticaria (P<.01), as the severity and prevalence of the urticarial response increased along with α- and β-tryptase gene ratios.8

 

 

Urticaria and angioedema also were seen in 51% (36/70) of patients in a cohort of HaT patients in the United Kingdom, in which 41% (29/70) also had skin flushing. In contrast to prior studies, these manifestations were not more common in patients with gene triplications or quintuplications than those with duplications.7 In another recent retrospective evaluation conducted at Brigham and Women’s Hospital (Boston, Massachusetts)(N=101), 80% of patients aged 4 to 85 years with confirmed diagnoses of HaT had skin manifestations such as urticaria, flushing, and pruritus.4

HaT and Mast Cell Activation Syndrome—In 2019, a Mast Cell Disorders Committee Work Group Report outlined recommendations for diagnosing and treating primary mast cell activation syndrome (MCAS), a disorder in which mast cells seem to be more easily activated. Mast cell activation syndrome is defined as a primary clinical condition in which there are episodic signs and symptoms of systemic anaphylaxis (Table) concurrently affecting at least 2 organ systems, resulting from secreted mast cell mediators.9,11 The 2019 report also touched on clinical criteria that lack precision for diagnosing MCAS yet are in use, including dermographism and several types of rashes.9 Episode triggers frequent in MCAS include hot water, alcohol, stress, exercise, infection, hormonal changes, and physical stimuli.

Symptoms of MCAS vs HaT

Hereditary alpha tryptasemia has been suggested to be a risk factor for MCAS, which also can be associated with SM and clonal MCAS.9 Patients with MCAS should be tested for increased α-tryptase gene copy number given the overlap in symptoms, the likely predisposition of those with HaT to develop MCAS, and the fact that these patients could be at an increased risk for anaphylaxis.4,7,9,11 However, the clinical phenotype for HaT includes allergic disorders affecting the skin as well as neuropsychiatric and connective tissue abnormalities that are distinctive from MCAS. Although HaT may be considered a heritable risk factor for MCAS, MCAS is only 1 potential phenotype associated with HaT.9

Implications of HaT

Hereditary alpha tryptasemia should be considered in all patients with basal tryptase levels greater than 8 ng/mL. Cutaneous symptoms are among the most common presentations for individuals with HaT and can include AD, chronic or episodic urticaria, pruritus, flushing, and angioedema. However, HaT is unique because of the coupling of these common dermatologic findings with other abnormalities, including abdominal pain and diarrhea, hypermobile joints, and autonomic dysfunction. Patients with HaT also may manifest psychiatric concerns of anxiety, depression, and chronic pain, all of which have been linked to this disorder.

It is unclear in HaT if the presence of extra-allelic copies of tryptase in an individual is directly pathogenic. The effects of increased basal tryptase and α2β2 tetramers have been shown to likely be responsible for some of the clinical features in these individuals but also may magnify other individual underlying disease(s) or diathesis in which mast cells are naturally abundant.8 In the skin, this increased mast cell activation and subsequent histamine release frequently are visible as dermatographia and urticaria. However, mast cell numbers also are known to be increased in both psoriatic and AD skin lesions,12 thus severe presentation of these diseases in conjunction with the other symptoms associated with mast cell activation should prompt suspicion for HaT.

Effects of HaT on Other Cutaneous Disease—Given the increase of mast cells in AD skin lesions and fact that 94% of patients in the 2014 Lyons et al1 study cited a history of AD, HaT may be a risk factor in the development of AD. Interestingly, in addition to the increased mast cells in AD lesions, PAR2+ nerve fibers also are increased in AD lesions and have been implicated in the nonhistaminergic pruritus experienced by patients with AD.12 Thus, given the proposed propensity for α2β2 tetramers to activate PAR2, it is possible this mechanism may contribute to severe pruritus in individuals with AD and concurrent HaT, as those with HaT express increased α2β2 tetramers. However, no study to date has directly compared AD symptoms in patients with concurrent HaT vs patients without it. Further research is needed on how HaT impacts other allergic and inflammatory skin diseases such as AD and psoriasis, but one may reasonably consider HaT when treating chronic inflammatory skin diseases refractory to typical interventions and/or severe presentations. Although HaT is an autosomal-dominant disorder, it is not detected by standard whole exome sequencing or microarrays. A commercial test is available, utilizing a buccal swab to test for TPSAB1 copy number.

HaT and Mast Cell Disorders—When evaluating someone with suspected HaT, it is important to screen for other symptoms of mast cell activation. For instance, in the GI tract increased mast cell activation results in activation of motor neurons and nociceptors and increases secretion and peristalsis with consequent bloating, abdominal pain, and diarrhea.10 Likewise, tryptase also has neuromodulatory effects that amplify the perception of pain and are likely responsible for the feelings of hyperalgesia reported in patients with HaT.13

 

 

There is substantial overlap in the clinical pictures of HaT and MCAS, and HaT is considered a heritable risk factor for MCAS. Consequently, any patient undergoing workup for MCAS also should be tested for HaT. Although HaT is associated with consistently elevated tryptase, MCAS is episodic in nature, and an increase in tryptase levels of at least 20% plus 2 ng/mL from baseline only in the presence of other symptoms reflective of mast cell activation (Table) is a prerequisite for diagnosis.9 Chronic signs and symptoms of atopy, chronic urticaria, and severe asthma are not indicative of MCAS but are frequently seen in HaT.

Another cause of persistently elevated tryptase levels is SM. Systemic mastocytosis is defined by aberrant clonal mast cell expansion and systemic involvement11 and can cause persistent symptoms, unlike MCAS alone. However, SM also can be associated with MCAS.9 Notably, a baseline serum tryptase level greater than 20 ng/mL—much higher than the threshold of greater than 8 ng/mL for suspicion of HaT—is seen in 75% of SM cases and is part of the minor diagnostic criteria for the disease.9,11 However, the 2016 study identifying increased TPSAB1 α-tryptase–encoding sequences as the causative entity for HaT by Lyons et al2 found the average (SD) basal serum tryptase level in individuals with α-tryptase–encoding sequence duplications to be 15 (5) ng/mL and 24 (6) ng/mL in those with triplications. Thus, there likely is no threshold for elevated baseline tryptase levels that would indicate SM over HaT as a more likely diagnosis. However, SM will present with new persistently elevated tryptase levels, whereas the elevation in HaT is believed to be lifelong.5 Also in contrast to HaT, SM can present with liver, spleen, and lymph node involvement; bone sclerosis; and cytopenia.11,14

Mastocytosis is much rarer than HaT, with an estimated prevalence of 9 cases per 100,000 individuals in the United States.11 Although HaT diagnostic testing is noninvasive, SM requires a bone marrow biopsy for definitive diagnosis. Given the likely much higher prevalence of HaT than SM and the patient burden of a bone marrow biopsy, HaT should be considered before proceeding with a bone marrow biopsy to evaluate for SM when a patient presents with persistent systemic symptoms of mast cell activation and elevated baseline tryptase levels. Furthermore, it also would be prudent to test for HaT in patients with known SM, as a cohort study by Lyons et al5 indicated that HaT is likely more common in those with SM (12.2% [10/82] of cohort with known SM vs 5.3% of 398 controls), and patients with concurrent SM and HaT were at a higher risk for severe anaphylaxis (RR=9.5; P=.007).

Studies thus far surrounding HaT have not evaluated timing of initial symptom onset or age of initial presentation for HaT. Furthermore, there is no guarantee that those with increased TPSAB1 copy number will be symptomatic, as there have been reports of asymptomatic individuals with HaT who had basal serum levels greater than 8 ng/mL.7 As research into HaT continues and larger cohorts are evaluated, questions surrounding timing of symptom onset and various factors that may make someone more likely to display a particular phenotype will be answered.

Treatment—Long-term prognosis for individuals with HaT is largely unknown. Unfortunately, there are limited data to support a single effective treatment strategy for managing HaT, and treatment has varied based on predominant symptoms. For cutaneous and GI tract symptoms, trials of maximal H1 and H2 antihistamines twice daily have been recommended.4 Omalizumab was reported to improve chronic urticaria in 3 of 3 patients, showing potential promise as a treatment.4 Mast cell stabilizers, such as oral cromolyn, have been used for severe GI symptoms, while some patients also have reported improvement with oral ketotifen.6 Other medications, such as tricyclic antidepressants, clemastine fumarate, and gabapentin, have been beneficial anecdotally.6 Given the lack of harmful effects seen in individuals who are α-tryptase deficient, α-tryptase inhibition is an intriguing target for future therapies.

Conclusion

Patients who present with a constellation of dermatologic, allergic, GI tract, neuropsychiatric, respiratory, autonomic, and connective tissue abnormalities consistent with HaT may receive a prompt diagnosis if the association is recognized. The full relationship between HaT and other chronic dermatologic disorders is still unknown. Ultimately, heightened interest and research into HaT will lead to more treatment options available for affected patients.

References

1. Lyons JJ, Sun G, Stone KD, et al. Mendelian inheritance of elevated serum tryptase associated with atopy and connective tissue abnormalities. J Allergy Clin Immunol. 2014;133:1471-1474.

2. Lyons JJ, Yu X, Hughes JD, et al. Elevated basal serum tryptase identifies a multisystem disorder associated with increased TPSAB1 copy number. Nat Genet. 2016;48:1564-1569.

3. Schwartz L. Diagnostic value of tryptase in anaphylaxis and mastocytosis. Immunol Allergy Clin North Am. 2006;6:451-463.

4. Giannetti MP, Weller E, Bormans C, et al. Hereditary alpha-tryptasemia in 101 patients with mast cell activation–related symptomatology including anaphylaxis. Ann Allergy Asthma Immunol. 2021;126:655-660.

5. Lyons JJ, Chovanec J, O’Connell MP, et al. Heritable risk for severe anaphylaxis associated with increased α-tryptase–encoding germline copy number at TPSAB1. J Allergy Clin Immunol. 2020;147:622-632.

6. Lyons JJ. Hereditary alpha tryptasemia: genotyping and associated clinical features. Immunol Allergy Clin North Am. 2018;38:483-495.

7. Robey RC, Wilcock A, Bonin H, et al. Hereditary alpha-tryptasemia: UK prevalence and variability in disease expression. J Allergy Clin Immunol Pract. 2020;8:3549-3556.

8. Le QT, Lyons JJ, Naranjo AN, et al. Impact of naturally forming human α/β-tryptase heterotetramers in the pathogenesis of hereditary α-tryptasemia. J Exp Med. 2019;216:2348-2361.

9. Weiler CR, Austen KF, Akin C, et al. AAAAI Mast Cell Disorders Committee Work Group Report: mast cell activation syndrome (MCAS) diagnosis and management. J Allergy Clin Immunol. 2019;144:883-896.

10. Ramsay DB, Stephen S, Borum M, et al. Mast cells in gastrointestinal disease. Gastroenterol Hepatol (N Y). 2010;6:772-777.

11. Giannetti A, Filice E, Caffarelli C, et al. Mast cell activation disorders. Medicina (Kaunas). 2021;57:124.

12. Siiskonen H, Harvima I. Mast cells and sensory nerves contribute to neurogenic inflammation and pruritus in chronic skin inflammation. Front Cell Neurosci. 2019;13:422.

13. Varrassi G, Fusco M, Skaper SD, et al. A pharmacological rationale to reduce the incidence of opioid induced tolerance and hyperalgesia: a review. Pain Ther. 2018;7:59-75.

14. Núñez E, Moreno-Borque R, García-Montero A, et al. Serum tryptase monitoring in indolent systemic mastocytosis: association with disease features and patient outcome. PLoS One. 2013;8:E76116.

Article PDF
Author and Disclosure Information

Drs. Kranyak and Shuler are from the University of South Carolina School of Medicine, Greenville. Dr. Wine Lee is from the Departments of Dermatology and Pediatrics, Medical University of South Carolina, Charleston.

Dr. Kranyak reports no conflict of interest. Dr. Shuler is a speaker for AbbVie, Eli Lilly and Company, Janssen, Pfizer, and Sanofi-Regeneron. Dr. Wine Lee has received research support from AbbVie, Amgen, Amyrt, Arcutis, Avita, Castle Creek, Celgene, Eli Lilly and Company, Galderma, Incyte Corporation, Janssen, Kiniksa, Mayne Pharmaceuticals, Moonlake Pharmaceuticals, Novartis, Pfizer, Sanofi-Regeneron, Target Pharma, Timber Pharmaceuticals, Trevi Therapeutics, and UCB. She has received research fees from Amyrt, Castle Creek, Eli Lilly and Company, Novartis, Pfizer, and Regeneron, as well as consulting fees from AbbVie, Krystal Biotech, and Pyramid Bioscience.

Correspondence: Allison Kranyak, MD, 607 Grove Rd, Greenville, SC 29605 ([email protected]).

Issue
Cutis - 111(1)
Publications
Topics
Page Number
49-52
Sections
Author and Disclosure Information

Drs. Kranyak and Shuler are from the University of South Carolina School of Medicine, Greenville. Dr. Wine Lee is from the Departments of Dermatology and Pediatrics, Medical University of South Carolina, Charleston.

Dr. Kranyak reports no conflict of interest. Dr. Shuler is a speaker for AbbVie, Eli Lilly and Company, Janssen, Pfizer, and Sanofi-Regeneron. Dr. Wine Lee has received research support from AbbVie, Amgen, Amyrt, Arcutis, Avita, Castle Creek, Celgene, Eli Lilly and Company, Galderma, Incyte Corporation, Janssen, Kiniksa, Mayne Pharmaceuticals, Moonlake Pharmaceuticals, Novartis, Pfizer, Sanofi-Regeneron, Target Pharma, Timber Pharmaceuticals, Trevi Therapeutics, and UCB. She has received research fees from Amyrt, Castle Creek, Eli Lilly and Company, Novartis, Pfizer, and Regeneron, as well as consulting fees from AbbVie, Krystal Biotech, and Pyramid Bioscience.

Correspondence: Allison Kranyak, MD, 607 Grove Rd, Greenville, SC 29605 ([email protected]).

Author and Disclosure Information

Drs. Kranyak and Shuler are from the University of South Carolina School of Medicine, Greenville. Dr. Wine Lee is from the Departments of Dermatology and Pediatrics, Medical University of South Carolina, Charleston.

Dr. Kranyak reports no conflict of interest. Dr. Shuler is a speaker for AbbVie, Eli Lilly and Company, Janssen, Pfizer, and Sanofi-Regeneron. Dr. Wine Lee has received research support from AbbVie, Amgen, Amyrt, Arcutis, Avita, Castle Creek, Celgene, Eli Lilly and Company, Galderma, Incyte Corporation, Janssen, Kiniksa, Mayne Pharmaceuticals, Moonlake Pharmaceuticals, Novartis, Pfizer, Sanofi-Regeneron, Target Pharma, Timber Pharmaceuticals, Trevi Therapeutics, and UCB. She has received research fees from Amyrt, Castle Creek, Eli Lilly and Company, Novartis, Pfizer, and Regeneron, as well as consulting fees from AbbVie, Krystal Biotech, and Pyramid Bioscience.

Correspondence: Allison Kranyak, MD, 607 Grove Rd, Greenville, SC 29605 ([email protected]).

Article PDF
Article PDF

Hereditary alpha tryptasemia (HaT), an autosomal-dominant disorder of tryptase overproduction, was first described in 2014 by Lyons et al.1 It has been associated with multiple dermatologic, allergic, gastrointestinal (GI) tract, neuropsychiatric, respiratory, autonomic, and connective tissue abnormalities. These multisystem concerns may include cutaneous flushing, chronic pruritus, urticaria, GI tract symptoms, arthralgia, and autonomic dysfunction.2 The diverse symptoms and the recent discovery of HaT make recognition of this disorder challenging. Currently, it also is believed that HaT is associated with an elevated risk for anaphylaxis and is a biomarker for severe symptoms in disorders with increased mast cell burden such as mastocytosis.3-5

Given the potential cutaneous manifestations and the fact that dermatologic symptoms may be the initial presentation of HaT, awareness and recognition of this condition by dermatologists are essential for diagnosis and treatment. This review summarizes the cutaneous presentations consistent with HaT and discusses various conditions that share overlapping dermatologic symptoms with HaT.

Background on HaT

Mast cells are known to secrete several vasoactive mediators including tryptase and histamine when activated by foreign substances, similar to IgE-mediated hypersensitivity reactions. In their baseline state, mast cells continuously secrete immature forms of tryptases called protryptases.6 These protryptases come in 2 forms: α and β. Although mature tryptase is acutely elevatedin anaphylaxis, persistently elevated total serum tryptase levels frequently are regarded as indicative of a systemic mast cell disorder such as systemic mastocytosis (SM).3 Despite the wide-ranging phenotype of HaT, all individuals with the disorder have an elevated basal serum tryptase level (>8 ng/mL). Hereditary alpha tryptasemia has been identified as another possible cause of persistently elevated levels.2,6

Genetics and Epidemiology of HaT—The humantryptase locus at chromosome 16p13.3 is composed of 4 paralog genes: TPSG1, TPSB2, TPSAB1, and TPSD1.4 Only TPSAB1 encodes for α-tryptase, while both TPSB2 and TPSAB1 encode for β-tryptase.4 Hereditary alpha tryptasemia is an autosomal-dominant disorder resulting from a copy number increase in the α-tryptase encoding sequence within the TPSAB1 gene. Despite the wide-ranging phenotype of HaT, all individuals identified with the disorder have a basal serum tryptase level greater than 8 ng/mL, with mean (SD) levels of 15 (5) ng/mL and 24 (6) ng/mL with gene duplication and triplication, respectively (reference range, 0–11.4 ng/mL).2,6 Hereditary alpha tryptasemia likely is common and largely undiagnosed, with a recently estimated prevalence of 5% in the United Kingdom7 and 5.6% in a cohort of 125 individuals from Italy, Slovenia, and the United States.5

Implications of Increased α-tryptase Levels—After an inciting stimulus, the active portions of α-protryptase and β-protryptase are secreted as tetramers by activated mast cells via degranulation. In vitro, β-tryptase homotetramers have been found to play a role in anaphylaxis, while α-homotetramers are nearly inactive.8,9 Recently, however, it has been discovered that α2β2 tetramers also can form and do so in a higher ratio in individuals with increased α-tryptase–encoding gene copies, such as those with HaT.8 These heterotetramers exhibit unique properties compared with the homotetramers and may stimulate epidermal growth factor–like module-containing mucinlike hormone receptor 2 and protease-activated receptor 2 (PAR2). Epidermal growth factor–like module-containing mucinlike hormone receptor 2 activation likely contributes to vibratory urticaria in patients, while activation of PAR2 may have a range of clinical effects, including worsening asthma, inflammatory bowel disease, pruritus, and the exacerbation of dermal inflammation and hyperalgesia.8,10 Thus, α- and β-tryptase tetramers can be considered mediators that may influence the severity of disorders in which mast cells are naturally prevalent and likely contribute to the phenotype of those with HaT.7 Furthermore, these characteristics have been shown to potentially increase in severity with increasing tryptase levels and with increased TPSAB1 duplications.1,2 In contrast, more than 25% of the population is deficient in α-tryptase without known deleterious effects.5

Cutaneous Manifestations of HaT

A case series reported by Lyons et al1 in 2014 detailed persistent elevated basal serum tryptase levels in 9 families with an autosomal-dominant pattern of inheritance. In this cohort, 31 of 33 (94%) affected individuals had a history of atopic dermatitis (AD), and 26 of 33 (79%) affected individuals reported symptoms consistent with mast cell degranulation, including urticaria; flushing; and/or crampy abdominal pain unprovoked or triggered by heat, exercise, vibration, stress, certain foods, or minor physical stimulation.1 A later report by Lyons et al2 in 2016 identified the TPSAB1 α-tryptase–encoding sequence copy number increase as the causative entity for HaT by examining a group of 96 patients from 35 families with frequent recurrent cutaneous flushing and pruritus, sometimes associated with urticaria and sleep disruption. Flushing and pruritus were found in 45% (33/73) of those with a TPSAB1 duplication and 80% (12/15) of those with a triplication (P=.022), suggesting a gene dose effect regarding α-tryptase encoding sequence copy number and these symptoms.2

A 2019 study further explored the clinical finding of urticaria in patients with HaT by specifically examining if vibration-induced urticaria was affected by TPSAB1 gene dosage.8 A cohort of 56 volunteers—35 healthy and 21 with HaT—underwent tryptase genotyping and cutaneous vibratory challenge. The presence of TPSAB1 was significantly correlated with induction of vibration-induced urticaria (P<.01), as the severity and prevalence of the urticarial response increased along with α- and β-tryptase gene ratios.8

 

 

Urticaria and angioedema also were seen in 51% (36/70) of patients in a cohort of HaT patients in the United Kingdom, in which 41% (29/70) also had skin flushing. In contrast to prior studies, these manifestations were not more common in patients with gene triplications or quintuplications than those with duplications.7 In another recent retrospective evaluation conducted at Brigham and Women’s Hospital (Boston, Massachusetts)(N=101), 80% of patients aged 4 to 85 years with confirmed diagnoses of HaT had skin manifestations such as urticaria, flushing, and pruritus.4

HaT and Mast Cell Activation Syndrome—In 2019, a Mast Cell Disorders Committee Work Group Report outlined recommendations for diagnosing and treating primary mast cell activation syndrome (MCAS), a disorder in which mast cells seem to be more easily activated. Mast cell activation syndrome is defined as a primary clinical condition in which there are episodic signs and symptoms of systemic anaphylaxis (Table) concurrently affecting at least 2 organ systems, resulting from secreted mast cell mediators.9,11 The 2019 report also touched on clinical criteria that lack precision for diagnosing MCAS yet are in use, including dermographism and several types of rashes.9 Episode triggers frequent in MCAS include hot water, alcohol, stress, exercise, infection, hormonal changes, and physical stimuli.

Symptoms of MCAS vs HaT

Hereditary alpha tryptasemia has been suggested to be a risk factor for MCAS, which also can be associated with SM and clonal MCAS.9 Patients with MCAS should be tested for increased α-tryptase gene copy number given the overlap in symptoms, the likely predisposition of those with HaT to develop MCAS, and the fact that these patients could be at an increased risk for anaphylaxis.4,7,9,11 However, the clinical phenotype for HaT includes allergic disorders affecting the skin as well as neuropsychiatric and connective tissue abnormalities that are distinctive from MCAS. Although HaT may be considered a heritable risk factor for MCAS, MCAS is only 1 potential phenotype associated with HaT.9

Implications of HaT

Hereditary alpha tryptasemia should be considered in all patients with basal tryptase levels greater than 8 ng/mL. Cutaneous symptoms are among the most common presentations for individuals with HaT and can include AD, chronic or episodic urticaria, pruritus, flushing, and angioedema. However, HaT is unique because of the coupling of these common dermatologic findings with other abnormalities, including abdominal pain and diarrhea, hypermobile joints, and autonomic dysfunction. Patients with HaT also may manifest psychiatric concerns of anxiety, depression, and chronic pain, all of which have been linked to this disorder.

It is unclear in HaT if the presence of extra-allelic copies of tryptase in an individual is directly pathogenic. The effects of increased basal tryptase and α2β2 tetramers have been shown to likely be responsible for some of the clinical features in these individuals but also may magnify other individual underlying disease(s) or diathesis in which mast cells are naturally abundant.8 In the skin, this increased mast cell activation and subsequent histamine release frequently are visible as dermatographia and urticaria. However, mast cell numbers also are known to be increased in both psoriatic and AD skin lesions,12 thus severe presentation of these diseases in conjunction with the other symptoms associated with mast cell activation should prompt suspicion for HaT.

Effects of HaT on Other Cutaneous Disease—Given the increase of mast cells in AD skin lesions and fact that 94% of patients in the 2014 Lyons et al1 study cited a history of AD, HaT may be a risk factor in the development of AD. Interestingly, in addition to the increased mast cells in AD lesions, PAR2+ nerve fibers also are increased in AD lesions and have been implicated in the nonhistaminergic pruritus experienced by patients with AD.12 Thus, given the proposed propensity for α2β2 tetramers to activate PAR2, it is possible this mechanism may contribute to severe pruritus in individuals with AD and concurrent HaT, as those with HaT express increased α2β2 tetramers. However, no study to date has directly compared AD symptoms in patients with concurrent HaT vs patients without it. Further research is needed on how HaT impacts other allergic and inflammatory skin diseases such as AD and psoriasis, but one may reasonably consider HaT when treating chronic inflammatory skin diseases refractory to typical interventions and/or severe presentations. Although HaT is an autosomal-dominant disorder, it is not detected by standard whole exome sequencing or microarrays. A commercial test is available, utilizing a buccal swab to test for TPSAB1 copy number.

HaT and Mast Cell Disorders—When evaluating someone with suspected HaT, it is important to screen for other symptoms of mast cell activation. For instance, in the GI tract increased mast cell activation results in activation of motor neurons and nociceptors and increases secretion and peristalsis with consequent bloating, abdominal pain, and diarrhea.10 Likewise, tryptase also has neuromodulatory effects that amplify the perception of pain and are likely responsible for the feelings of hyperalgesia reported in patients with HaT.13

 

 

There is substantial overlap in the clinical pictures of HaT and MCAS, and HaT is considered a heritable risk factor for MCAS. Consequently, any patient undergoing workup for MCAS also should be tested for HaT. Although HaT is associated with consistently elevated tryptase, MCAS is episodic in nature, and an increase in tryptase levels of at least 20% plus 2 ng/mL from baseline only in the presence of other symptoms reflective of mast cell activation (Table) is a prerequisite for diagnosis.9 Chronic signs and symptoms of atopy, chronic urticaria, and severe asthma are not indicative of MCAS but are frequently seen in HaT.

Another cause of persistently elevated tryptase levels is SM. Systemic mastocytosis is defined by aberrant clonal mast cell expansion and systemic involvement11 and can cause persistent symptoms, unlike MCAS alone. However, SM also can be associated with MCAS.9 Notably, a baseline serum tryptase level greater than 20 ng/mL—much higher than the threshold of greater than 8 ng/mL for suspicion of HaT—is seen in 75% of SM cases and is part of the minor diagnostic criteria for the disease.9,11 However, the 2016 study identifying increased TPSAB1 α-tryptase–encoding sequences as the causative entity for HaT by Lyons et al2 found the average (SD) basal serum tryptase level in individuals with α-tryptase–encoding sequence duplications to be 15 (5) ng/mL and 24 (6) ng/mL in those with triplications. Thus, there likely is no threshold for elevated baseline tryptase levels that would indicate SM over HaT as a more likely diagnosis. However, SM will present with new persistently elevated tryptase levels, whereas the elevation in HaT is believed to be lifelong.5 Also in contrast to HaT, SM can present with liver, spleen, and lymph node involvement; bone sclerosis; and cytopenia.11,14

Mastocytosis is much rarer than HaT, with an estimated prevalence of 9 cases per 100,000 individuals in the United States.11 Although HaT diagnostic testing is noninvasive, SM requires a bone marrow biopsy for definitive diagnosis. Given the likely much higher prevalence of HaT than SM and the patient burden of a bone marrow biopsy, HaT should be considered before proceeding with a bone marrow biopsy to evaluate for SM when a patient presents with persistent systemic symptoms of mast cell activation and elevated baseline tryptase levels. Furthermore, it also would be prudent to test for HaT in patients with known SM, as a cohort study by Lyons et al5 indicated that HaT is likely more common in those with SM (12.2% [10/82] of cohort with known SM vs 5.3% of 398 controls), and patients with concurrent SM and HaT were at a higher risk for severe anaphylaxis (RR=9.5; P=.007).

Studies thus far surrounding HaT have not evaluated timing of initial symptom onset or age of initial presentation for HaT. Furthermore, there is no guarantee that those with increased TPSAB1 copy number will be symptomatic, as there have been reports of asymptomatic individuals with HaT who had basal serum levels greater than 8 ng/mL.7 As research into HaT continues and larger cohorts are evaluated, questions surrounding timing of symptom onset and various factors that may make someone more likely to display a particular phenotype will be answered.

Treatment—Long-term prognosis for individuals with HaT is largely unknown. Unfortunately, there are limited data to support a single effective treatment strategy for managing HaT, and treatment has varied based on predominant symptoms. For cutaneous and GI tract symptoms, trials of maximal H1 and H2 antihistamines twice daily have been recommended.4 Omalizumab was reported to improve chronic urticaria in 3 of 3 patients, showing potential promise as a treatment.4 Mast cell stabilizers, such as oral cromolyn, have been used for severe GI symptoms, while some patients also have reported improvement with oral ketotifen.6 Other medications, such as tricyclic antidepressants, clemastine fumarate, and gabapentin, have been beneficial anecdotally.6 Given the lack of harmful effects seen in individuals who are α-tryptase deficient, α-tryptase inhibition is an intriguing target for future therapies.

Conclusion

Patients who present with a constellation of dermatologic, allergic, GI tract, neuropsychiatric, respiratory, autonomic, and connective tissue abnormalities consistent with HaT may receive a prompt diagnosis if the association is recognized. The full relationship between HaT and other chronic dermatologic disorders is still unknown. Ultimately, heightened interest and research into HaT will lead to more treatment options available for affected patients.

Hereditary alpha tryptasemia (HaT), an autosomal-dominant disorder of tryptase overproduction, was first described in 2014 by Lyons et al.1 It has been associated with multiple dermatologic, allergic, gastrointestinal (GI) tract, neuropsychiatric, respiratory, autonomic, and connective tissue abnormalities. These multisystem concerns may include cutaneous flushing, chronic pruritus, urticaria, GI tract symptoms, arthralgia, and autonomic dysfunction.2 The diverse symptoms and the recent discovery of HaT make recognition of this disorder challenging. Currently, it also is believed that HaT is associated with an elevated risk for anaphylaxis and is a biomarker for severe symptoms in disorders with increased mast cell burden such as mastocytosis.3-5

Given the potential cutaneous manifestations and the fact that dermatologic symptoms may be the initial presentation of HaT, awareness and recognition of this condition by dermatologists are essential for diagnosis and treatment. This review summarizes the cutaneous presentations consistent with HaT and discusses various conditions that share overlapping dermatologic symptoms with HaT.

Background on HaT

Mast cells are known to secrete several vasoactive mediators including tryptase and histamine when activated by foreign substances, similar to IgE-mediated hypersensitivity reactions. In their baseline state, mast cells continuously secrete immature forms of tryptases called protryptases.6 These protryptases come in 2 forms: α and β. Although mature tryptase is acutely elevatedin anaphylaxis, persistently elevated total serum tryptase levels frequently are regarded as indicative of a systemic mast cell disorder such as systemic mastocytosis (SM).3 Despite the wide-ranging phenotype of HaT, all individuals with the disorder have an elevated basal serum tryptase level (>8 ng/mL). Hereditary alpha tryptasemia has been identified as another possible cause of persistently elevated levels.2,6

Genetics and Epidemiology of HaT—The humantryptase locus at chromosome 16p13.3 is composed of 4 paralog genes: TPSG1, TPSB2, TPSAB1, and TPSD1.4 Only TPSAB1 encodes for α-tryptase, while both TPSB2 and TPSAB1 encode for β-tryptase.4 Hereditary alpha tryptasemia is an autosomal-dominant disorder resulting from a copy number increase in the α-tryptase encoding sequence within the TPSAB1 gene. Despite the wide-ranging phenotype of HaT, all individuals identified with the disorder have a basal serum tryptase level greater than 8 ng/mL, with mean (SD) levels of 15 (5) ng/mL and 24 (6) ng/mL with gene duplication and triplication, respectively (reference range, 0–11.4 ng/mL).2,6 Hereditary alpha tryptasemia likely is common and largely undiagnosed, with a recently estimated prevalence of 5% in the United Kingdom7 and 5.6% in a cohort of 125 individuals from Italy, Slovenia, and the United States.5

Implications of Increased α-tryptase Levels—After an inciting stimulus, the active portions of α-protryptase and β-protryptase are secreted as tetramers by activated mast cells via degranulation. In vitro, β-tryptase homotetramers have been found to play a role in anaphylaxis, while α-homotetramers are nearly inactive.8,9 Recently, however, it has been discovered that α2β2 tetramers also can form and do so in a higher ratio in individuals with increased α-tryptase–encoding gene copies, such as those with HaT.8 These heterotetramers exhibit unique properties compared with the homotetramers and may stimulate epidermal growth factor–like module-containing mucinlike hormone receptor 2 and protease-activated receptor 2 (PAR2). Epidermal growth factor–like module-containing mucinlike hormone receptor 2 activation likely contributes to vibratory urticaria in patients, while activation of PAR2 may have a range of clinical effects, including worsening asthma, inflammatory bowel disease, pruritus, and the exacerbation of dermal inflammation and hyperalgesia.8,10 Thus, α- and β-tryptase tetramers can be considered mediators that may influence the severity of disorders in which mast cells are naturally prevalent and likely contribute to the phenotype of those with HaT.7 Furthermore, these characteristics have been shown to potentially increase in severity with increasing tryptase levels and with increased TPSAB1 duplications.1,2 In contrast, more than 25% of the population is deficient in α-tryptase without known deleterious effects.5

Cutaneous Manifestations of HaT

A case series reported by Lyons et al1 in 2014 detailed persistent elevated basal serum tryptase levels in 9 families with an autosomal-dominant pattern of inheritance. In this cohort, 31 of 33 (94%) affected individuals had a history of atopic dermatitis (AD), and 26 of 33 (79%) affected individuals reported symptoms consistent with mast cell degranulation, including urticaria; flushing; and/or crampy abdominal pain unprovoked or triggered by heat, exercise, vibration, stress, certain foods, or minor physical stimulation.1 A later report by Lyons et al2 in 2016 identified the TPSAB1 α-tryptase–encoding sequence copy number increase as the causative entity for HaT by examining a group of 96 patients from 35 families with frequent recurrent cutaneous flushing and pruritus, sometimes associated with urticaria and sleep disruption. Flushing and pruritus were found in 45% (33/73) of those with a TPSAB1 duplication and 80% (12/15) of those with a triplication (P=.022), suggesting a gene dose effect regarding α-tryptase encoding sequence copy number and these symptoms.2

A 2019 study further explored the clinical finding of urticaria in patients with HaT by specifically examining if vibration-induced urticaria was affected by TPSAB1 gene dosage.8 A cohort of 56 volunteers—35 healthy and 21 with HaT—underwent tryptase genotyping and cutaneous vibratory challenge. The presence of TPSAB1 was significantly correlated with induction of vibration-induced urticaria (P<.01), as the severity and prevalence of the urticarial response increased along with α- and β-tryptase gene ratios.8

 

 

Urticaria and angioedema also were seen in 51% (36/70) of patients in a cohort of HaT patients in the United Kingdom, in which 41% (29/70) also had skin flushing. In contrast to prior studies, these manifestations were not more common in patients with gene triplications or quintuplications than those with duplications.7 In another recent retrospective evaluation conducted at Brigham and Women’s Hospital (Boston, Massachusetts)(N=101), 80% of patients aged 4 to 85 years with confirmed diagnoses of HaT had skin manifestations such as urticaria, flushing, and pruritus.4

HaT and Mast Cell Activation Syndrome—In 2019, a Mast Cell Disorders Committee Work Group Report outlined recommendations for diagnosing and treating primary mast cell activation syndrome (MCAS), a disorder in which mast cells seem to be more easily activated. Mast cell activation syndrome is defined as a primary clinical condition in which there are episodic signs and symptoms of systemic anaphylaxis (Table) concurrently affecting at least 2 organ systems, resulting from secreted mast cell mediators.9,11 The 2019 report also touched on clinical criteria that lack precision for diagnosing MCAS yet are in use, including dermographism and several types of rashes.9 Episode triggers frequent in MCAS include hot water, alcohol, stress, exercise, infection, hormonal changes, and physical stimuli.

Symptoms of MCAS vs HaT

Hereditary alpha tryptasemia has been suggested to be a risk factor for MCAS, which also can be associated with SM and clonal MCAS.9 Patients with MCAS should be tested for increased α-tryptase gene copy number given the overlap in symptoms, the likely predisposition of those with HaT to develop MCAS, and the fact that these patients could be at an increased risk for anaphylaxis.4,7,9,11 However, the clinical phenotype for HaT includes allergic disorders affecting the skin as well as neuropsychiatric and connective tissue abnormalities that are distinctive from MCAS. Although HaT may be considered a heritable risk factor for MCAS, MCAS is only 1 potential phenotype associated with HaT.9

Implications of HaT

Hereditary alpha tryptasemia should be considered in all patients with basal tryptase levels greater than 8 ng/mL. Cutaneous symptoms are among the most common presentations for individuals with HaT and can include AD, chronic or episodic urticaria, pruritus, flushing, and angioedema. However, HaT is unique because of the coupling of these common dermatologic findings with other abnormalities, including abdominal pain and diarrhea, hypermobile joints, and autonomic dysfunction. Patients with HaT also may manifest psychiatric concerns of anxiety, depression, and chronic pain, all of which have been linked to this disorder.

It is unclear in HaT if the presence of extra-allelic copies of tryptase in an individual is directly pathogenic. The effects of increased basal tryptase and α2β2 tetramers have been shown to likely be responsible for some of the clinical features in these individuals but also may magnify other individual underlying disease(s) or diathesis in which mast cells are naturally abundant.8 In the skin, this increased mast cell activation and subsequent histamine release frequently are visible as dermatographia and urticaria. However, mast cell numbers also are known to be increased in both psoriatic and AD skin lesions,12 thus severe presentation of these diseases in conjunction with the other symptoms associated with mast cell activation should prompt suspicion for HaT.

Effects of HaT on Other Cutaneous Disease—Given the increase of mast cells in AD skin lesions and fact that 94% of patients in the 2014 Lyons et al1 study cited a history of AD, HaT may be a risk factor in the development of AD. Interestingly, in addition to the increased mast cells in AD lesions, PAR2+ nerve fibers also are increased in AD lesions and have been implicated in the nonhistaminergic pruritus experienced by patients with AD.12 Thus, given the proposed propensity for α2β2 tetramers to activate PAR2, it is possible this mechanism may contribute to severe pruritus in individuals with AD and concurrent HaT, as those with HaT express increased α2β2 tetramers. However, no study to date has directly compared AD symptoms in patients with concurrent HaT vs patients without it. Further research is needed on how HaT impacts other allergic and inflammatory skin diseases such as AD and psoriasis, but one may reasonably consider HaT when treating chronic inflammatory skin diseases refractory to typical interventions and/or severe presentations. Although HaT is an autosomal-dominant disorder, it is not detected by standard whole exome sequencing or microarrays. A commercial test is available, utilizing a buccal swab to test for TPSAB1 copy number.

HaT and Mast Cell Disorders—When evaluating someone with suspected HaT, it is important to screen for other symptoms of mast cell activation. For instance, in the GI tract increased mast cell activation results in activation of motor neurons and nociceptors and increases secretion and peristalsis with consequent bloating, abdominal pain, and diarrhea.10 Likewise, tryptase also has neuromodulatory effects that amplify the perception of pain and are likely responsible for the feelings of hyperalgesia reported in patients with HaT.13

 

 

There is substantial overlap in the clinical pictures of HaT and MCAS, and HaT is considered a heritable risk factor for MCAS. Consequently, any patient undergoing workup for MCAS also should be tested for HaT. Although HaT is associated with consistently elevated tryptase, MCAS is episodic in nature, and an increase in tryptase levels of at least 20% plus 2 ng/mL from baseline only in the presence of other symptoms reflective of mast cell activation (Table) is a prerequisite for diagnosis.9 Chronic signs and symptoms of atopy, chronic urticaria, and severe asthma are not indicative of MCAS but are frequently seen in HaT.

Another cause of persistently elevated tryptase levels is SM. Systemic mastocytosis is defined by aberrant clonal mast cell expansion and systemic involvement11 and can cause persistent symptoms, unlike MCAS alone. However, SM also can be associated with MCAS.9 Notably, a baseline serum tryptase level greater than 20 ng/mL—much higher than the threshold of greater than 8 ng/mL for suspicion of HaT—is seen in 75% of SM cases and is part of the minor diagnostic criteria for the disease.9,11 However, the 2016 study identifying increased TPSAB1 α-tryptase–encoding sequences as the causative entity for HaT by Lyons et al2 found the average (SD) basal serum tryptase level in individuals with α-tryptase–encoding sequence duplications to be 15 (5) ng/mL and 24 (6) ng/mL in those with triplications. Thus, there likely is no threshold for elevated baseline tryptase levels that would indicate SM over HaT as a more likely diagnosis. However, SM will present with new persistently elevated tryptase levels, whereas the elevation in HaT is believed to be lifelong.5 Also in contrast to HaT, SM can present with liver, spleen, and lymph node involvement; bone sclerosis; and cytopenia.11,14

Mastocytosis is much rarer than HaT, with an estimated prevalence of 9 cases per 100,000 individuals in the United States.11 Although HaT diagnostic testing is noninvasive, SM requires a bone marrow biopsy for definitive diagnosis. Given the likely much higher prevalence of HaT than SM and the patient burden of a bone marrow biopsy, HaT should be considered before proceeding with a bone marrow biopsy to evaluate for SM when a patient presents with persistent systemic symptoms of mast cell activation and elevated baseline tryptase levels. Furthermore, it also would be prudent to test for HaT in patients with known SM, as a cohort study by Lyons et al5 indicated that HaT is likely more common in those with SM (12.2% [10/82] of cohort with known SM vs 5.3% of 398 controls), and patients with concurrent SM and HaT were at a higher risk for severe anaphylaxis (RR=9.5; P=.007).

Studies thus far surrounding HaT have not evaluated timing of initial symptom onset or age of initial presentation for HaT. Furthermore, there is no guarantee that those with increased TPSAB1 copy number will be symptomatic, as there have been reports of asymptomatic individuals with HaT who had basal serum levels greater than 8 ng/mL.7 As research into HaT continues and larger cohorts are evaluated, questions surrounding timing of symptom onset and various factors that may make someone more likely to display a particular phenotype will be answered.

Treatment—Long-term prognosis for individuals with HaT is largely unknown. Unfortunately, there are limited data to support a single effective treatment strategy for managing HaT, and treatment has varied based on predominant symptoms. For cutaneous and GI tract symptoms, trials of maximal H1 and H2 antihistamines twice daily have been recommended.4 Omalizumab was reported to improve chronic urticaria in 3 of 3 patients, showing potential promise as a treatment.4 Mast cell stabilizers, such as oral cromolyn, have been used for severe GI symptoms, while some patients also have reported improvement with oral ketotifen.6 Other medications, such as tricyclic antidepressants, clemastine fumarate, and gabapentin, have been beneficial anecdotally.6 Given the lack of harmful effects seen in individuals who are α-tryptase deficient, α-tryptase inhibition is an intriguing target for future therapies.

Conclusion

Patients who present with a constellation of dermatologic, allergic, GI tract, neuropsychiatric, respiratory, autonomic, and connective tissue abnormalities consistent with HaT may receive a prompt diagnosis if the association is recognized. The full relationship between HaT and other chronic dermatologic disorders is still unknown. Ultimately, heightened interest and research into HaT will lead to more treatment options available for affected patients.

References

1. Lyons JJ, Sun G, Stone KD, et al. Mendelian inheritance of elevated serum tryptase associated with atopy and connective tissue abnormalities. J Allergy Clin Immunol. 2014;133:1471-1474.

2. Lyons JJ, Yu X, Hughes JD, et al. Elevated basal serum tryptase identifies a multisystem disorder associated with increased TPSAB1 copy number. Nat Genet. 2016;48:1564-1569.

3. Schwartz L. Diagnostic value of tryptase in anaphylaxis and mastocytosis. Immunol Allergy Clin North Am. 2006;6:451-463.

4. Giannetti MP, Weller E, Bormans C, et al. Hereditary alpha-tryptasemia in 101 patients with mast cell activation–related symptomatology including anaphylaxis. Ann Allergy Asthma Immunol. 2021;126:655-660.

5. Lyons JJ, Chovanec J, O’Connell MP, et al. Heritable risk for severe anaphylaxis associated with increased α-tryptase–encoding germline copy number at TPSAB1. J Allergy Clin Immunol. 2020;147:622-632.

6. Lyons JJ. Hereditary alpha tryptasemia: genotyping and associated clinical features. Immunol Allergy Clin North Am. 2018;38:483-495.

7. Robey RC, Wilcock A, Bonin H, et al. Hereditary alpha-tryptasemia: UK prevalence and variability in disease expression. J Allergy Clin Immunol Pract. 2020;8:3549-3556.

8. Le QT, Lyons JJ, Naranjo AN, et al. Impact of naturally forming human α/β-tryptase heterotetramers in the pathogenesis of hereditary α-tryptasemia. J Exp Med. 2019;216:2348-2361.

9. Weiler CR, Austen KF, Akin C, et al. AAAAI Mast Cell Disorders Committee Work Group Report: mast cell activation syndrome (MCAS) diagnosis and management. J Allergy Clin Immunol. 2019;144:883-896.

10. Ramsay DB, Stephen S, Borum M, et al. Mast cells in gastrointestinal disease. Gastroenterol Hepatol (N Y). 2010;6:772-777.

11. Giannetti A, Filice E, Caffarelli C, et al. Mast cell activation disorders. Medicina (Kaunas). 2021;57:124.

12. Siiskonen H, Harvima I. Mast cells and sensory nerves contribute to neurogenic inflammation and pruritus in chronic skin inflammation. Front Cell Neurosci. 2019;13:422.

13. Varrassi G, Fusco M, Skaper SD, et al. A pharmacological rationale to reduce the incidence of opioid induced tolerance and hyperalgesia: a review. Pain Ther. 2018;7:59-75.

14. Núñez E, Moreno-Borque R, García-Montero A, et al. Serum tryptase monitoring in indolent systemic mastocytosis: association with disease features and patient outcome. PLoS One. 2013;8:E76116.

References

1. Lyons JJ, Sun G, Stone KD, et al. Mendelian inheritance of elevated serum tryptase associated with atopy and connective tissue abnormalities. J Allergy Clin Immunol. 2014;133:1471-1474.

2. Lyons JJ, Yu X, Hughes JD, et al. Elevated basal serum tryptase identifies a multisystem disorder associated with increased TPSAB1 copy number. Nat Genet. 2016;48:1564-1569.

3. Schwartz L. Diagnostic value of tryptase in anaphylaxis and mastocytosis. Immunol Allergy Clin North Am. 2006;6:451-463.

4. Giannetti MP, Weller E, Bormans C, et al. Hereditary alpha-tryptasemia in 101 patients with mast cell activation–related symptomatology including anaphylaxis. Ann Allergy Asthma Immunol. 2021;126:655-660.

5. Lyons JJ, Chovanec J, O’Connell MP, et al. Heritable risk for severe anaphylaxis associated with increased α-tryptase–encoding germline copy number at TPSAB1. J Allergy Clin Immunol. 2020;147:622-632.

6. Lyons JJ. Hereditary alpha tryptasemia: genotyping and associated clinical features. Immunol Allergy Clin North Am. 2018;38:483-495.

7. Robey RC, Wilcock A, Bonin H, et al. Hereditary alpha-tryptasemia: UK prevalence and variability in disease expression. J Allergy Clin Immunol Pract. 2020;8:3549-3556.

8. Le QT, Lyons JJ, Naranjo AN, et al. Impact of naturally forming human α/β-tryptase heterotetramers in the pathogenesis of hereditary α-tryptasemia. J Exp Med. 2019;216:2348-2361.

9. Weiler CR, Austen KF, Akin C, et al. AAAAI Mast Cell Disorders Committee Work Group Report: mast cell activation syndrome (MCAS) diagnosis and management. J Allergy Clin Immunol. 2019;144:883-896.

10. Ramsay DB, Stephen S, Borum M, et al. Mast cells in gastrointestinal disease. Gastroenterol Hepatol (N Y). 2010;6:772-777.

11. Giannetti A, Filice E, Caffarelli C, et al. Mast cell activation disorders. Medicina (Kaunas). 2021;57:124.

12. Siiskonen H, Harvima I. Mast cells and sensory nerves contribute to neurogenic inflammation and pruritus in chronic skin inflammation. Front Cell Neurosci. 2019;13:422.

13. Varrassi G, Fusco M, Skaper SD, et al. A pharmacological rationale to reduce the incidence of opioid induced tolerance and hyperalgesia: a review. Pain Ther. 2018;7:59-75.

14. Núñez E, Moreno-Borque R, García-Montero A, et al. Serum tryptase monitoring in indolent systemic mastocytosis: association with disease features and patient outcome. PLoS One. 2013;8:E76116.

Issue
Cutis - 111(1)
Issue
Cutis - 111(1)
Page Number
49-52
Page Number
49-52
Publications
Publications
Topics
Article Type
Display Headline
Cutaneous Manifestations in Hereditary Alpha Tryptasemia
Display Headline
Cutaneous Manifestations in Hereditary Alpha Tryptasemia
Sections
Inside the Article

Practice Points

  • Chronic or episodic urticaria, flushing, and pruritus are the most consistent cutaneous abnormalities associated with hereditary alpha tryptasemia (HaT), but HaT also may augment symptoms of other underlying inflammatory skin disorders, such as atopic dermatitis and psoriasis.
  • Individuals with episodic dermatologic manifestations indicative of mast cell activation accompanied by symptoms affecting 1 or more organ systems should be evaluated for mast cell activation syndrome as well as HaT.
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