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

Elephantiasis Nostras Verrucosa Secondary to Scleroderma

Article Type
Changed
Tue, 02/19/2019 - 08:57
Display Headline
Elephantiasis Nostras Verrucosa Secondary to Scleroderma

To the Editor:

Elephantiasis nostras verrucosa (ENV) is a skin disorder caused by marked underlying lymphedema that leads to hyperkeratosis, papillomatosis, and verrucous growths on the epidermis.1 The pathophysiology of ENV relates to noninfectious lymphatic obstruction and lymphatic fibrosis secondary to venous stasis, malignancy, radiation therapy, or trauma.2 We present an unusual case of lymphedema and subsequent ENV limited to the arms and hands in a patient with scleroderma, an autoimmune fibrosing disorder.

A 54-year-old woman with a 5-year history of scleroderma presented to our dermatology clinic for treatment of progressive skin changes including pruritus, tightness, finger ulcerations, and pus exuding from papules on the dorsal arms and hands. She had been experiencing several systemic symptoms including dysphagia and lung involvement, necessitating oxygen therapy and a continuous positive airway pressure device for pulmonary arterial hypertension. A computed tomography scan of the lungs demonstrated an increase in ground-glass infiltrates in the right lower lobe and an air-fluid level in the esophagus. At the time of presentation, she was being treated with bosentan and sildenafil for pulmonary arterial hypertension, in addition to prednisone, venlafaxine, lansoprazole, metoclopramide, levothyroxine, temazepam, aspirin, and oxycodone. In the 2 years prior to presentation, she had been treated with intravenous cyclophosphamide once monthly for 6 months, adalimumab for 1 year, and 1 session of photodynamic therapy to the arms, all without benefit.



Physical examination showed cutaneous signs of scleroderma including marked sclerosis of the skin on the face, hands, V of the neck, proximal arms, and mid and proximal thighs. Excoriated papules with overlying crusting and pustulation were superimposed on the sclerotic skin of the arms (Figure 1).

Figure 1. Left forearm sclerosis with overlying papules and crust in a patient with elephantiasis nostras verrucosa.


A superinfection was diagnosed and treated with cephalexin 500 mg 4 times daily for 2 weeks; thereafter, mupirocin cream twice daily was used as needed. She was prescribed fexofenadine 180 mg twice daily and doxepin 20 mg at bedtime for pruritus.

 

 

At 3-week follow-up, a trial of narrowband UVB therapy was recommended for control of pruritus. Two weeks later, a modified wet-wrap regimen using clobetasol ointment 0.5% twice daily covered with wet gauze followed by a self-adherent dressing was initiated only on the right arm for comparison purposes. This treatment was not successful. A biopsy taken from the left arm showed lymphedema with perivascular fibroplasia and epidermal hyperplasia consistent with ENV (Figure 2).

Figure 2. Biopsy of the left arm showed lymphedema with perivascular fibroplasia and epidermal hyperplasia consistent with elephantiasis nostras verrucosa (H&E, original magnification ×40).


Two months after her initial presentation, we instituted treatment with tazarotene gel 0.1% twice daily to the arms as well as a water-based topical emulsion to the finger ulcerations and a healing ointment to the hands. A month later, the patient reported no benefit with tazarotene. She desired more flexibility in her arms and hands; therefore, after a discussion with her rheumatologist, biweekly psoralen plus UVA (PUVA) therapy was initiated. Five months after presentation, methotrexate (MTX) 15 mg once weekly with folic acid 1 mg once daily was added. The PUVA therapy and MTX were stopped 3 months later due to lack of treatment benefit.

The patient was referred to vascular medicine for possible compression therapy. It was determined that her vasculature was intact, but compression therapy was contraindicated due to underlying systemic sclerosis. She was subsequently prescribed mycophenolate mofetil 1000 mg twice daily by her rheumatologist. The options of serial excisions or laser resurfacing were presented, but she declined.

Elephantiasis nostras verrucosa is differentiated from elephantiasis tropica, which is caused by a filarial infection of the lymphatic system. The chronic obstructive lymphedema characteristic of ENV can present as a result of various primary or secondary etiologies including trauma, malignancy, venous stasis, inflammation, or infection.3 In systemic sclerosis, extravascular fibrosis theoretically can lead to lymphatic obstruction and subsequent lymphatic stasis. In turn, the pathophysiology of dermal and subcutaneous fibrosis likely reflects autoantibodies (eg, anticardiolipin antibodies) that can damage lymphatic and nonlymphatic vessels.4,5 With prolonged lymphostasis, excess protein-rich interstitial fluid accumulates, inducing fibroblast proliferation and inhibiting the local immune response.1

As the underlying mechanism of ENV, fibrosis of lymphatic vessels in systemic sclerosis is not well documented. Characteristic features of systemic sclerosis include extensive fibrosis, fibroproliferative vasculopathy, and inflammation, which are all possible mechanisms for the internal lymphatic obstruction resulting in the skin changes observed in ENV.6 It seemed unusual that the fibrotic changes of lymphatic vessels in our patient were extensive enough to cause ENV of the upper extremities; lower extremity involvement is the more common presentation because of the greater likelihood of lymphedema manifesting in the legs and feet. Lower extremity ENV has been reported in association with scleroderma.7,8

Regarding therapeutic options, Boyd et al9 reported a good response in a patient with ENV of the abdomen who was treated with topical tazarotene. Additionally, PUVA and MTX have been reported to be beneficial for the progressive skin changes of systemic sclerosis.10 Mycophenolate mofetil has been used in patients who fail MTX therapy because of its antifibrotic properties without the side-effect profiles of other immunosuppressives, such as imatinib.10,11 In our patient, skin lesions persisted following these varied approaches, and compression therapy was not advised due to the underlying sclerosis.



Because options for medical treatment of severe ENV are limited, surgical debridement of the affected limb often remains the only viable option in advanced cases.12 A PubMed search of articles indexed for MEDLINE using the terms elephantiasis (MeSH terms) or elephantiasis (all fields) and scleroderma, systemic (MeSH terms) or scleroderma (all fields) and systemic (all fields) or systemic scleroderma (all fields) or scleroderma (all fields) or scleroderma, localized (MeSH terms) or scleroderma (all fields) and localized (all fields) or localized scleroderma (all fields) yielded only 1 other case report of lower extremity ENV in a patient with systemic sclerosis who ultimately required bilateral leg amputation.8 When possible, avoiding lymphostasis through compression and control of any underlying infections is important in the treatment and prevention of ENV.3

References
  1. Sisto K, Khachemoune A. Elephantiasis nostras verrucosa: a review. Am J Clin Dermatol. 2008;9:141-146.
  2. Schissel DJ, Hivnor C, Elston DM. Elephantiasis nostras verrucosa. Cutis. 1998;62:77-80.
  3. Duckworth A, Husain J, DeHeer P. Elephantiasis nostras verrucosa or ‘mossy foot lesions’ in lymphedema praecox. J Am Podiatr Med Assoc. 2008;98:66-69.
  4. Assous N, Allanore Y, Batteaux F, et al. Prevalence of antiphospholipid antibodies in systemic sclerosis and association with primitive pulmonary arterial hypertension and endothelial injury. Clin Exp Rheumatol. 2005;23:199-204.
  5. Derrett-Smith EC, Dooley A, Gilbane AJ, et al. Endothelial injury in a transforming growth-factor-dependent mouse model of scleroderma induces pulmonary arterial hypertension. Arthritis Rheum. 2013;65:2928-2939.
  6. Pattanaik M, Brown M, Postlethwaite A. Vascular involvement in systemic sclerosis (scleroderma). J Inflamm Res. 2011;4:105-125.
  7. Kerchner K, Fleischer A, Yosipovitch G. Lower extremity lymphedema update: pathophysiology, diagnosis and treatment guidelines. J Am Acad Dermatol. 2008;59:324-331.
  8. Chatterjee S, Karai L. Elephantiasis nostras verrucosa in a patient with systemic sclerosis. Clin Exp Dermatol. 2009;34:e696-e698.
  9. Boyd J, Sloan S, Meffert J. Elephantiasis nostrum verrucosa of the abdomen: clinical results with tazarotene. J Drugs Dermatol. 2004;3:446-448.
  10. Fett, N. Scleroderma: nomenclature, etiology, pathogenesis, prognosis, and treatments: facts and controversies. Clin Dermatol. 2013;31:432-437.
  11. Moinzadeh P, Krieg T, Hunzelmann N. Imatinib treatment of generalized localized scleroderma (morphea). J Am Acad Dermatol. 2010;63:e102-e104.
  12. Iwao F, Sato-Matsumura KC, Sawamura D, et al. Elephantiasis nostras verrucosa successfully treated by surgical debridement. Dermatol Surg. 2004;30:939-941.
Article PDF
Author and Disclosure Information

Dr. Lee is from the Section of Dermatology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire. Dr. Bershow is from the Department of Dermatology, Minneapolis VA Healthcare System. Dr. Crowson is from the Department of Dermatopathology, University of Oklahoma and Regional Medical Laboratory, St. John Medical Center, Tulsa.

The authors report no conflict of interest.

The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of Veterans Affairs or the US government.

Correspondence: Andrea Bershow, MD, 1 Veterans Dr, Department 111K, Minneapolis, MN 55417 ([email protected]).

Issue
Cutis - 103(2)
Publications
Topics
Page Number
E4-E6
Sections
Author and Disclosure Information

Dr. Lee is from the Section of Dermatology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire. Dr. Bershow is from the Department of Dermatology, Minneapolis VA Healthcare System. Dr. Crowson is from the Department of Dermatopathology, University of Oklahoma and Regional Medical Laboratory, St. John Medical Center, Tulsa.

The authors report no conflict of interest.

The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of Veterans Affairs or the US government.

Correspondence: Andrea Bershow, MD, 1 Veterans Dr, Department 111K, Minneapolis, MN 55417 ([email protected]).

Author and Disclosure Information

Dr. Lee is from the Section of Dermatology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire. Dr. Bershow is from the Department of Dermatology, Minneapolis VA Healthcare System. Dr. Crowson is from the Department of Dermatopathology, University of Oklahoma and Regional Medical Laboratory, St. John Medical Center, Tulsa.

The authors report no conflict of interest.

The views expressed in this article are those of the authors and do not necessarily reflect the official policy or position of the Department of Veterans Affairs or the US government.

Correspondence: Andrea Bershow, MD, 1 Veterans Dr, Department 111K, Minneapolis, MN 55417 ([email protected]).

Article PDF
Article PDF

To the Editor:

Elephantiasis nostras verrucosa (ENV) is a skin disorder caused by marked underlying lymphedema that leads to hyperkeratosis, papillomatosis, and verrucous growths on the epidermis.1 The pathophysiology of ENV relates to noninfectious lymphatic obstruction and lymphatic fibrosis secondary to venous stasis, malignancy, radiation therapy, or trauma.2 We present an unusual case of lymphedema and subsequent ENV limited to the arms and hands in a patient with scleroderma, an autoimmune fibrosing disorder.

A 54-year-old woman with a 5-year history of scleroderma presented to our dermatology clinic for treatment of progressive skin changes including pruritus, tightness, finger ulcerations, and pus exuding from papules on the dorsal arms and hands. She had been experiencing several systemic symptoms including dysphagia and lung involvement, necessitating oxygen therapy and a continuous positive airway pressure device for pulmonary arterial hypertension. A computed tomography scan of the lungs demonstrated an increase in ground-glass infiltrates in the right lower lobe and an air-fluid level in the esophagus. At the time of presentation, she was being treated with bosentan and sildenafil for pulmonary arterial hypertension, in addition to prednisone, venlafaxine, lansoprazole, metoclopramide, levothyroxine, temazepam, aspirin, and oxycodone. In the 2 years prior to presentation, she had been treated with intravenous cyclophosphamide once monthly for 6 months, adalimumab for 1 year, and 1 session of photodynamic therapy to the arms, all without benefit.



Physical examination showed cutaneous signs of scleroderma including marked sclerosis of the skin on the face, hands, V of the neck, proximal arms, and mid and proximal thighs. Excoriated papules with overlying crusting and pustulation were superimposed on the sclerotic skin of the arms (Figure 1).

Figure 1. Left forearm sclerosis with overlying papules and crust in a patient with elephantiasis nostras verrucosa.


A superinfection was diagnosed and treated with cephalexin 500 mg 4 times daily for 2 weeks; thereafter, mupirocin cream twice daily was used as needed. She was prescribed fexofenadine 180 mg twice daily and doxepin 20 mg at bedtime for pruritus.

 

 

At 3-week follow-up, a trial of narrowband UVB therapy was recommended for control of pruritus. Two weeks later, a modified wet-wrap regimen using clobetasol ointment 0.5% twice daily covered with wet gauze followed by a self-adherent dressing was initiated only on the right arm for comparison purposes. This treatment was not successful. A biopsy taken from the left arm showed lymphedema with perivascular fibroplasia and epidermal hyperplasia consistent with ENV (Figure 2).

Figure 2. Biopsy of the left arm showed lymphedema with perivascular fibroplasia and epidermal hyperplasia consistent with elephantiasis nostras verrucosa (H&E, original magnification ×40).


Two months after her initial presentation, we instituted treatment with tazarotene gel 0.1% twice daily to the arms as well as a water-based topical emulsion to the finger ulcerations and a healing ointment to the hands. A month later, the patient reported no benefit with tazarotene. She desired more flexibility in her arms and hands; therefore, after a discussion with her rheumatologist, biweekly psoralen plus UVA (PUVA) therapy was initiated. Five months after presentation, methotrexate (MTX) 15 mg once weekly with folic acid 1 mg once daily was added. The PUVA therapy and MTX were stopped 3 months later due to lack of treatment benefit.

The patient was referred to vascular medicine for possible compression therapy. It was determined that her vasculature was intact, but compression therapy was contraindicated due to underlying systemic sclerosis. She was subsequently prescribed mycophenolate mofetil 1000 mg twice daily by her rheumatologist. The options of serial excisions or laser resurfacing were presented, but she declined.

Elephantiasis nostras verrucosa is differentiated from elephantiasis tropica, which is caused by a filarial infection of the lymphatic system. The chronic obstructive lymphedema characteristic of ENV can present as a result of various primary or secondary etiologies including trauma, malignancy, venous stasis, inflammation, or infection.3 In systemic sclerosis, extravascular fibrosis theoretically can lead to lymphatic obstruction and subsequent lymphatic stasis. In turn, the pathophysiology of dermal and subcutaneous fibrosis likely reflects autoantibodies (eg, anticardiolipin antibodies) that can damage lymphatic and nonlymphatic vessels.4,5 With prolonged lymphostasis, excess protein-rich interstitial fluid accumulates, inducing fibroblast proliferation and inhibiting the local immune response.1

As the underlying mechanism of ENV, fibrosis of lymphatic vessels in systemic sclerosis is not well documented. Characteristic features of systemic sclerosis include extensive fibrosis, fibroproliferative vasculopathy, and inflammation, which are all possible mechanisms for the internal lymphatic obstruction resulting in the skin changes observed in ENV.6 It seemed unusual that the fibrotic changes of lymphatic vessels in our patient were extensive enough to cause ENV of the upper extremities; lower extremity involvement is the more common presentation because of the greater likelihood of lymphedema manifesting in the legs and feet. Lower extremity ENV has been reported in association with scleroderma.7,8

Regarding therapeutic options, Boyd et al9 reported a good response in a patient with ENV of the abdomen who was treated with topical tazarotene. Additionally, PUVA and MTX have been reported to be beneficial for the progressive skin changes of systemic sclerosis.10 Mycophenolate mofetil has been used in patients who fail MTX therapy because of its antifibrotic properties without the side-effect profiles of other immunosuppressives, such as imatinib.10,11 In our patient, skin lesions persisted following these varied approaches, and compression therapy was not advised due to the underlying sclerosis.



Because options for medical treatment of severe ENV are limited, surgical debridement of the affected limb often remains the only viable option in advanced cases.12 A PubMed search of articles indexed for MEDLINE using the terms elephantiasis (MeSH terms) or elephantiasis (all fields) and scleroderma, systemic (MeSH terms) or scleroderma (all fields) and systemic (all fields) or systemic scleroderma (all fields) or scleroderma (all fields) or scleroderma, localized (MeSH terms) or scleroderma (all fields) and localized (all fields) or localized scleroderma (all fields) yielded only 1 other case report of lower extremity ENV in a patient with systemic sclerosis who ultimately required bilateral leg amputation.8 When possible, avoiding lymphostasis through compression and control of any underlying infections is important in the treatment and prevention of ENV.3

To the Editor:

Elephantiasis nostras verrucosa (ENV) is a skin disorder caused by marked underlying lymphedema that leads to hyperkeratosis, papillomatosis, and verrucous growths on the epidermis.1 The pathophysiology of ENV relates to noninfectious lymphatic obstruction and lymphatic fibrosis secondary to venous stasis, malignancy, radiation therapy, or trauma.2 We present an unusual case of lymphedema and subsequent ENV limited to the arms and hands in a patient with scleroderma, an autoimmune fibrosing disorder.

A 54-year-old woman with a 5-year history of scleroderma presented to our dermatology clinic for treatment of progressive skin changes including pruritus, tightness, finger ulcerations, and pus exuding from papules on the dorsal arms and hands. She had been experiencing several systemic symptoms including dysphagia and lung involvement, necessitating oxygen therapy and a continuous positive airway pressure device for pulmonary arterial hypertension. A computed tomography scan of the lungs demonstrated an increase in ground-glass infiltrates in the right lower lobe and an air-fluid level in the esophagus. At the time of presentation, she was being treated with bosentan and sildenafil for pulmonary arterial hypertension, in addition to prednisone, venlafaxine, lansoprazole, metoclopramide, levothyroxine, temazepam, aspirin, and oxycodone. In the 2 years prior to presentation, she had been treated with intravenous cyclophosphamide once monthly for 6 months, adalimumab for 1 year, and 1 session of photodynamic therapy to the arms, all without benefit.



Physical examination showed cutaneous signs of scleroderma including marked sclerosis of the skin on the face, hands, V of the neck, proximal arms, and mid and proximal thighs. Excoriated papules with overlying crusting and pustulation were superimposed on the sclerotic skin of the arms (Figure 1).

Figure 1. Left forearm sclerosis with overlying papules and crust in a patient with elephantiasis nostras verrucosa.


A superinfection was diagnosed and treated with cephalexin 500 mg 4 times daily for 2 weeks; thereafter, mupirocin cream twice daily was used as needed. She was prescribed fexofenadine 180 mg twice daily and doxepin 20 mg at bedtime for pruritus.

 

 

At 3-week follow-up, a trial of narrowband UVB therapy was recommended for control of pruritus. Two weeks later, a modified wet-wrap regimen using clobetasol ointment 0.5% twice daily covered with wet gauze followed by a self-adherent dressing was initiated only on the right arm for comparison purposes. This treatment was not successful. A biopsy taken from the left arm showed lymphedema with perivascular fibroplasia and epidermal hyperplasia consistent with ENV (Figure 2).

Figure 2. Biopsy of the left arm showed lymphedema with perivascular fibroplasia and epidermal hyperplasia consistent with elephantiasis nostras verrucosa (H&E, original magnification ×40).


Two months after her initial presentation, we instituted treatment with tazarotene gel 0.1% twice daily to the arms as well as a water-based topical emulsion to the finger ulcerations and a healing ointment to the hands. A month later, the patient reported no benefit with tazarotene. She desired more flexibility in her arms and hands; therefore, after a discussion with her rheumatologist, biweekly psoralen plus UVA (PUVA) therapy was initiated. Five months after presentation, methotrexate (MTX) 15 mg once weekly with folic acid 1 mg once daily was added. The PUVA therapy and MTX were stopped 3 months later due to lack of treatment benefit.

The patient was referred to vascular medicine for possible compression therapy. It was determined that her vasculature was intact, but compression therapy was contraindicated due to underlying systemic sclerosis. She was subsequently prescribed mycophenolate mofetil 1000 mg twice daily by her rheumatologist. The options of serial excisions or laser resurfacing were presented, but she declined.

Elephantiasis nostras verrucosa is differentiated from elephantiasis tropica, which is caused by a filarial infection of the lymphatic system. The chronic obstructive lymphedema characteristic of ENV can present as a result of various primary or secondary etiologies including trauma, malignancy, venous stasis, inflammation, or infection.3 In systemic sclerosis, extravascular fibrosis theoretically can lead to lymphatic obstruction and subsequent lymphatic stasis. In turn, the pathophysiology of dermal and subcutaneous fibrosis likely reflects autoantibodies (eg, anticardiolipin antibodies) that can damage lymphatic and nonlymphatic vessels.4,5 With prolonged lymphostasis, excess protein-rich interstitial fluid accumulates, inducing fibroblast proliferation and inhibiting the local immune response.1

As the underlying mechanism of ENV, fibrosis of lymphatic vessels in systemic sclerosis is not well documented. Characteristic features of systemic sclerosis include extensive fibrosis, fibroproliferative vasculopathy, and inflammation, which are all possible mechanisms for the internal lymphatic obstruction resulting in the skin changes observed in ENV.6 It seemed unusual that the fibrotic changes of lymphatic vessels in our patient were extensive enough to cause ENV of the upper extremities; lower extremity involvement is the more common presentation because of the greater likelihood of lymphedema manifesting in the legs and feet. Lower extremity ENV has been reported in association with scleroderma.7,8

Regarding therapeutic options, Boyd et al9 reported a good response in a patient with ENV of the abdomen who was treated with topical tazarotene. Additionally, PUVA and MTX have been reported to be beneficial for the progressive skin changes of systemic sclerosis.10 Mycophenolate mofetil has been used in patients who fail MTX therapy because of its antifibrotic properties without the side-effect profiles of other immunosuppressives, such as imatinib.10,11 In our patient, skin lesions persisted following these varied approaches, and compression therapy was not advised due to the underlying sclerosis.



Because options for medical treatment of severe ENV are limited, surgical debridement of the affected limb often remains the only viable option in advanced cases.12 A PubMed search of articles indexed for MEDLINE using the terms elephantiasis (MeSH terms) or elephantiasis (all fields) and scleroderma, systemic (MeSH terms) or scleroderma (all fields) and systemic (all fields) or systemic scleroderma (all fields) or scleroderma (all fields) or scleroderma, localized (MeSH terms) or scleroderma (all fields) and localized (all fields) or localized scleroderma (all fields) yielded only 1 other case report of lower extremity ENV in a patient with systemic sclerosis who ultimately required bilateral leg amputation.8 When possible, avoiding lymphostasis through compression and control of any underlying infections is important in the treatment and prevention of ENV.3

References
  1. Sisto K, Khachemoune A. Elephantiasis nostras verrucosa: a review. Am J Clin Dermatol. 2008;9:141-146.
  2. Schissel DJ, Hivnor C, Elston DM. Elephantiasis nostras verrucosa. Cutis. 1998;62:77-80.
  3. Duckworth A, Husain J, DeHeer P. Elephantiasis nostras verrucosa or ‘mossy foot lesions’ in lymphedema praecox. J Am Podiatr Med Assoc. 2008;98:66-69.
  4. Assous N, Allanore Y, Batteaux F, et al. Prevalence of antiphospholipid antibodies in systemic sclerosis and association with primitive pulmonary arterial hypertension and endothelial injury. Clin Exp Rheumatol. 2005;23:199-204.
  5. Derrett-Smith EC, Dooley A, Gilbane AJ, et al. Endothelial injury in a transforming growth-factor-dependent mouse model of scleroderma induces pulmonary arterial hypertension. Arthritis Rheum. 2013;65:2928-2939.
  6. Pattanaik M, Brown M, Postlethwaite A. Vascular involvement in systemic sclerosis (scleroderma). J Inflamm Res. 2011;4:105-125.
  7. Kerchner K, Fleischer A, Yosipovitch G. Lower extremity lymphedema update: pathophysiology, diagnosis and treatment guidelines. J Am Acad Dermatol. 2008;59:324-331.
  8. Chatterjee S, Karai L. Elephantiasis nostras verrucosa in a patient with systemic sclerosis. Clin Exp Dermatol. 2009;34:e696-e698.
  9. Boyd J, Sloan S, Meffert J. Elephantiasis nostrum verrucosa of the abdomen: clinical results with tazarotene. J Drugs Dermatol. 2004;3:446-448.
  10. Fett, N. Scleroderma: nomenclature, etiology, pathogenesis, prognosis, and treatments: facts and controversies. Clin Dermatol. 2013;31:432-437.
  11. Moinzadeh P, Krieg T, Hunzelmann N. Imatinib treatment of generalized localized scleroderma (morphea). J Am Acad Dermatol. 2010;63:e102-e104.
  12. Iwao F, Sato-Matsumura KC, Sawamura D, et al. Elephantiasis nostras verrucosa successfully treated by surgical debridement. Dermatol Surg. 2004;30:939-941.
References
  1. Sisto K, Khachemoune A. Elephantiasis nostras verrucosa: a review. Am J Clin Dermatol. 2008;9:141-146.
  2. Schissel DJ, Hivnor C, Elston DM. Elephantiasis nostras verrucosa. Cutis. 1998;62:77-80.
  3. Duckworth A, Husain J, DeHeer P. Elephantiasis nostras verrucosa or ‘mossy foot lesions’ in lymphedema praecox. J Am Podiatr Med Assoc. 2008;98:66-69.
  4. Assous N, Allanore Y, Batteaux F, et al. Prevalence of antiphospholipid antibodies in systemic sclerosis and association with primitive pulmonary arterial hypertension and endothelial injury. Clin Exp Rheumatol. 2005;23:199-204.
  5. Derrett-Smith EC, Dooley A, Gilbane AJ, et al. Endothelial injury in a transforming growth-factor-dependent mouse model of scleroderma induces pulmonary arterial hypertension. Arthritis Rheum. 2013;65:2928-2939.
  6. Pattanaik M, Brown M, Postlethwaite A. Vascular involvement in systemic sclerosis (scleroderma). J Inflamm Res. 2011;4:105-125.
  7. Kerchner K, Fleischer A, Yosipovitch G. Lower extremity lymphedema update: pathophysiology, diagnosis and treatment guidelines. J Am Acad Dermatol. 2008;59:324-331.
  8. Chatterjee S, Karai L. Elephantiasis nostras verrucosa in a patient with systemic sclerosis. Clin Exp Dermatol. 2009;34:e696-e698.
  9. Boyd J, Sloan S, Meffert J. Elephantiasis nostrum verrucosa of the abdomen: clinical results with tazarotene. J Drugs Dermatol. 2004;3:446-448.
  10. Fett, N. Scleroderma: nomenclature, etiology, pathogenesis, prognosis, and treatments: facts and controversies. Clin Dermatol. 2013;31:432-437.
  11. Moinzadeh P, Krieg T, Hunzelmann N. Imatinib treatment of generalized localized scleroderma (morphea). J Am Acad Dermatol. 2010;63:e102-e104.
  12. Iwao F, Sato-Matsumura KC, Sawamura D, et al. Elephantiasis nostras verrucosa successfully treated by surgical debridement. Dermatol Surg. 2004;30:939-941.
Issue
Cutis - 103(2)
Issue
Cutis - 103(2)
Page Number
E4-E6
Page Number
E4-E6
Publications
Publications
Topics
Article Type
Display Headline
Elephantiasis Nostras Verrucosa Secondary to Scleroderma
Display Headline
Elephantiasis Nostras Verrucosa Secondary to Scleroderma
Sections
Inside the Article

Practice Points

  • Scleroderma rarely may lead to elephantiasis nostras verrucosa (ENV) of the upper extremities.
  • Avoiding lymphostasis through compression and control of concomitant skin and soft tissue infections is important in the treatment and prevention of ENV.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Neurofibromatosis Type 1 in the Setting of Systemic Lupus Erythematosus

Article Type
Changed
Tue, 02/19/2019 - 09:02
Display Headline
Neurofibromatosis Type 1 in the Setting of Systemic Lupus Erythematosus

To the Editor:

Patients with concurrent neurofibromatosis type 1 (NF-1) and systemic lupus erythematosus (SLE) rarely have been reported in the literature. Neurofibromatosis type 1 is one of the most common genetic disorders, with a worldwide birth incidence of 1 in 2500 individuals and prevalence of 1 in 4000 individuals.1 The incidence and prevalence of SLE varies widely depending on race and geographic location. Estimated incidence rates for SLE range from 1 to 25 per 100,000 individuals annually in North America, South America, Europe, and Asia.2,3 The reported worldwide prevalence is 20 to 150 cases per 100,000 individuals annually.2,4,5

Given the high prevalence of both conditions, the association between SLE and NF-1 likely is underrecognized; therefore, identifying more patients with concurrent SLE and NF-1 and describing the interplay between the 2 conditions may have important therapeutic implications. We present the case of a middle-aged woman with a history of SLE who had cutaneous lesions characteristic of NF-1 to further the understanding of these concurrent conditions.



A middle-aged woman presented to our academic dermatology clinic for evaluation and removal of dark spots that had been present diffusely on the trunk and extremities since birth. She reported a history of SLE with lupus nephritis, hypertension, and a nodular goiter following a partial thyroidectomy. She noted that she did not seek treatment for the skin findings sooner because she was more concerned about her other medical conditions; however, because she felt these conditions were now stable, she decided to seek treatment for the “rash.” Physical examination revealed hundreds of café au lait macules and numerous neurofibromas diffusely distributed on the trunk and extremities (Figure 1) as well as bilateral axillary freckling. A clinical diagnosis of NF-1 was made.

Figure 1. Café au lait macules and neurofibromas on the upper back.


When questioned, the patient reported that she may have been diagnosed with NF-1 in the past by another physician, but she did not recall it specifically. The patient was advised that there were no treatments for the café au lait macules. We notified her other physicians of the NF-1 diagnosis so she could be monitored for systemic conditions related to NF-1, including optic gliomas, pheochromocytoma, renal artery stenosis, and internal neurofibromas. We also referred the patient for genetic counseling; of note, the patient reported she had 4 children without any evidence of similar skin lesions or chronic health problems.

 

 

A PubMed search of articles indexed for MEDLINE using the terms systemic lupus and neurofibromatosis yielded 8 cases of patients having both SLE and NF-1 (including our case).6-11 Our patient reported having multiple lesions since birth, decades before the onset and diagnosis of SLE. In 3 other cases, patients were diagnosed with SLE and then presented with neurofibromas, leading to NF-1 diagnosis.In the discussion of those 3 cases, it was proposed that immune system alterations caused by SLE leading to viral illness may have predisposed the patients to the development of tumors and other collagen diseases, or it could be coincidental.6,7 In another case, a patient with NF-1 developed SLE, which was thought to be coincidental.8 Akyuz et al9 described the case of a pediatric patient with NF-1 who subsequently was diagnosed with SLE. The authors suggested that the lack of neurofibromin contributed to the development of SLE, an autoimmune condition. Under normal circumstances, neurofibromin acts as a guanosine triphosphatase–activating protein for RAS in T cells.10 CD8+ T-cell function also is impaired in patients with SLE.9 Additionally, it has been reported that anti–double-stranded DNA antibodies and immune complexes were present in NF-1 patients, even though there were low titers.12 Thus, the authors proposed that the lack of neurofibromin led to dysregulation of the RAS pathway and impairment of T cells, creating an immune milieu that predisposed the patient to development of SLE. Our case gives additional credence to this theory, as our patient had a similar clinical course: the café au lait macules were present since birth and the symptoms of SLE surfaced much later in her late 20s and 30s. Another case by Makino and Tampo10 described a patient with a history of SLE who was later diagnosed with NF-1 based on choroidal findings highly specific for NF-1 but did not have other classic findings of NF-1. The authors mentioned that there might be a potential relationship between these two disorders but did not speculate any theory in particular for their case.10



The interplay between an autoimmune condition such as SLE and NF-1, a condition traditionally thought to be due to a genetic mutation, may have greater clinical and therapeutic implications beyond just these two disorders. Although it is well established that RAS pathway disruption causes NF-1, it has been uncovered that dysfunction in the RAS pathway also can contribute to melanoma oncogenesis.13,14 These insights have led to the development of and approval of targeted drugs designed to inhibit the RAS pathway (eg, vemurafenib, dabrafenib, trametinib).14-17 Melanoma also is considered a “model” tumor for studying the relationship between the immune system and cancer.18AKT is a signal transduction pathway that promotes cell survival and growth in various cancers.15 In addition, deactivation of MEK (part of the RAS pathway) can cause activation of AKT (protein kinase B) signaling and lupus like autoimmune conditions19 (Figure 2). Likewise, an understanding of the RAS pathway and T-cell function in patients with both SLE and NF-1 may give us more information about melanoma and other cancers.

Figure 2. MAPK pathways and potential interplay between neurofibromatosis type 1, systemic lupus erythematosus, and melanoma. Mutated BRAF leads to activation of the MEK/ERK pathway and development of melanoma. MEK-1 inhibition results in activation of P13K/AKT signaling and breach of peripheral tolerance and development of lupuslike autoimmune disease. Both neurofibromatosis type 1 and systemic lupus erythematosus are related to activated RAS. Neurofibromin 1 (NF1) leads to phosphorylation of RAS-GDP, resulting in the activated form of RAS (RAS-GTP).


Our case also is instructive in another point: our patient had never sought treatment for her skin lesions, as she said she had other more serious health conditions. Closer evaluation of her skin condition may have led to earlier diagnosis of NF-1, which has important health implications. The average lifespan of patients with NF-1 is 10 to 15 years lower than the general population, with cancer being the leading cause of death.20 Malignant peripheral nerve sheath tumors are the most common malignant tumors observed in such patients.21-23 Other cancers that are associated with NF-1 include rhabdomyosarcomas, gastrointestinal stromal tumors, neuroectodermal tumors, pheochromocytomas, and breast carcinomas.23

To make a clinical diagnosis of NF-1, a patient must have 2 of 7 cardinal clinical features as defined by the National Institutes of Health (Table).24 In our patient with hundreds of café au lait macules and dozens of neurofibromas, the diagnosis was clear; however, in other patients, the skin findings of NF-1 may not be as prominent. A patient could meet criteria for NF-1 diagnosis with the inconspicuous presentation of 6 café au lait macules and either 1 plexiform neurofibroma or 2 neurofibromas (of any type) on the entire body.



We recommend that patients with SLE undergo skin examinations to look for more subtle presentations of NF-1. Earlier diagnosis will help to initiate close monitoring of the disorder’s associated systemic health risks. In addition, the identification of more patients with both NF-1 and SLE may help shed light on the etiology of both conditions.

References
  1. Carey JC, Baty BJ, Johnson JP, et al. The genetic aspects of neurofibromatosis. Ann N Y Acad Sci. 1986;486:45-56.
  2. Pons-Estel GJ, Alarcón GS, Scofield L, et al. Understanding the epidemiology and progression of systemic lupus erythematosus. Semin Arthritis Rheum. 2010;39:257-268.
  3. Danchenko N, Satia JA, Anthony MS. Epidemiology of systemic lupus erythematosus: a comparison of worldwide disease burden. Lupus. 2006;15:308-318.
  4. Lawrence RC, Helmick CG, Arnett FC, et al. Estimates of the prevalence of arthritis and selected musculoskeletal disorders in the United States. Arthritis Rheum. 1998;41:778-799.
  5. Chakravarty EF, Bush TM, Manzi S, et al. Prevalence of adult systemic lupus erythematosus in California and Pennsylvania in 2000: estimates obtained using hospitalization data. Arthritis Rheum. 2007;56:2092-2094.
  6. Bitnun S, Bassan H. Letter: neurofibromatosis and SLE. N Engl J Med. 1975;292:429-430.
  7. Riccardi VM. Neurofibromatosis in a patient with systemic lupus erythematosus. Arthritis Rheum. 1983;26:574.
  8. Corominas H, Guardiola JM, Matas L, et al. Neurofibromatosis and systemic lupus erythematosus. a matter of coincidence? Clin Rhematol. 2003;22:496-497.
  9. Akyuz SG, Caltik A, Bulbul M, et al. An unusual pediatric case with neurofibromatosis and systemic lupus erythematosus. Rheumatol Int. 2012;32:2345-47.
  10. Makino S, Tampo H. Rare and unusual choroidal abnormalities in a patient with systemic lupus erythematosus. Case Rep Ophthalmol. 2013;4:81-86.
  11. Galvan JM, Hofkamp MP. Usefulness of intrapartum magnetic resonance imaging for a parturient with neurofibromatosis type I during induction of labor for preeclampsia. Proc (Bayl Univ Med Cent). 2018;31:92-93.
  12. Gerosa PL, Vai C, Bizzozer L, et al. Immunological and clinical surveillance in Recklinghausen’s neurofibromatosis (NF1). Panminerva Med. 1993;35:80-85.
  13. Busca R, Abbe P, Mantoux F, et al. RAS mediates the cAMP-dependent activation of extracellular signal-regulated kinases (ERKs) in melanocytes. EMBO J. 2000;19:2900-2910.
  14. Sullivan RJ, Flaherty K. MAP kinase signaling and inhibition in melanoma. Oncogene. 2013;32:2373-2379.
  15. Hennessy BT, Smith DL, Ram PT, et al. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005;12:988-1004.
  16. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507-2516.
  17. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380:358-365.
  18. Maio M. Melanoma as a model tumour for immuno-oncology. Ann Oncol. 2012;23:(suppl 8):viii10-4.
  19. Zmajkovicova K, Jesenberger V, Catalanotti F, et al. MEK1 is required for PTEN membrane recruitment, AKT regulation, and the maintenance of peripheral tolerance. Mol Cell. 2013;50:43-55.
  20. Patil S, Chamberlain RS. Neoplasms associated with germline and somatic NF1 gene mutations. Oncologist. 2012;17:101-116.
  21. Carroll SL, Ratner N. How does the Schwann cell lineage form tumors in NF1? Glia. 2008;56:1590-1605.
  22. Rasmussen SA, Friedman JM. NF1 gene and neurofibromatosis 1. Am J Epidemiol. 2000;151:33-40.
  23. Yohay K. Neurofibromatosis type 1 and associated malignancies. Curr Neurol Neurosci Rep. 2009;9:247-253.
  24. Neurofibromatosis. conference statement. National Institutes of Health Consensus Development Conference. Arch Neurol. 1988;45:575-78.
Article PDF
Author and Disclosure Information

Dr. Dhandha is from Maine-Dartmouth Family Medicine Residency Dermatology, Augusta. Dr. Chu is from OncoDerm Associates, St. Louis, Missouri. Dr. Guo is from the Department of Dermatology, Saint Louis University.

The authors report no conflict of interest.

Correspondence: Maulik M. Dhandha MD, MDFMR Dermatology, 6 E Chestnut St, Ballard Center, 3rd Floor, Augusta, ME 04330 ([email protected]).

Issue
Cutis - 103(2)
Publications
Topics
Page Number
E9-E12
Sections
Author and Disclosure Information

Dr. Dhandha is from Maine-Dartmouth Family Medicine Residency Dermatology, Augusta. Dr. Chu is from OncoDerm Associates, St. Louis, Missouri. Dr. Guo is from the Department of Dermatology, Saint Louis University.

The authors report no conflict of interest.

Correspondence: Maulik M. Dhandha MD, MDFMR Dermatology, 6 E Chestnut St, Ballard Center, 3rd Floor, Augusta, ME 04330 ([email protected]).

Author and Disclosure Information

Dr. Dhandha is from Maine-Dartmouth Family Medicine Residency Dermatology, Augusta. Dr. Chu is from OncoDerm Associates, St. Louis, Missouri. Dr. Guo is from the Department of Dermatology, Saint Louis University.

The authors report no conflict of interest.

Correspondence: Maulik M. Dhandha MD, MDFMR Dermatology, 6 E Chestnut St, Ballard Center, 3rd Floor, Augusta, ME 04330 ([email protected]).

Article PDF
Article PDF

To the Editor:

Patients with concurrent neurofibromatosis type 1 (NF-1) and systemic lupus erythematosus (SLE) rarely have been reported in the literature. Neurofibromatosis type 1 is one of the most common genetic disorders, with a worldwide birth incidence of 1 in 2500 individuals and prevalence of 1 in 4000 individuals.1 The incidence and prevalence of SLE varies widely depending on race and geographic location. Estimated incidence rates for SLE range from 1 to 25 per 100,000 individuals annually in North America, South America, Europe, and Asia.2,3 The reported worldwide prevalence is 20 to 150 cases per 100,000 individuals annually.2,4,5

Given the high prevalence of both conditions, the association between SLE and NF-1 likely is underrecognized; therefore, identifying more patients with concurrent SLE and NF-1 and describing the interplay between the 2 conditions may have important therapeutic implications. We present the case of a middle-aged woman with a history of SLE who had cutaneous lesions characteristic of NF-1 to further the understanding of these concurrent conditions.



A middle-aged woman presented to our academic dermatology clinic for evaluation and removal of dark spots that had been present diffusely on the trunk and extremities since birth. She reported a history of SLE with lupus nephritis, hypertension, and a nodular goiter following a partial thyroidectomy. She noted that she did not seek treatment for the skin findings sooner because she was more concerned about her other medical conditions; however, because she felt these conditions were now stable, she decided to seek treatment for the “rash.” Physical examination revealed hundreds of café au lait macules and numerous neurofibromas diffusely distributed on the trunk and extremities (Figure 1) as well as bilateral axillary freckling. A clinical diagnosis of NF-1 was made.

Figure 1. Café au lait macules and neurofibromas on the upper back.


When questioned, the patient reported that she may have been diagnosed with NF-1 in the past by another physician, but she did not recall it specifically. The patient was advised that there were no treatments for the café au lait macules. We notified her other physicians of the NF-1 diagnosis so she could be monitored for systemic conditions related to NF-1, including optic gliomas, pheochromocytoma, renal artery stenosis, and internal neurofibromas. We also referred the patient for genetic counseling; of note, the patient reported she had 4 children without any evidence of similar skin lesions or chronic health problems.

 

 

A PubMed search of articles indexed for MEDLINE using the terms systemic lupus and neurofibromatosis yielded 8 cases of patients having both SLE and NF-1 (including our case).6-11 Our patient reported having multiple lesions since birth, decades before the onset and diagnosis of SLE. In 3 other cases, patients were diagnosed with SLE and then presented with neurofibromas, leading to NF-1 diagnosis.In the discussion of those 3 cases, it was proposed that immune system alterations caused by SLE leading to viral illness may have predisposed the patients to the development of tumors and other collagen diseases, or it could be coincidental.6,7 In another case, a patient with NF-1 developed SLE, which was thought to be coincidental.8 Akyuz et al9 described the case of a pediatric patient with NF-1 who subsequently was diagnosed with SLE. The authors suggested that the lack of neurofibromin contributed to the development of SLE, an autoimmune condition. Under normal circumstances, neurofibromin acts as a guanosine triphosphatase–activating protein for RAS in T cells.10 CD8+ T-cell function also is impaired in patients with SLE.9 Additionally, it has been reported that anti–double-stranded DNA antibodies and immune complexes were present in NF-1 patients, even though there were low titers.12 Thus, the authors proposed that the lack of neurofibromin led to dysregulation of the RAS pathway and impairment of T cells, creating an immune milieu that predisposed the patient to development of SLE. Our case gives additional credence to this theory, as our patient had a similar clinical course: the café au lait macules were present since birth and the symptoms of SLE surfaced much later in her late 20s and 30s. Another case by Makino and Tampo10 described a patient with a history of SLE who was later diagnosed with NF-1 based on choroidal findings highly specific for NF-1 but did not have other classic findings of NF-1. The authors mentioned that there might be a potential relationship between these two disorders but did not speculate any theory in particular for their case.10



The interplay between an autoimmune condition such as SLE and NF-1, a condition traditionally thought to be due to a genetic mutation, may have greater clinical and therapeutic implications beyond just these two disorders. Although it is well established that RAS pathway disruption causes NF-1, it has been uncovered that dysfunction in the RAS pathway also can contribute to melanoma oncogenesis.13,14 These insights have led to the development of and approval of targeted drugs designed to inhibit the RAS pathway (eg, vemurafenib, dabrafenib, trametinib).14-17 Melanoma also is considered a “model” tumor for studying the relationship between the immune system and cancer.18AKT is a signal transduction pathway that promotes cell survival and growth in various cancers.15 In addition, deactivation of MEK (part of the RAS pathway) can cause activation of AKT (protein kinase B) signaling and lupus like autoimmune conditions19 (Figure 2). Likewise, an understanding of the RAS pathway and T-cell function in patients with both SLE and NF-1 may give us more information about melanoma and other cancers.

Figure 2. MAPK pathways and potential interplay between neurofibromatosis type 1, systemic lupus erythematosus, and melanoma. Mutated BRAF leads to activation of the MEK/ERK pathway and development of melanoma. MEK-1 inhibition results in activation of P13K/AKT signaling and breach of peripheral tolerance and development of lupuslike autoimmune disease. Both neurofibromatosis type 1 and systemic lupus erythematosus are related to activated RAS. Neurofibromin 1 (NF1) leads to phosphorylation of RAS-GDP, resulting in the activated form of RAS (RAS-GTP).


Our case also is instructive in another point: our patient had never sought treatment for her skin lesions, as she said she had other more serious health conditions. Closer evaluation of her skin condition may have led to earlier diagnosis of NF-1, which has important health implications. The average lifespan of patients with NF-1 is 10 to 15 years lower than the general population, with cancer being the leading cause of death.20 Malignant peripheral nerve sheath tumors are the most common malignant tumors observed in such patients.21-23 Other cancers that are associated with NF-1 include rhabdomyosarcomas, gastrointestinal stromal tumors, neuroectodermal tumors, pheochromocytomas, and breast carcinomas.23

To make a clinical diagnosis of NF-1, a patient must have 2 of 7 cardinal clinical features as defined by the National Institutes of Health (Table).24 In our patient with hundreds of café au lait macules and dozens of neurofibromas, the diagnosis was clear; however, in other patients, the skin findings of NF-1 may not be as prominent. A patient could meet criteria for NF-1 diagnosis with the inconspicuous presentation of 6 café au lait macules and either 1 plexiform neurofibroma or 2 neurofibromas (of any type) on the entire body.



We recommend that patients with SLE undergo skin examinations to look for more subtle presentations of NF-1. Earlier diagnosis will help to initiate close monitoring of the disorder’s associated systemic health risks. In addition, the identification of more patients with both NF-1 and SLE may help shed light on the etiology of both conditions.

To the Editor:

Patients with concurrent neurofibromatosis type 1 (NF-1) and systemic lupus erythematosus (SLE) rarely have been reported in the literature. Neurofibromatosis type 1 is one of the most common genetic disorders, with a worldwide birth incidence of 1 in 2500 individuals and prevalence of 1 in 4000 individuals.1 The incidence and prevalence of SLE varies widely depending on race and geographic location. Estimated incidence rates for SLE range from 1 to 25 per 100,000 individuals annually in North America, South America, Europe, and Asia.2,3 The reported worldwide prevalence is 20 to 150 cases per 100,000 individuals annually.2,4,5

Given the high prevalence of both conditions, the association between SLE and NF-1 likely is underrecognized; therefore, identifying more patients with concurrent SLE and NF-1 and describing the interplay between the 2 conditions may have important therapeutic implications. We present the case of a middle-aged woman with a history of SLE who had cutaneous lesions characteristic of NF-1 to further the understanding of these concurrent conditions.



A middle-aged woman presented to our academic dermatology clinic for evaluation and removal of dark spots that had been present diffusely on the trunk and extremities since birth. She reported a history of SLE with lupus nephritis, hypertension, and a nodular goiter following a partial thyroidectomy. She noted that she did not seek treatment for the skin findings sooner because she was more concerned about her other medical conditions; however, because she felt these conditions were now stable, she decided to seek treatment for the “rash.” Physical examination revealed hundreds of café au lait macules and numerous neurofibromas diffusely distributed on the trunk and extremities (Figure 1) as well as bilateral axillary freckling. A clinical diagnosis of NF-1 was made.

Figure 1. Café au lait macules and neurofibromas on the upper back.


When questioned, the patient reported that she may have been diagnosed with NF-1 in the past by another physician, but she did not recall it specifically. The patient was advised that there were no treatments for the café au lait macules. We notified her other physicians of the NF-1 diagnosis so she could be monitored for systemic conditions related to NF-1, including optic gliomas, pheochromocytoma, renal artery stenosis, and internal neurofibromas. We also referred the patient for genetic counseling; of note, the patient reported she had 4 children without any evidence of similar skin lesions or chronic health problems.

 

 

A PubMed search of articles indexed for MEDLINE using the terms systemic lupus and neurofibromatosis yielded 8 cases of patients having both SLE and NF-1 (including our case).6-11 Our patient reported having multiple lesions since birth, decades before the onset and diagnosis of SLE. In 3 other cases, patients were diagnosed with SLE and then presented with neurofibromas, leading to NF-1 diagnosis.In the discussion of those 3 cases, it was proposed that immune system alterations caused by SLE leading to viral illness may have predisposed the patients to the development of tumors and other collagen diseases, or it could be coincidental.6,7 In another case, a patient with NF-1 developed SLE, which was thought to be coincidental.8 Akyuz et al9 described the case of a pediatric patient with NF-1 who subsequently was diagnosed with SLE. The authors suggested that the lack of neurofibromin contributed to the development of SLE, an autoimmune condition. Under normal circumstances, neurofibromin acts as a guanosine triphosphatase–activating protein for RAS in T cells.10 CD8+ T-cell function also is impaired in patients with SLE.9 Additionally, it has been reported that anti–double-stranded DNA antibodies and immune complexes were present in NF-1 patients, even though there were low titers.12 Thus, the authors proposed that the lack of neurofibromin led to dysregulation of the RAS pathway and impairment of T cells, creating an immune milieu that predisposed the patient to development of SLE. Our case gives additional credence to this theory, as our patient had a similar clinical course: the café au lait macules were present since birth and the symptoms of SLE surfaced much later in her late 20s and 30s. Another case by Makino and Tampo10 described a patient with a history of SLE who was later diagnosed with NF-1 based on choroidal findings highly specific for NF-1 but did not have other classic findings of NF-1. The authors mentioned that there might be a potential relationship between these two disorders but did not speculate any theory in particular for their case.10



The interplay between an autoimmune condition such as SLE and NF-1, a condition traditionally thought to be due to a genetic mutation, may have greater clinical and therapeutic implications beyond just these two disorders. Although it is well established that RAS pathway disruption causes NF-1, it has been uncovered that dysfunction in the RAS pathway also can contribute to melanoma oncogenesis.13,14 These insights have led to the development of and approval of targeted drugs designed to inhibit the RAS pathway (eg, vemurafenib, dabrafenib, trametinib).14-17 Melanoma also is considered a “model” tumor for studying the relationship between the immune system and cancer.18AKT is a signal transduction pathway that promotes cell survival and growth in various cancers.15 In addition, deactivation of MEK (part of the RAS pathway) can cause activation of AKT (protein kinase B) signaling and lupus like autoimmune conditions19 (Figure 2). Likewise, an understanding of the RAS pathway and T-cell function in patients with both SLE and NF-1 may give us more information about melanoma and other cancers.

Figure 2. MAPK pathways and potential interplay between neurofibromatosis type 1, systemic lupus erythematosus, and melanoma. Mutated BRAF leads to activation of the MEK/ERK pathway and development of melanoma. MEK-1 inhibition results in activation of P13K/AKT signaling and breach of peripheral tolerance and development of lupuslike autoimmune disease. Both neurofibromatosis type 1 and systemic lupus erythematosus are related to activated RAS. Neurofibromin 1 (NF1) leads to phosphorylation of RAS-GDP, resulting in the activated form of RAS (RAS-GTP).


Our case also is instructive in another point: our patient had never sought treatment for her skin lesions, as she said she had other more serious health conditions. Closer evaluation of her skin condition may have led to earlier diagnosis of NF-1, which has important health implications. The average lifespan of patients with NF-1 is 10 to 15 years lower than the general population, with cancer being the leading cause of death.20 Malignant peripheral nerve sheath tumors are the most common malignant tumors observed in such patients.21-23 Other cancers that are associated with NF-1 include rhabdomyosarcomas, gastrointestinal stromal tumors, neuroectodermal tumors, pheochromocytomas, and breast carcinomas.23

To make a clinical diagnosis of NF-1, a patient must have 2 of 7 cardinal clinical features as defined by the National Institutes of Health (Table).24 In our patient with hundreds of café au lait macules and dozens of neurofibromas, the diagnosis was clear; however, in other patients, the skin findings of NF-1 may not be as prominent. A patient could meet criteria for NF-1 diagnosis with the inconspicuous presentation of 6 café au lait macules and either 1 plexiform neurofibroma or 2 neurofibromas (of any type) on the entire body.



We recommend that patients with SLE undergo skin examinations to look for more subtle presentations of NF-1. Earlier diagnosis will help to initiate close monitoring of the disorder’s associated systemic health risks. In addition, the identification of more patients with both NF-1 and SLE may help shed light on the etiology of both conditions.

References
  1. Carey JC, Baty BJ, Johnson JP, et al. The genetic aspects of neurofibromatosis. Ann N Y Acad Sci. 1986;486:45-56.
  2. Pons-Estel GJ, Alarcón GS, Scofield L, et al. Understanding the epidemiology and progression of systemic lupus erythematosus. Semin Arthritis Rheum. 2010;39:257-268.
  3. Danchenko N, Satia JA, Anthony MS. Epidemiology of systemic lupus erythematosus: a comparison of worldwide disease burden. Lupus. 2006;15:308-318.
  4. Lawrence RC, Helmick CG, Arnett FC, et al. Estimates of the prevalence of arthritis and selected musculoskeletal disorders in the United States. Arthritis Rheum. 1998;41:778-799.
  5. Chakravarty EF, Bush TM, Manzi S, et al. Prevalence of adult systemic lupus erythematosus in California and Pennsylvania in 2000: estimates obtained using hospitalization data. Arthritis Rheum. 2007;56:2092-2094.
  6. Bitnun S, Bassan H. Letter: neurofibromatosis and SLE. N Engl J Med. 1975;292:429-430.
  7. Riccardi VM. Neurofibromatosis in a patient with systemic lupus erythematosus. Arthritis Rheum. 1983;26:574.
  8. Corominas H, Guardiola JM, Matas L, et al. Neurofibromatosis and systemic lupus erythematosus. a matter of coincidence? Clin Rhematol. 2003;22:496-497.
  9. Akyuz SG, Caltik A, Bulbul M, et al. An unusual pediatric case with neurofibromatosis and systemic lupus erythematosus. Rheumatol Int. 2012;32:2345-47.
  10. Makino S, Tampo H. Rare and unusual choroidal abnormalities in a patient with systemic lupus erythematosus. Case Rep Ophthalmol. 2013;4:81-86.
  11. Galvan JM, Hofkamp MP. Usefulness of intrapartum magnetic resonance imaging for a parturient with neurofibromatosis type I during induction of labor for preeclampsia. Proc (Bayl Univ Med Cent). 2018;31:92-93.
  12. Gerosa PL, Vai C, Bizzozer L, et al. Immunological and clinical surveillance in Recklinghausen’s neurofibromatosis (NF1). Panminerva Med. 1993;35:80-85.
  13. Busca R, Abbe P, Mantoux F, et al. RAS mediates the cAMP-dependent activation of extracellular signal-regulated kinases (ERKs) in melanocytes. EMBO J. 2000;19:2900-2910.
  14. Sullivan RJ, Flaherty K. MAP kinase signaling and inhibition in melanoma. Oncogene. 2013;32:2373-2379.
  15. Hennessy BT, Smith DL, Ram PT, et al. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005;12:988-1004.
  16. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507-2516.
  17. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380:358-365.
  18. Maio M. Melanoma as a model tumour for immuno-oncology. Ann Oncol. 2012;23:(suppl 8):viii10-4.
  19. Zmajkovicova K, Jesenberger V, Catalanotti F, et al. MEK1 is required for PTEN membrane recruitment, AKT regulation, and the maintenance of peripheral tolerance. Mol Cell. 2013;50:43-55.
  20. Patil S, Chamberlain RS. Neoplasms associated with germline and somatic NF1 gene mutations. Oncologist. 2012;17:101-116.
  21. Carroll SL, Ratner N. How does the Schwann cell lineage form tumors in NF1? Glia. 2008;56:1590-1605.
  22. Rasmussen SA, Friedman JM. NF1 gene and neurofibromatosis 1. Am J Epidemiol. 2000;151:33-40.
  23. Yohay K. Neurofibromatosis type 1 and associated malignancies. Curr Neurol Neurosci Rep. 2009;9:247-253.
  24. Neurofibromatosis. conference statement. National Institutes of Health Consensus Development Conference. Arch Neurol. 1988;45:575-78.
References
  1. Carey JC, Baty BJ, Johnson JP, et al. The genetic aspects of neurofibromatosis. Ann N Y Acad Sci. 1986;486:45-56.
  2. Pons-Estel GJ, Alarcón GS, Scofield L, et al. Understanding the epidemiology and progression of systemic lupus erythematosus. Semin Arthritis Rheum. 2010;39:257-268.
  3. Danchenko N, Satia JA, Anthony MS. Epidemiology of systemic lupus erythematosus: a comparison of worldwide disease burden. Lupus. 2006;15:308-318.
  4. Lawrence RC, Helmick CG, Arnett FC, et al. Estimates of the prevalence of arthritis and selected musculoskeletal disorders in the United States. Arthritis Rheum. 1998;41:778-799.
  5. Chakravarty EF, Bush TM, Manzi S, et al. Prevalence of adult systemic lupus erythematosus in California and Pennsylvania in 2000: estimates obtained using hospitalization data. Arthritis Rheum. 2007;56:2092-2094.
  6. Bitnun S, Bassan H. Letter: neurofibromatosis and SLE. N Engl J Med. 1975;292:429-430.
  7. Riccardi VM. Neurofibromatosis in a patient with systemic lupus erythematosus. Arthritis Rheum. 1983;26:574.
  8. Corominas H, Guardiola JM, Matas L, et al. Neurofibromatosis and systemic lupus erythematosus. a matter of coincidence? Clin Rhematol. 2003;22:496-497.
  9. Akyuz SG, Caltik A, Bulbul M, et al. An unusual pediatric case with neurofibromatosis and systemic lupus erythematosus. Rheumatol Int. 2012;32:2345-47.
  10. Makino S, Tampo H. Rare and unusual choroidal abnormalities in a patient with systemic lupus erythematosus. Case Rep Ophthalmol. 2013;4:81-86.
  11. Galvan JM, Hofkamp MP. Usefulness of intrapartum magnetic resonance imaging for a parturient with neurofibromatosis type I during induction of labor for preeclampsia. Proc (Bayl Univ Med Cent). 2018;31:92-93.
  12. Gerosa PL, Vai C, Bizzozer L, et al. Immunological and clinical surveillance in Recklinghausen’s neurofibromatosis (NF1). Panminerva Med. 1993;35:80-85.
  13. Busca R, Abbe P, Mantoux F, et al. RAS mediates the cAMP-dependent activation of extracellular signal-regulated kinases (ERKs) in melanocytes. EMBO J. 2000;19:2900-2910.
  14. Sullivan RJ, Flaherty K. MAP kinase signaling and inhibition in melanoma. Oncogene. 2013;32:2373-2379.
  15. Hennessy BT, Smith DL, Ram PT, et al. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 2005;12:988-1004.
  16. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507-2516.
  17. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380:358-365.
  18. Maio M. Melanoma as a model tumour for immuno-oncology. Ann Oncol. 2012;23:(suppl 8):viii10-4.
  19. Zmajkovicova K, Jesenberger V, Catalanotti F, et al. MEK1 is required for PTEN membrane recruitment, AKT regulation, and the maintenance of peripheral tolerance. Mol Cell. 2013;50:43-55.
  20. Patil S, Chamberlain RS. Neoplasms associated with germline and somatic NF1 gene mutations. Oncologist. 2012;17:101-116.
  21. Carroll SL, Ratner N. How does the Schwann cell lineage form tumors in NF1? Glia. 2008;56:1590-1605.
  22. Rasmussen SA, Friedman JM. NF1 gene and neurofibromatosis 1. Am J Epidemiol. 2000;151:33-40.
  23. Yohay K. Neurofibromatosis type 1 and associated malignancies. Curr Neurol Neurosci Rep. 2009;9:247-253.
  24. Neurofibromatosis. conference statement. National Institutes of Health Consensus Development Conference. Arch Neurol. 1988;45:575-78.
Issue
Cutis - 103(2)
Issue
Cutis - 103(2)
Page Number
E9-E12
Page Number
E9-E12
Publications
Publications
Topics
Article Type
Display Headline
Neurofibromatosis Type 1 in the Setting of Systemic Lupus Erythematosus
Display Headline
Neurofibromatosis Type 1 in the Setting of Systemic Lupus Erythematosus
Sections
Inside the Article

Practice Points

  • Patients with neurofibromatosis type 1 (NF-1) benefit from early diagnosis and long-term follow-up.
  • Patients with systemic lupus erythematosus (SLE) may develop different malignancies given the immune dysregulation. We recommend that patients with SLE undergo detailed skin examinations to check for subtle clues for NF-1.
  • Similarly, patients with NF-1 can develop SLE later in life.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Miscommunication With Dermatology Patients: Are We Speaking the Same Language?

Article Type
Changed
Fri, 10/25/2019 - 11:00
Display Headline
Miscommunication With Dermatology Patients: Are We Speaking the Same Language?

I was a third-year medical student, dutifully reviewing discharge instructions with a patient and her family. The patient’s adult daughter asked, “What about that diet you put her on?” As they looked at me quizzically, I looked back equally confused, until it clicked: We needed to talk about the word diet. In everyday conversation, diet generally is understood to mean restriction of food to lose weight, which is what the family hoped would be prescribed for their obese family member. I needed to tell them that I was sorry for the misunderstanding. If they overheard us “ordering a diet,” we simply meant providing trays of hospital food.

We become so familiar with the language of our profession that we do not remember it may be foreign to our patients. In dermatology, we are aware that our specialty is full of esoteric jargon and complex concepts that need to be carefully explained to our patients in simpler terms. But since that incident in medical school, I have been interested in the more insidious potential misunderstandings that can arise from words as seemingly simple as diet. There are many examples in dermatology, particularly in the way we prescribe topical therapy and use trade names.

Topical Therapy

Instructions for systemic medications may be as simple as “take 1 pill twice daily.” Prescriptions for topical medications can be written with an equally simple patient signature such as “apply twice daily to affected area,” but the simplicity is deceptive. The direction to “apply” may seem intuitive to the prescriber, but we do not always specify the amount. Sunscreen, for example, is notoriously underapplied when the actual amount of product needed for protection is not demonstrated.1 One study of new dermatology patients given a prescription for a new topical medication found that the majority of patients underdosed.2

Determination of an “affected area,” regardless of whether the site is indicated, can be even less straightforward. In acne treatment, the affected area is the whole region prone to acne breakouts, whereas in psoriasis it may be discrete psoriatic plaques. We may believe our explanations are perfectly clear, but we have all seen patients spot treating their acne or psoriasis patients covering entire territories of normal skin with topical steroids, despite our education. One study of eczema action plans found that there was considerable variability in the way different providers described disease flares that require treatment. For example, redness was only used as a descriptor of an eczema flare in 68.2% of eczema action plans studied.3 Ensuring our patients understand our criteria for skin requiring topical treatment may mean the difference between treatment success and failure and also may help to avoid unnecessary side effects.



Adherence to topical medication regimens is poor, and inadequate patient education is only one factor.4,5 One study found that more than one-third of new prescriptions for topical medications were never even filled.6 However, improving our communication about application of topical drugs is one way we must address the complicated issue of adherence.

 

 

Trade Names

In dermatology, we often use trade names to refer to our medications, even if we do not intend to reference the brand name of the drug specifically. We may tell a patient to use Lidex (Medicis Pharmaceutical Corporation) for her hands but then send an escript to her pharmacy for fluocinonide. Trade names are designed to roll off the tongue, in contrast to the unwieldy, clumsily long generic names assigned to many of our medications.

Substituting trade names may facilitate more natural conversation to promote patient understanding in some cases; however, there are pitfalls associated with this habit. First, we may be doing our patients a disservice if we do not clarify when it would be acceptable to substitute with the generic when the medication is available over-the-counter. If we decide to treat with Rogaine (Johnson & Johnson Consumer Inc) but do not suggest the option of purchasing the generic minoxidil, the patient could be unnecessarily overpaying for a brand name by following our instructions.



Conversely, there are scenarios in which the use of a brand name is actually not specific enough. A patient once told me she was using Differin (Galderma Laboratories, LP) as discussed at her prior visit, but she revealed she was washing it off after application. I initially assumed she misunderstood that adapalene was a gel to be applied and left on. After additional questioning, however, it became clear that she purchased the Differin gentle cleanser, a nonmedicated facial wash, rather than the retinoid we had intended for her. I had not considered that Differin would market an entire line of skin care products but now realize we must be cautious using Differin and adapalene interchangeably. Other examples include popular over-the-counter antihistamine brands such as Allegra (Chattem, a Sanofi company) or Benadryl (Johnson & Johnson Consumer Inc) that market multiple products with different active ingredients.

Final Thoughts

The smooth transfer of information between physician and patient is key to a healthy therapeutic relationship. In residency and throughout our careers, we will continue to develop and refine our communication skills to best serve our patients. We should pay particular attention to the unexpected and surprising ways in which we fail to adequately communicate, make note of these patterns, and share with our colleagues so that we can all learn from our collective experiences.

References
  1. Schneider, J. The teaspoon rule of applying sunscreen. Arch Dermatol. 2002;138:838-839.
  2. Storm A, Benfeldt E, Andersen SE, et al. A prospective study of patient adherence to topical treatments: 95% of patients underdose. J Am Acad Dermatol. 2008;59:975-980.
  3. Stringer T, Yin HS, Gittler J, et al. The readability, suitability, and content features of eczema action plans in the United States. Pediatr Dermatol. 2018;35:800-807.
  4. Hougeir FG, Cook-Bolden FE, Rodriguez D, et al. Critical considerations on optimizing topical corticosteroid therapy. J Clin Aesthet Dermatol. 2015;8(suppl 1):S2-S14.
  5. Savary J, Ortonne JP, Aractingi S. The right dose in the right place: an overview of current prescription, instruction and application modalities for topical psoriasis treatments. J Eur Acad Dermatol Venereol. 2005;19:14-17.
  6. Storm A, Anderson SE, Benfeldt E, et al. One in 3 prescriptions are never redeemed: primary nonadherence in an outpatient clinic. J Am Acad Dermatol. 2008;59:27-33.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Cleveland Clinic Foundation, Ohio.

The author reports no conflict of interest.

Correspondence: Elisabeth Tracey, MD, Department of Dermatology, 9500 Euclid Ave, A60, Cleveland, OH 44195 ([email protected]).

Issue
Cutis - 103(2)
Publications
Topics
Page Number
E27-E28
Sections
Author and Disclosure Information

From the Department of Dermatology, Cleveland Clinic Foundation, Ohio.

The author reports no conflict of interest.

Correspondence: Elisabeth Tracey, MD, Department of Dermatology, 9500 Euclid Ave, A60, Cleveland, OH 44195 ([email protected]).

Author and Disclosure Information

From the Department of Dermatology, Cleveland Clinic Foundation, Ohio.

The author reports no conflict of interest.

Correspondence: Elisabeth Tracey, MD, Department of Dermatology, 9500 Euclid Ave, A60, Cleveland, OH 44195 ([email protected]).

Article PDF
Article PDF

I was a third-year medical student, dutifully reviewing discharge instructions with a patient and her family. The patient’s adult daughter asked, “What about that diet you put her on?” As they looked at me quizzically, I looked back equally confused, until it clicked: We needed to talk about the word diet. In everyday conversation, diet generally is understood to mean restriction of food to lose weight, which is what the family hoped would be prescribed for their obese family member. I needed to tell them that I was sorry for the misunderstanding. If they overheard us “ordering a diet,” we simply meant providing trays of hospital food.

We become so familiar with the language of our profession that we do not remember it may be foreign to our patients. In dermatology, we are aware that our specialty is full of esoteric jargon and complex concepts that need to be carefully explained to our patients in simpler terms. But since that incident in medical school, I have been interested in the more insidious potential misunderstandings that can arise from words as seemingly simple as diet. There are many examples in dermatology, particularly in the way we prescribe topical therapy and use trade names.

Topical Therapy

Instructions for systemic medications may be as simple as “take 1 pill twice daily.” Prescriptions for topical medications can be written with an equally simple patient signature such as “apply twice daily to affected area,” but the simplicity is deceptive. The direction to “apply” may seem intuitive to the prescriber, but we do not always specify the amount. Sunscreen, for example, is notoriously underapplied when the actual amount of product needed for protection is not demonstrated.1 One study of new dermatology patients given a prescription for a new topical medication found that the majority of patients underdosed.2

Determination of an “affected area,” regardless of whether the site is indicated, can be even less straightforward. In acne treatment, the affected area is the whole region prone to acne breakouts, whereas in psoriasis it may be discrete psoriatic plaques. We may believe our explanations are perfectly clear, but we have all seen patients spot treating their acne or psoriasis patients covering entire territories of normal skin with topical steroids, despite our education. One study of eczema action plans found that there was considerable variability in the way different providers described disease flares that require treatment. For example, redness was only used as a descriptor of an eczema flare in 68.2% of eczema action plans studied.3 Ensuring our patients understand our criteria for skin requiring topical treatment may mean the difference between treatment success and failure and also may help to avoid unnecessary side effects.



Adherence to topical medication regimens is poor, and inadequate patient education is only one factor.4,5 One study found that more than one-third of new prescriptions for topical medications were never even filled.6 However, improving our communication about application of topical drugs is one way we must address the complicated issue of adherence.

 

 

Trade Names

In dermatology, we often use trade names to refer to our medications, even if we do not intend to reference the brand name of the drug specifically. We may tell a patient to use Lidex (Medicis Pharmaceutical Corporation) for her hands but then send an escript to her pharmacy for fluocinonide. Trade names are designed to roll off the tongue, in contrast to the unwieldy, clumsily long generic names assigned to many of our medications.

Substituting trade names may facilitate more natural conversation to promote patient understanding in some cases; however, there are pitfalls associated with this habit. First, we may be doing our patients a disservice if we do not clarify when it would be acceptable to substitute with the generic when the medication is available over-the-counter. If we decide to treat with Rogaine (Johnson & Johnson Consumer Inc) but do not suggest the option of purchasing the generic minoxidil, the patient could be unnecessarily overpaying for a brand name by following our instructions.



Conversely, there are scenarios in which the use of a brand name is actually not specific enough. A patient once told me she was using Differin (Galderma Laboratories, LP) as discussed at her prior visit, but she revealed she was washing it off after application. I initially assumed she misunderstood that adapalene was a gel to be applied and left on. After additional questioning, however, it became clear that she purchased the Differin gentle cleanser, a nonmedicated facial wash, rather than the retinoid we had intended for her. I had not considered that Differin would market an entire line of skin care products but now realize we must be cautious using Differin and adapalene interchangeably. Other examples include popular over-the-counter antihistamine brands such as Allegra (Chattem, a Sanofi company) or Benadryl (Johnson & Johnson Consumer Inc) that market multiple products with different active ingredients.

Final Thoughts

The smooth transfer of information between physician and patient is key to a healthy therapeutic relationship. In residency and throughout our careers, we will continue to develop and refine our communication skills to best serve our patients. We should pay particular attention to the unexpected and surprising ways in which we fail to adequately communicate, make note of these patterns, and share with our colleagues so that we can all learn from our collective experiences.

I was a third-year medical student, dutifully reviewing discharge instructions with a patient and her family. The patient’s adult daughter asked, “What about that diet you put her on?” As they looked at me quizzically, I looked back equally confused, until it clicked: We needed to talk about the word diet. In everyday conversation, diet generally is understood to mean restriction of food to lose weight, which is what the family hoped would be prescribed for their obese family member. I needed to tell them that I was sorry for the misunderstanding. If they overheard us “ordering a diet,” we simply meant providing trays of hospital food.

We become so familiar with the language of our profession that we do not remember it may be foreign to our patients. In dermatology, we are aware that our specialty is full of esoteric jargon and complex concepts that need to be carefully explained to our patients in simpler terms. But since that incident in medical school, I have been interested in the more insidious potential misunderstandings that can arise from words as seemingly simple as diet. There are many examples in dermatology, particularly in the way we prescribe topical therapy and use trade names.

Topical Therapy

Instructions for systemic medications may be as simple as “take 1 pill twice daily.” Prescriptions for topical medications can be written with an equally simple patient signature such as “apply twice daily to affected area,” but the simplicity is deceptive. The direction to “apply” may seem intuitive to the prescriber, but we do not always specify the amount. Sunscreen, for example, is notoriously underapplied when the actual amount of product needed for protection is not demonstrated.1 One study of new dermatology patients given a prescription for a new topical medication found that the majority of patients underdosed.2

Determination of an “affected area,” regardless of whether the site is indicated, can be even less straightforward. In acne treatment, the affected area is the whole region prone to acne breakouts, whereas in psoriasis it may be discrete psoriatic plaques. We may believe our explanations are perfectly clear, but we have all seen patients spot treating their acne or psoriasis patients covering entire territories of normal skin with topical steroids, despite our education. One study of eczema action plans found that there was considerable variability in the way different providers described disease flares that require treatment. For example, redness was only used as a descriptor of an eczema flare in 68.2% of eczema action plans studied.3 Ensuring our patients understand our criteria for skin requiring topical treatment may mean the difference between treatment success and failure and also may help to avoid unnecessary side effects.



Adherence to topical medication regimens is poor, and inadequate patient education is only one factor.4,5 One study found that more than one-third of new prescriptions for topical medications were never even filled.6 However, improving our communication about application of topical drugs is one way we must address the complicated issue of adherence.

 

 

Trade Names

In dermatology, we often use trade names to refer to our medications, even if we do not intend to reference the brand name of the drug specifically. We may tell a patient to use Lidex (Medicis Pharmaceutical Corporation) for her hands but then send an escript to her pharmacy for fluocinonide. Trade names are designed to roll off the tongue, in contrast to the unwieldy, clumsily long generic names assigned to many of our medications.

Substituting trade names may facilitate more natural conversation to promote patient understanding in some cases; however, there are pitfalls associated with this habit. First, we may be doing our patients a disservice if we do not clarify when it would be acceptable to substitute with the generic when the medication is available over-the-counter. If we decide to treat with Rogaine (Johnson & Johnson Consumer Inc) but do not suggest the option of purchasing the generic minoxidil, the patient could be unnecessarily overpaying for a brand name by following our instructions.



Conversely, there are scenarios in which the use of a brand name is actually not specific enough. A patient once told me she was using Differin (Galderma Laboratories, LP) as discussed at her prior visit, but she revealed she was washing it off after application. I initially assumed she misunderstood that adapalene was a gel to be applied and left on. After additional questioning, however, it became clear that she purchased the Differin gentle cleanser, a nonmedicated facial wash, rather than the retinoid we had intended for her. I had not considered that Differin would market an entire line of skin care products but now realize we must be cautious using Differin and adapalene interchangeably. Other examples include popular over-the-counter antihistamine brands such as Allegra (Chattem, a Sanofi company) or Benadryl (Johnson & Johnson Consumer Inc) that market multiple products with different active ingredients.

Final Thoughts

The smooth transfer of information between physician and patient is key to a healthy therapeutic relationship. In residency and throughout our careers, we will continue to develop and refine our communication skills to best serve our patients. We should pay particular attention to the unexpected and surprising ways in which we fail to adequately communicate, make note of these patterns, and share with our colleagues so that we can all learn from our collective experiences.

References
  1. Schneider, J. The teaspoon rule of applying sunscreen. Arch Dermatol. 2002;138:838-839.
  2. Storm A, Benfeldt E, Andersen SE, et al. A prospective study of patient adherence to topical treatments: 95% of patients underdose. J Am Acad Dermatol. 2008;59:975-980.
  3. Stringer T, Yin HS, Gittler J, et al. The readability, suitability, and content features of eczema action plans in the United States. Pediatr Dermatol. 2018;35:800-807.
  4. Hougeir FG, Cook-Bolden FE, Rodriguez D, et al. Critical considerations on optimizing topical corticosteroid therapy. J Clin Aesthet Dermatol. 2015;8(suppl 1):S2-S14.
  5. Savary J, Ortonne JP, Aractingi S. The right dose in the right place: an overview of current prescription, instruction and application modalities for topical psoriasis treatments. J Eur Acad Dermatol Venereol. 2005;19:14-17.
  6. Storm A, Anderson SE, Benfeldt E, et al. One in 3 prescriptions are never redeemed: primary nonadherence in an outpatient clinic. J Am Acad Dermatol. 2008;59:27-33.
References
  1. Schneider, J. The teaspoon rule of applying sunscreen. Arch Dermatol. 2002;138:838-839.
  2. Storm A, Benfeldt E, Andersen SE, et al. A prospective study of patient adherence to topical treatments: 95% of patients underdose. J Am Acad Dermatol. 2008;59:975-980.
  3. Stringer T, Yin HS, Gittler J, et al. The readability, suitability, and content features of eczema action plans in the United States. Pediatr Dermatol. 2018;35:800-807.
  4. Hougeir FG, Cook-Bolden FE, Rodriguez D, et al. Critical considerations on optimizing topical corticosteroid therapy. J Clin Aesthet Dermatol. 2015;8(suppl 1):S2-S14.
  5. Savary J, Ortonne JP, Aractingi S. The right dose in the right place: an overview of current prescription, instruction and application modalities for topical psoriasis treatments. J Eur Acad Dermatol Venereol. 2005;19:14-17.
  6. Storm A, Anderson SE, Benfeldt E, et al. One in 3 prescriptions are never redeemed: primary nonadherence in an outpatient clinic. J Am Acad Dermatol. 2008;59:27-33.
Issue
Cutis - 103(2)
Issue
Cutis - 103(2)
Page Number
E27-E28
Page Number
E27-E28
Publications
Publications
Topics
Article Type
Display Headline
Miscommunication With Dermatology Patients: Are We Speaking the Same Language?
Display Headline
Miscommunication With Dermatology Patients: Are We Speaking the Same Language?
Sections
Inside the Article

Resident Pearl

  • It is not just the esoteric jargon and complex pathophysiologic concepts in dermatology that can challenge effective communication with our patients. We face potential for misunderstanding even in situations that may seem straightforward. Vigilance in avoiding ambiguity in all our exchanges with patients can help foster therapeutic relationships and optimize patient care.
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.
Article PDF Media

February 2019 Highlights

Article Type
Changed
Mon, 02/11/2019 - 11:32
Display Headline
February 2019 Highlights
Publications
Publications
Publications
Article Type
Display Headline
February 2019 Highlights
Display Headline
February 2019 Highlights
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 02/07/2019 - 10:45
Un-Gate On Date
Thu, 02/07/2019 - 10:45
Use ProPublica
CFC Schedule Remove Status
Thu, 02/07/2019 - 10:45

Nail Psoriasis Tips

Article Type
Changed
Mon, 02/11/2019 - 11:13
Display Headline
Nail Psoriasis Tips

What does your patient need to know at the first visit?

Patient education is important initially. There are several causes for nail dystrophy. Oftentimes, when patients present, they believe that they have onychomycosis. Therefore, it is important to counsel individuals with potential nail psoriasis (Figure) and to discuss the differential diagnosis of the condition.

Nail matrix psoriasis demonstrating pitting and onycholysis. Photograph courtesy of Antonella Tosti, MD (Miami, Florida). Reprinted with permission from Cutis. 2013;92:129-135.

The presence of psoriasis on other areas of the body and the absence of fungal infection on the soles of the feet and in between the toes increases the likelihood of nail psoriasis. The most accurate test to perform is a nail clipping with subsequent periodic acid–Schiff stain. It is important to remember, however, that nail psoriasis and fungal infection of the nail can coexist.

Once the diagnosis of nail psoriasis is established, it is important to review gentle care of the nails. A thorough discussion of therapeutic options is helpful. Patients also should be advised that the presence of nail psoriasis can increase the likelihood of the development of
psoriatic arthritis.

What are your go-to treatments?

Prior to the development of biologic therapies, topical treatments were the mainstay of treatment. Topical corticosteroid preparations can be used around and under the nail. Other therapeutic options include topical calcipotriene and topical retinoids.

Intralesional injection is another therapeutic option. Injection into the nail bed is useful for the treatment of nail bed symptoms of nail psoriasis such as onycholysis. Injection into the proximal nail fold can ameliorate signs of nail matrix psoriasis such as nail pitting. Although injection can be effective, it also can be painful; therefore, many patients do not opt to have this therapy performed.

Systemic therapy has been shown to be highly effective in improving nail psoriasis. There has been a good amount of data from studies specifically done in nail psoriasis and nail data that have been taken from larger phase 3 trials (Elewski et al; van de Kerkhof et al). Therefore, several of the biologics on the market as well apremilast are good options for the treatment of nail psoriasis. When using a systemic agent, it is important to carefully review the benefits and risks of each therapy with patients. Because the nail grows slowly, improvement can be gradual and take several months to peak.

How do you keep patients compliant with treatment?

Because nail psoriasis causes distress among patients, it generally is not too hard for them to be compliant. Of course, it is important to have regular follow-up to monitor progress and to reinforce the importance of continued therapy. At the end of the day, however, treatment success is the best asset to encourage continued compliance.

Resources for Patients
Managing nail psoriasis
http://www.psoriasis.org/about-psoriasis/specific-locations/hands-feet-nails/managing-nail-psoriasis

What is nail psoriasis, and how can I treat it?
http://www.aad.org/public/diseases/scaly-skin/psoriasis/diagnosis-and-treatment-of-psoriasis/what-is-nail-psoriasis-and-how-can-i-treat-it

Suggested Readings
Elewski BE, Okun MM, Papp K, et al. Adalimumab for nail psoriasis: efficacy and safety from the first 26 weeks of phase 3, randomized, placebo controlled trial. J Am Acad Dermatol. 2018;78:90.e1-99.e1.

Van de Kerkhof P, Guenther L, Gottlieb AB, et al. Ixekizumab treatment improves fingernail psoriasis in patients with moderate-to-severe psoriasis: results from the randomized, controlled, and open-label phases of UNCOVER-3. J Eur Acad Dermatol Venereol. 2017;31:477-482.

Yin N, Choudhary S, Nouri K. Pulsed dye laser for the treatment of nail psoriasis. Cutis. 2013;92:129-135.

Article PDF
Author and Disclosure Information

From the Icahn School of Medicine at Mount Sinai, New York, New York.

Dr. Weinberg is on the speaker’s bureau for AbbVie; Amgen Inc; Eli Lilly and Company; Novartis; and Sun Pharmaceutical Industries, Ltd.

Correspondence: Jeffrey M. Weinberg, MD, 10 Union Square E, Ste 3C, New York, NY 10003 ([email protected]).

Issue
Cutis - 103(2)
Publications
Topics
Page Number
109-110
Sections
Author and Disclosure Information

From the Icahn School of Medicine at Mount Sinai, New York, New York.

Dr. Weinberg is on the speaker’s bureau for AbbVie; Amgen Inc; Eli Lilly and Company; Novartis; and Sun Pharmaceutical Industries, Ltd.

Correspondence: Jeffrey M. Weinberg, MD, 10 Union Square E, Ste 3C, New York, NY 10003 ([email protected]).

Author and Disclosure Information

From the Icahn School of Medicine at Mount Sinai, New York, New York.

Dr. Weinberg is on the speaker’s bureau for AbbVie; Amgen Inc; Eli Lilly and Company; Novartis; and Sun Pharmaceutical Industries, Ltd.

Correspondence: Jeffrey M. Weinberg, MD, 10 Union Square E, Ste 3C, New York, NY 10003 ([email protected]).

Article PDF
Article PDF
Related Articles

What does your patient need to know at the first visit?

Patient education is important initially. There are several causes for nail dystrophy. Oftentimes, when patients present, they believe that they have onychomycosis. Therefore, it is important to counsel individuals with potential nail psoriasis (Figure) and to discuss the differential diagnosis of the condition.

Nail matrix psoriasis demonstrating pitting and onycholysis. Photograph courtesy of Antonella Tosti, MD (Miami, Florida). Reprinted with permission from Cutis. 2013;92:129-135.

The presence of psoriasis on other areas of the body and the absence of fungal infection on the soles of the feet and in between the toes increases the likelihood of nail psoriasis. The most accurate test to perform is a nail clipping with subsequent periodic acid–Schiff stain. It is important to remember, however, that nail psoriasis and fungal infection of the nail can coexist.

Once the diagnosis of nail psoriasis is established, it is important to review gentle care of the nails. A thorough discussion of therapeutic options is helpful. Patients also should be advised that the presence of nail psoriasis can increase the likelihood of the development of
psoriatic arthritis.

What are your go-to treatments?

Prior to the development of biologic therapies, topical treatments were the mainstay of treatment. Topical corticosteroid preparations can be used around and under the nail. Other therapeutic options include topical calcipotriene and topical retinoids.

Intralesional injection is another therapeutic option. Injection into the nail bed is useful for the treatment of nail bed symptoms of nail psoriasis such as onycholysis. Injection into the proximal nail fold can ameliorate signs of nail matrix psoriasis such as nail pitting. Although injection can be effective, it also can be painful; therefore, many patients do not opt to have this therapy performed.

Systemic therapy has been shown to be highly effective in improving nail psoriasis. There has been a good amount of data from studies specifically done in nail psoriasis and nail data that have been taken from larger phase 3 trials (Elewski et al; van de Kerkhof et al). Therefore, several of the biologics on the market as well apremilast are good options for the treatment of nail psoriasis. When using a systemic agent, it is important to carefully review the benefits and risks of each therapy with patients. Because the nail grows slowly, improvement can be gradual and take several months to peak.

How do you keep patients compliant with treatment?

Because nail psoriasis causes distress among patients, it generally is not too hard for them to be compliant. Of course, it is important to have regular follow-up to monitor progress and to reinforce the importance of continued therapy. At the end of the day, however, treatment success is the best asset to encourage continued compliance.

Resources for Patients
Managing nail psoriasis
http://www.psoriasis.org/about-psoriasis/specific-locations/hands-feet-nails/managing-nail-psoriasis

What is nail psoriasis, and how can I treat it?
http://www.aad.org/public/diseases/scaly-skin/psoriasis/diagnosis-and-treatment-of-psoriasis/what-is-nail-psoriasis-and-how-can-i-treat-it

Suggested Readings
Elewski BE, Okun MM, Papp K, et al. Adalimumab for nail psoriasis: efficacy and safety from the first 26 weeks of phase 3, randomized, placebo controlled trial. J Am Acad Dermatol. 2018;78:90.e1-99.e1.

Van de Kerkhof P, Guenther L, Gottlieb AB, et al. Ixekizumab treatment improves fingernail psoriasis in patients with moderate-to-severe psoriasis: results from the randomized, controlled, and open-label phases of UNCOVER-3. J Eur Acad Dermatol Venereol. 2017;31:477-482.

Yin N, Choudhary S, Nouri K. Pulsed dye laser for the treatment of nail psoriasis. Cutis. 2013;92:129-135.

What does your patient need to know at the first visit?

Patient education is important initially. There are several causes for nail dystrophy. Oftentimes, when patients present, they believe that they have onychomycosis. Therefore, it is important to counsel individuals with potential nail psoriasis (Figure) and to discuss the differential diagnosis of the condition.

Nail matrix psoriasis demonstrating pitting and onycholysis. Photograph courtesy of Antonella Tosti, MD (Miami, Florida). Reprinted with permission from Cutis. 2013;92:129-135.

The presence of psoriasis on other areas of the body and the absence of fungal infection on the soles of the feet and in between the toes increases the likelihood of nail psoriasis. The most accurate test to perform is a nail clipping with subsequent periodic acid–Schiff stain. It is important to remember, however, that nail psoriasis and fungal infection of the nail can coexist.

Once the diagnosis of nail psoriasis is established, it is important to review gentle care of the nails. A thorough discussion of therapeutic options is helpful. Patients also should be advised that the presence of nail psoriasis can increase the likelihood of the development of
psoriatic arthritis.

What are your go-to treatments?

Prior to the development of biologic therapies, topical treatments were the mainstay of treatment. Topical corticosteroid preparations can be used around and under the nail. Other therapeutic options include topical calcipotriene and topical retinoids.

Intralesional injection is another therapeutic option. Injection into the nail bed is useful for the treatment of nail bed symptoms of nail psoriasis such as onycholysis. Injection into the proximal nail fold can ameliorate signs of nail matrix psoriasis such as nail pitting. Although injection can be effective, it also can be painful; therefore, many patients do not opt to have this therapy performed.

Systemic therapy has been shown to be highly effective in improving nail psoriasis. There has been a good amount of data from studies specifically done in nail psoriasis and nail data that have been taken from larger phase 3 trials (Elewski et al; van de Kerkhof et al). Therefore, several of the biologics on the market as well apremilast are good options for the treatment of nail psoriasis. When using a systemic agent, it is important to carefully review the benefits and risks of each therapy with patients. Because the nail grows slowly, improvement can be gradual and take several months to peak.

How do you keep patients compliant with treatment?

Because nail psoriasis causes distress among patients, it generally is not too hard for them to be compliant. Of course, it is important to have regular follow-up to monitor progress and to reinforce the importance of continued therapy. At the end of the day, however, treatment success is the best asset to encourage continued compliance.

Resources for Patients
Managing nail psoriasis
http://www.psoriasis.org/about-psoriasis/specific-locations/hands-feet-nails/managing-nail-psoriasis

What is nail psoriasis, and how can I treat it?
http://www.aad.org/public/diseases/scaly-skin/psoriasis/diagnosis-and-treatment-of-psoriasis/what-is-nail-psoriasis-and-how-can-i-treat-it

Suggested Readings
Elewski BE, Okun MM, Papp K, et al. Adalimumab for nail psoriasis: efficacy and safety from the first 26 weeks of phase 3, randomized, placebo controlled trial. J Am Acad Dermatol. 2018;78:90.e1-99.e1.

Van de Kerkhof P, Guenther L, Gottlieb AB, et al. Ixekizumab treatment improves fingernail psoriasis in patients with moderate-to-severe psoriasis: results from the randomized, controlled, and open-label phases of UNCOVER-3. J Eur Acad Dermatol Venereol. 2017;31:477-482.

Yin N, Choudhary S, Nouri K. Pulsed dye laser for the treatment of nail psoriasis. Cutis. 2013;92:129-135.

Issue
Cutis - 103(2)
Issue
Cutis - 103(2)
Page Number
109-110
Page Number
109-110
Publications
Publications
Topics
Article Type
Display Headline
Nail Psoriasis Tips
Display Headline
Nail Psoriasis Tips
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
CFC Schedule Remove Status
Thu, 08/02/2018 - 09:15
Article PDF Media

Granuloma Annulare: A Retrospective Series of 133 Patients

Article Type
Changed
Mon, 02/11/2019 - 11:15
Display Headline
Granuloma Annulare: A Retrospective Series of 133 Patients

Granuloma annulare (GA) is a granulomatous skin disorder of uncertain etiology. A number of clinical variants exist, most commonly localized annular plaques on the hands or feet, generalized lesions, or subcutaneous nodules in children. Histologically, GA exhibits granulomatous inflammation with either interstitial or palisading lymphocytes and histiocytes with mucin deposition.

Few data exist regarding the epidemiology of GA. Although the pathogenesis of GA is unknown, associations between GA and underlying systemic processes, such as diabetes mellitus, hyperlipidemia, thyroid disease, and human immunodeficiency virus (HIV), have been suggested.



The purpose of this retrospective study was to determine the number of cases of GA seen annually at the Department of Dermatology at the University of Pennsylvania (Philadelphia, Pennsylvania) from 2008 to 2014. Additionally, we reviewed all cases of biopsy-proven GA from 2010 to 2014 and reported the demographics, underlying medical comorbidities, medications, treatments, and outcomes seen in this patient population.

Methods

We identified the number of outpatients presenting with GA annually using PennSeek, a tool developed by the Penn Medicine Data Analytics Center to search electronic medical records (EMRs). We queried the EMR database to determine the number of discrete patients seen at the Department of Dermatology at the University of Pennsylvania annually from 2008 (the year the EMR was established) to 2014. We then used PennSeek to determine the number of patients given a diagnosis of GA annually from 2008 to 2014 based on the International Classification of Diseases, Ninth Revision (ICD-9).

After using PennSeek to identify all patients given the ICD-9 diagnosis of GA from 2008 to 2014, we reviewed the EMRs of these patients to identify cases that were biopsy proven. For the biopsy-proven cases of GA seen at the University of Pennsylvania from 2010 to 2014, we reviewed the EMRs of these patients for clinical characteristics and treatment outcomes. For each case, we recorded the patient’s age, sex, medical comorbidities, GA subtype, and medications.

This study was approved by the University of Pennsylvania’s institutional review board.

 

 

Results

On average, the percentage of patients given a diagnosis of GA annually was 0.22% (95% CI, 0.19%-0.24%). A Pearson χ2 test was used to determine if any single annual percentage was significantly different from the others. We found a P value of .321, which suggests that the percentage of patients with GA seen annually has been stable from 2008 to 2014 (Figure).

Proportion of patients diagnosed with granuloma annulare (GA) annually (2008-2014).

There were 133 cases of biopsy-proven GA that were reviewed for clinical characteristics; of them, 86.5% were female. Thyroid disease was noted in 30.1% of patients, hyperlipidemia in 30.1%, and hematologic malignancies in 3.8%. Type 1 diabetes mellitus was noted in 1.5% of patients. None of the patients were HIV-positive, 1.5% were hepatitis B–positive, and 2.3% were hepatitis C–positive. Of the 133 cases, 64.7% had localized GA and 30.8% had generalized GA. Photosensitive and papular GA were rarer (1.5% and 2.3% of cases, respectively). Use of a selective serotonin reuptake inhibitor (SSRI) was noted in 18.1% of patients; use of a calcium channel blocker was noted in 9.0% (Table 1).



The most commonly prescribed treatment of GA was topical steroids; 30.9% of patients who were prescribed a topical steroid experienced improvement of their condition. Intralesional triamcinolone was the second most prescribed treatment of GA, with an improvement rate of 40.0% (Table 2).

Comment

We attempted to determine the period of prevalence of GA in a tertiary care, university-based referral practice and evaluate disease associations, treatments, and outcomes of patients with biopsy-proven GA. Our calculated period prevalence of GA of 0.22% to 0.27% is consistent with another review, which reported that 0.1% to 0.4% of new patients presenting to a dermatology practice were given a diagnosis of GA.1 More than 85% of the cases we reviewed were seen in females, a finding that is more heavily skewed compared to prior reports that have suggested a female to male ratio of approximately 1:1 to 2:1.1-7 Our findings suggest that GA is a female-predominant condition, or women may be more likely to seek evaluation for the condition.

More than 95% of the cases we reviewed were localized (64.7%) or generalized (30.8%) GA, making these variants the most common forms of GA, which is consistent with prior reports.1-3,8,9 Other varieties of GA—drug induced, patch, perforating, photosensitive, palmar, and papular—appear rare. Because this study was conducted at an adult hospital, subcutaneous GA, which often is seen in children, may be underrepresented. As a retrospective chart review, it is possible that documentation is insufficient to capture each rare variant.

 

 


Concomitant Disorders and Unrelated Medical Therapy
Hypothyroidism is statistically significantly overrepresented in our patient population (30.1%) compared with an average prevalence of 1% to 2% in iodine-replete populations (Fisher exact test, P<.001).10 This finding is consistent with prior small studies and cases series, which have suggested an association between autoimmune thyroiditis and GA.11-14

Despite prior reports of a possible association between HIV and GA,15-24 none of our patients had a diagnosis of HIV. However, many of our patients were not tested for HIV, which confounds our results and may represent a practice gap in the field.

At 1.5%, the prevalence of type 1 diabetes mellitus in our patients is slightly higher than the national average of 0.3%.25 However, based on a Fisher exact test of analysis of proportions, this difference is not statistically significant (P=.106).

At 1.5% and 2.3%, the prevalence of hepatitis B and hepatitis C, respectively, in our patients is slightly higher than the national average of 0.5% and 1%, respectively.26 However, based on a Fisher exact test of analysis of proportions, these differences are not statistically significant (P=.142 and P=.146, respectively).

Given the high prevalence of hyperlipidemia in the United States (31.7%), this disease is not overrepresented in our sample (30.1%), though others have suggested there may be a connection.27,28 Based on a Fisher exact test, this difference of proportions is not statistically significant (P=.780).

Selective serotonin reuptake inhibitor use is common in the United States; approximately 11% of Americans older than 12 years use an SSRI.29 At 18.1%, the use of SSRIs in our patient group was statistically significantly higher than the national average (Fisher exact test, P=.017), suggesting a possible association between SSRI use and development of GA, warranting further investigation.

The use of calcium channel blockers, interferon, and tumor necrosis factor inhibitors was not significantly associated with GA in our series.

GA Therapy
The most commonly used treatments for GA in our study were topical steroids and intralesional triamcinolone, followed by hydroxychloroquine; all treatments employed exhibited a widely variable response. Assessing treatment response via retrospective chart review is challenging and response rates may not be accurately captured.

Study Limitations
Our study had several limitations. In calculating the period prevalence of GA, our query was limited by the number of years that the EMR has been in place. The number of cases we reviewed for clinical characteristics was limited to 133, as many cases with the ICD-9 diagnosis of GA were not biopsy proven and therefore were not included in our review. Many of the cases we reviewed were lost to follow-up, which prevented us from determining treatment outcomes.



Another weakness of our study was that our query did not provide an estimate of incidence or prevalence of GA overall, as this analysis was not a population-based study. The power of our study was limited by the number of cases of GA seen annually and the number of patients lost to follow-up. Additionally, our study population may only be generalizable to other large academic centers.

Conclusion

This study further solidifies our understanding of the epidemiology of GA and diseases that can be associated with GA. We identified a higher female to male ratio than previous reports, and consistent with prior reports, we noted potential associations with conditions such as thyroid disease and hyperlipidemia. Our population demonstrated higher rates of SSRI use than expected, warranting further investigation. Dermatologists should be aware of potential disease associations with GA, but as a whole we need better data and larger studies to determine the appropriate evaluation and treatment for patients with GA.

References
  1. Muhlbauer JE. Granuloma annulare. J Am Acad Dermatol. 1980;3:217-230.
  2. Thornsberry LA, English JC 3rd. Etiology, diagnosis, and therapeutic management of granuloma annulare: an update. Am J Clin Dermatol. 2013;14:279-290.
  3. Wells RS, Smith MA. The natural history of granuloma annulare. Br J Dermatol. 1963;75:199-205.
  4. Wallet-Faber N, Farhi D, Gorin I, et al. Outcome of granuloma annulare: shorter duration is associated with younger age and recent onset. J Eur Acad Dermatol Venereol. 2010;24:103-104.
  5. Dahl MV. Granuloma annulare: long-term follow-up. Arch Dermatol. 2007;143:946-947.
  6. Yun JH, Lee JY, Kim MK, et al. Clinical and pathological features of generalized granuloma annulare with their correlation: a retrospective multicenter study in Korea. Ann Dermatol. 2009;21:113-119.
  7. Tan HH, Goh CL. Granuloma annulare: a review of 41 cases at the National Skin Centre. Ann Acad Med Singapore. 2000;29:714-718.
  8. Cyr PR. Diagnosis and management of granuloma annulare. Am Fam Physician. 2006;74:1729-1734.
  9. Smith MD, Downie JB, DiCostanzo D. Granuloma annulare. Int J Dermatol. 1997;36:326-333.
  10. Vanderpump MPJ. The epidemiology of thyroid diseases. In: Braverman LE, Utiger RD, eds. Werner and Ingbar’s The Thyroid: A Fundamental and Clinical Text. 9th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2005:398-496.
  11. Vázquez-López F, Pereiro M Jr, Manjón Haces JA, et al. Localized granuloma annulare and autoimmune thyroiditis in adult women: a case-control study. J Am Acad Dermatol. 2003;48:517-520.
  12. Vázquez-López F, González-López MA, Raya-Aguado C, et al. Localized granuloma annulare and autoimmune thyroiditis: a new case report. J Am Acad Dermatol. 2000;43(5, pt 2):943-945.
  13. Kappeler D, Troendle A, Mueller B. Localized granuloma annulare associated with autoimmune thyroid disease in a patient with a positive family history for autoimmune polyglandular syndrome type II. Eur J Endocrinol. 2001;145:101-102.
  14. Maschio M, Marigliano M, Sabbion A, et al. A rare case of granuloma annulare in a 5-year-old child with type 1 diabetes and autoimmune thyroiditis. Am J Dermatopathol. 2013;35:385-387.
  15. Smith NP. AIDS, Kaposi’s sarcoma and the dermatologist. J R Soc Med. 1985;78:97-99.
  16. Huerter CJ, Bass J, Bergfeld WF, et al. Perforating granuloma annulare in a patient with acquired immunodeficiency syndrome. Immunohistologic evaluation of the cellular infiltrate. Arch Dermatol. 1987;123:1217-1220.
  17. Jones SK, Harman RR. Atypical granuloma annulare in patients with the acquired immunodeficiency syndrome. J Am Acad Dermatol. 1989;20(2 pt 1):299-300.
  18. Devesa Parente JA, Dores JA, Aranha JM. Generalized perforating granuloma annulare: case report. Acta Dermatovenerol Croat. 2012;20:260-262.
  19. Ghadially R, Sibbald RG, Walter JB, et al. Granuloma annulare in patients with human immunodeficiency virus infections. J Am Acad Dermatol. 1989;20(2, pt 1):232-235.
  20. Toro JR, Chu P, Yen TS, et al. Granuloma annulare and human immunodeficiency virus infection. Arch Dermatol. 1999;135:1341-1346.
  21. Cohen PR. Granuloma annulare: a mucocutaneous condition in human immunodeficiency virus-infected patients. Arch Dermatol. 1999;135:1404-1407.
  22. O’Moore EJ, Nandawni R, Uthayakumar S, et al. HIV-associated granuloma annulare (HAGA): a report of six cases. Br J Dermatol. 2000;142:1054-1056.
  23. Kapembwa MS, Goolamali SK, Price A, et al. Granuloma annulare masquerading as molluscum contagiosum-like eruption in an HIV-positive African woman. J Am Acad Dermatol. 2003;49(suppl 2):S184-S186.
  24. Morris SD, Cerio R, Paige DG. An unusual presentation of diffuse granuloma annulare in an HIV-positive patient—immunohistochemical evidence of predominant CD8 lymphocytes. Clin Exp Dermatol. 2002;27:205-208.
  25. Maahs DM, West NA, Lawrence JM, et al. Epidemiology of type 1 diabetes. Endocrinol Metab Clin North Am. 2010;39:481-497.
  26. Centers for Disease Control and Prevention. Viral hepatitis surveillance—United States, 2010. www.cdc.gov/hepatitis/statistics/2010surveillance/commentary.htm. Accessed November 10, 2018.
  27. Mozaffarian D, Benjamin EJ, Go AS, et al; American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2015 update: a report from the American Heart Association. Circulation. 2015;131:E29-E322.
  28. Wu W, Robinson-Bostom L, Kokkotou E, et al. Dyslipidemia in granuloma annulare: a case-control study. Arch Dermatol. 2012;148:1131-1136.
  29. Pratt LA, Brody DJ, Gu Q. Antidepressant Use in Persons Aged 12 and Over: United States, 2005-2008. NCHS Data Brief, No. 76. Hyattsville, MD: National Center for Health Statistics; 2011. http://www.cdc.gov/nchs/data/databriefs/db76.htm. Updated October 19, 2011. Accessed June 1, 2014.
Author and Disclosure Information

From the Department of Dermatology, University of Pennsylvania, Philadelphia.

The authors report no conflict of interest.

Correspondence: Misha Rosenbach, MD, Department of Dermatology, University of Pennsylvania, 3600 Spruce St, 2 Maloney Bldg, Philadelphia, PA 19104 ([email protected]).

Issue
Cutis - 103(2)
Publications
Topics
Page Number
102-106
Sections
Author and Disclosure Information

From the Department of Dermatology, University of Pennsylvania, Philadelphia.

The authors report no conflict of interest.

Correspondence: Misha Rosenbach, MD, Department of Dermatology, University of Pennsylvania, 3600 Spruce St, 2 Maloney Bldg, Philadelphia, PA 19104 ([email protected]).

Author and Disclosure Information

From the Department of Dermatology, University of Pennsylvania, Philadelphia.

The authors report no conflict of interest.

Correspondence: Misha Rosenbach, MD, Department of Dermatology, University of Pennsylvania, 3600 Spruce St, 2 Maloney Bldg, Philadelphia, PA 19104 ([email protected]).

Granuloma annulare (GA) is a granulomatous skin disorder of uncertain etiology. A number of clinical variants exist, most commonly localized annular plaques on the hands or feet, generalized lesions, or subcutaneous nodules in children. Histologically, GA exhibits granulomatous inflammation with either interstitial or palisading lymphocytes and histiocytes with mucin deposition.

Few data exist regarding the epidemiology of GA. Although the pathogenesis of GA is unknown, associations between GA and underlying systemic processes, such as diabetes mellitus, hyperlipidemia, thyroid disease, and human immunodeficiency virus (HIV), have been suggested.



The purpose of this retrospective study was to determine the number of cases of GA seen annually at the Department of Dermatology at the University of Pennsylvania (Philadelphia, Pennsylvania) from 2008 to 2014. Additionally, we reviewed all cases of biopsy-proven GA from 2010 to 2014 and reported the demographics, underlying medical comorbidities, medications, treatments, and outcomes seen in this patient population.

Methods

We identified the number of outpatients presenting with GA annually using PennSeek, a tool developed by the Penn Medicine Data Analytics Center to search electronic medical records (EMRs). We queried the EMR database to determine the number of discrete patients seen at the Department of Dermatology at the University of Pennsylvania annually from 2008 (the year the EMR was established) to 2014. We then used PennSeek to determine the number of patients given a diagnosis of GA annually from 2008 to 2014 based on the International Classification of Diseases, Ninth Revision (ICD-9).

After using PennSeek to identify all patients given the ICD-9 diagnosis of GA from 2008 to 2014, we reviewed the EMRs of these patients to identify cases that were biopsy proven. For the biopsy-proven cases of GA seen at the University of Pennsylvania from 2010 to 2014, we reviewed the EMRs of these patients for clinical characteristics and treatment outcomes. For each case, we recorded the patient’s age, sex, medical comorbidities, GA subtype, and medications.

This study was approved by the University of Pennsylvania’s institutional review board.

 

 

Results

On average, the percentage of patients given a diagnosis of GA annually was 0.22% (95% CI, 0.19%-0.24%). A Pearson χ2 test was used to determine if any single annual percentage was significantly different from the others. We found a P value of .321, which suggests that the percentage of patients with GA seen annually has been stable from 2008 to 2014 (Figure).

Proportion of patients diagnosed with granuloma annulare (GA) annually (2008-2014).

There were 133 cases of biopsy-proven GA that were reviewed for clinical characteristics; of them, 86.5% were female. Thyroid disease was noted in 30.1% of patients, hyperlipidemia in 30.1%, and hematologic malignancies in 3.8%. Type 1 diabetes mellitus was noted in 1.5% of patients. None of the patients were HIV-positive, 1.5% were hepatitis B–positive, and 2.3% were hepatitis C–positive. Of the 133 cases, 64.7% had localized GA and 30.8% had generalized GA. Photosensitive and papular GA were rarer (1.5% and 2.3% of cases, respectively). Use of a selective serotonin reuptake inhibitor (SSRI) was noted in 18.1% of patients; use of a calcium channel blocker was noted in 9.0% (Table 1).



The most commonly prescribed treatment of GA was topical steroids; 30.9% of patients who were prescribed a topical steroid experienced improvement of their condition. Intralesional triamcinolone was the second most prescribed treatment of GA, with an improvement rate of 40.0% (Table 2).

Comment

We attempted to determine the period of prevalence of GA in a tertiary care, university-based referral practice and evaluate disease associations, treatments, and outcomes of patients with biopsy-proven GA. Our calculated period prevalence of GA of 0.22% to 0.27% is consistent with another review, which reported that 0.1% to 0.4% of new patients presenting to a dermatology practice were given a diagnosis of GA.1 More than 85% of the cases we reviewed were seen in females, a finding that is more heavily skewed compared to prior reports that have suggested a female to male ratio of approximately 1:1 to 2:1.1-7 Our findings suggest that GA is a female-predominant condition, or women may be more likely to seek evaluation for the condition.

More than 95% of the cases we reviewed were localized (64.7%) or generalized (30.8%) GA, making these variants the most common forms of GA, which is consistent with prior reports.1-3,8,9 Other varieties of GA—drug induced, patch, perforating, photosensitive, palmar, and papular—appear rare. Because this study was conducted at an adult hospital, subcutaneous GA, which often is seen in children, may be underrepresented. As a retrospective chart review, it is possible that documentation is insufficient to capture each rare variant.

 

 


Concomitant Disorders and Unrelated Medical Therapy
Hypothyroidism is statistically significantly overrepresented in our patient population (30.1%) compared with an average prevalence of 1% to 2% in iodine-replete populations (Fisher exact test, P<.001).10 This finding is consistent with prior small studies and cases series, which have suggested an association between autoimmune thyroiditis and GA.11-14

Despite prior reports of a possible association between HIV and GA,15-24 none of our patients had a diagnosis of HIV. However, many of our patients were not tested for HIV, which confounds our results and may represent a practice gap in the field.

At 1.5%, the prevalence of type 1 diabetes mellitus in our patients is slightly higher than the national average of 0.3%.25 However, based on a Fisher exact test of analysis of proportions, this difference is not statistically significant (P=.106).

At 1.5% and 2.3%, the prevalence of hepatitis B and hepatitis C, respectively, in our patients is slightly higher than the national average of 0.5% and 1%, respectively.26 However, based on a Fisher exact test of analysis of proportions, these differences are not statistically significant (P=.142 and P=.146, respectively).

Given the high prevalence of hyperlipidemia in the United States (31.7%), this disease is not overrepresented in our sample (30.1%), though others have suggested there may be a connection.27,28 Based on a Fisher exact test, this difference of proportions is not statistically significant (P=.780).

Selective serotonin reuptake inhibitor use is common in the United States; approximately 11% of Americans older than 12 years use an SSRI.29 At 18.1%, the use of SSRIs in our patient group was statistically significantly higher than the national average (Fisher exact test, P=.017), suggesting a possible association between SSRI use and development of GA, warranting further investigation.

The use of calcium channel blockers, interferon, and tumor necrosis factor inhibitors was not significantly associated with GA in our series.

GA Therapy
The most commonly used treatments for GA in our study were topical steroids and intralesional triamcinolone, followed by hydroxychloroquine; all treatments employed exhibited a widely variable response. Assessing treatment response via retrospective chart review is challenging and response rates may not be accurately captured.

Study Limitations
Our study had several limitations. In calculating the period prevalence of GA, our query was limited by the number of years that the EMR has been in place. The number of cases we reviewed for clinical characteristics was limited to 133, as many cases with the ICD-9 diagnosis of GA were not biopsy proven and therefore were not included in our review. Many of the cases we reviewed were lost to follow-up, which prevented us from determining treatment outcomes.



Another weakness of our study was that our query did not provide an estimate of incidence or prevalence of GA overall, as this analysis was not a population-based study. The power of our study was limited by the number of cases of GA seen annually and the number of patients lost to follow-up. Additionally, our study population may only be generalizable to other large academic centers.

Conclusion

This study further solidifies our understanding of the epidemiology of GA and diseases that can be associated with GA. We identified a higher female to male ratio than previous reports, and consistent with prior reports, we noted potential associations with conditions such as thyroid disease and hyperlipidemia. Our population demonstrated higher rates of SSRI use than expected, warranting further investigation. Dermatologists should be aware of potential disease associations with GA, but as a whole we need better data and larger studies to determine the appropriate evaluation and treatment for patients with GA.

Granuloma annulare (GA) is a granulomatous skin disorder of uncertain etiology. A number of clinical variants exist, most commonly localized annular plaques on the hands or feet, generalized lesions, or subcutaneous nodules in children. Histologically, GA exhibits granulomatous inflammation with either interstitial or palisading lymphocytes and histiocytes with mucin deposition.

Few data exist regarding the epidemiology of GA. Although the pathogenesis of GA is unknown, associations between GA and underlying systemic processes, such as diabetes mellitus, hyperlipidemia, thyroid disease, and human immunodeficiency virus (HIV), have been suggested.



The purpose of this retrospective study was to determine the number of cases of GA seen annually at the Department of Dermatology at the University of Pennsylvania (Philadelphia, Pennsylvania) from 2008 to 2014. Additionally, we reviewed all cases of biopsy-proven GA from 2010 to 2014 and reported the demographics, underlying medical comorbidities, medications, treatments, and outcomes seen in this patient population.

Methods

We identified the number of outpatients presenting with GA annually using PennSeek, a tool developed by the Penn Medicine Data Analytics Center to search electronic medical records (EMRs). We queried the EMR database to determine the number of discrete patients seen at the Department of Dermatology at the University of Pennsylvania annually from 2008 (the year the EMR was established) to 2014. We then used PennSeek to determine the number of patients given a diagnosis of GA annually from 2008 to 2014 based on the International Classification of Diseases, Ninth Revision (ICD-9).

After using PennSeek to identify all patients given the ICD-9 diagnosis of GA from 2008 to 2014, we reviewed the EMRs of these patients to identify cases that were biopsy proven. For the biopsy-proven cases of GA seen at the University of Pennsylvania from 2010 to 2014, we reviewed the EMRs of these patients for clinical characteristics and treatment outcomes. For each case, we recorded the patient’s age, sex, medical comorbidities, GA subtype, and medications.

This study was approved by the University of Pennsylvania’s institutional review board.

 

 

Results

On average, the percentage of patients given a diagnosis of GA annually was 0.22% (95% CI, 0.19%-0.24%). A Pearson χ2 test was used to determine if any single annual percentage was significantly different from the others. We found a P value of .321, which suggests that the percentage of patients with GA seen annually has been stable from 2008 to 2014 (Figure).

Proportion of patients diagnosed with granuloma annulare (GA) annually (2008-2014).

There were 133 cases of biopsy-proven GA that were reviewed for clinical characteristics; of them, 86.5% were female. Thyroid disease was noted in 30.1% of patients, hyperlipidemia in 30.1%, and hematologic malignancies in 3.8%. Type 1 diabetes mellitus was noted in 1.5% of patients. None of the patients were HIV-positive, 1.5% were hepatitis B–positive, and 2.3% were hepatitis C–positive. Of the 133 cases, 64.7% had localized GA and 30.8% had generalized GA. Photosensitive and papular GA were rarer (1.5% and 2.3% of cases, respectively). Use of a selective serotonin reuptake inhibitor (SSRI) was noted in 18.1% of patients; use of a calcium channel blocker was noted in 9.0% (Table 1).



The most commonly prescribed treatment of GA was topical steroids; 30.9% of patients who were prescribed a topical steroid experienced improvement of their condition. Intralesional triamcinolone was the second most prescribed treatment of GA, with an improvement rate of 40.0% (Table 2).

Comment

We attempted to determine the period of prevalence of GA in a tertiary care, university-based referral practice and evaluate disease associations, treatments, and outcomes of patients with biopsy-proven GA. Our calculated period prevalence of GA of 0.22% to 0.27% is consistent with another review, which reported that 0.1% to 0.4% of new patients presenting to a dermatology practice were given a diagnosis of GA.1 More than 85% of the cases we reviewed were seen in females, a finding that is more heavily skewed compared to prior reports that have suggested a female to male ratio of approximately 1:1 to 2:1.1-7 Our findings suggest that GA is a female-predominant condition, or women may be more likely to seek evaluation for the condition.

More than 95% of the cases we reviewed were localized (64.7%) or generalized (30.8%) GA, making these variants the most common forms of GA, which is consistent with prior reports.1-3,8,9 Other varieties of GA—drug induced, patch, perforating, photosensitive, palmar, and papular—appear rare. Because this study was conducted at an adult hospital, subcutaneous GA, which often is seen in children, may be underrepresented. As a retrospective chart review, it is possible that documentation is insufficient to capture each rare variant.

 

 


Concomitant Disorders and Unrelated Medical Therapy
Hypothyroidism is statistically significantly overrepresented in our patient population (30.1%) compared with an average prevalence of 1% to 2% in iodine-replete populations (Fisher exact test, P<.001).10 This finding is consistent with prior small studies and cases series, which have suggested an association between autoimmune thyroiditis and GA.11-14

Despite prior reports of a possible association between HIV and GA,15-24 none of our patients had a diagnosis of HIV. However, many of our patients were not tested for HIV, which confounds our results and may represent a practice gap in the field.

At 1.5%, the prevalence of type 1 diabetes mellitus in our patients is slightly higher than the national average of 0.3%.25 However, based on a Fisher exact test of analysis of proportions, this difference is not statistically significant (P=.106).

At 1.5% and 2.3%, the prevalence of hepatitis B and hepatitis C, respectively, in our patients is slightly higher than the national average of 0.5% and 1%, respectively.26 However, based on a Fisher exact test of analysis of proportions, these differences are not statistically significant (P=.142 and P=.146, respectively).

Given the high prevalence of hyperlipidemia in the United States (31.7%), this disease is not overrepresented in our sample (30.1%), though others have suggested there may be a connection.27,28 Based on a Fisher exact test, this difference of proportions is not statistically significant (P=.780).

Selective serotonin reuptake inhibitor use is common in the United States; approximately 11% of Americans older than 12 years use an SSRI.29 At 18.1%, the use of SSRIs in our patient group was statistically significantly higher than the national average (Fisher exact test, P=.017), suggesting a possible association between SSRI use and development of GA, warranting further investigation.

The use of calcium channel blockers, interferon, and tumor necrosis factor inhibitors was not significantly associated with GA in our series.

GA Therapy
The most commonly used treatments for GA in our study were topical steroids and intralesional triamcinolone, followed by hydroxychloroquine; all treatments employed exhibited a widely variable response. Assessing treatment response via retrospective chart review is challenging and response rates may not be accurately captured.

Study Limitations
Our study had several limitations. In calculating the period prevalence of GA, our query was limited by the number of years that the EMR has been in place. The number of cases we reviewed for clinical characteristics was limited to 133, as many cases with the ICD-9 diagnosis of GA were not biopsy proven and therefore were not included in our review. Many of the cases we reviewed were lost to follow-up, which prevented us from determining treatment outcomes.



Another weakness of our study was that our query did not provide an estimate of incidence or prevalence of GA overall, as this analysis was not a population-based study. The power of our study was limited by the number of cases of GA seen annually and the number of patients lost to follow-up. Additionally, our study population may only be generalizable to other large academic centers.

Conclusion

This study further solidifies our understanding of the epidemiology of GA and diseases that can be associated with GA. We identified a higher female to male ratio than previous reports, and consistent with prior reports, we noted potential associations with conditions such as thyroid disease and hyperlipidemia. Our population demonstrated higher rates of SSRI use than expected, warranting further investigation. Dermatologists should be aware of potential disease associations with GA, but as a whole we need better data and larger studies to determine the appropriate evaluation and treatment for patients with GA.

References
  1. Muhlbauer JE. Granuloma annulare. J Am Acad Dermatol. 1980;3:217-230.
  2. Thornsberry LA, English JC 3rd. Etiology, diagnosis, and therapeutic management of granuloma annulare: an update. Am J Clin Dermatol. 2013;14:279-290.
  3. Wells RS, Smith MA. The natural history of granuloma annulare. Br J Dermatol. 1963;75:199-205.
  4. Wallet-Faber N, Farhi D, Gorin I, et al. Outcome of granuloma annulare: shorter duration is associated with younger age and recent onset. J Eur Acad Dermatol Venereol. 2010;24:103-104.
  5. Dahl MV. Granuloma annulare: long-term follow-up. Arch Dermatol. 2007;143:946-947.
  6. Yun JH, Lee JY, Kim MK, et al. Clinical and pathological features of generalized granuloma annulare with their correlation: a retrospective multicenter study in Korea. Ann Dermatol. 2009;21:113-119.
  7. Tan HH, Goh CL. Granuloma annulare: a review of 41 cases at the National Skin Centre. Ann Acad Med Singapore. 2000;29:714-718.
  8. Cyr PR. Diagnosis and management of granuloma annulare. Am Fam Physician. 2006;74:1729-1734.
  9. Smith MD, Downie JB, DiCostanzo D. Granuloma annulare. Int J Dermatol. 1997;36:326-333.
  10. Vanderpump MPJ. The epidemiology of thyroid diseases. In: Braverman LE, Utiger RD, eds. Werner and Ingbar’s The Thyroid: A Fundamental and Clinical Text. 9th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2005:398-496.
  11. Vázquez-López F, Pereiro M Jr, Manjón Haces JA, et al. Localized granuloma annulare and autoimmune thyroiditis in adult women: a case-control study. J Am Acad Dermatol. 2003;48:517-520.
  12. Vázquez-López F, González-López MA, Raya-Aguado C, et al. Localized granuloma annulare and autoimmune thyroiditis: a new case report. J Am Acad Dermatol. 2000;43(5, pt 2):943-945.
  13. Kappeler D, Troendle A, Mueller B. Localized granuloma annulare associated with autoimmune thyroid disease in a patient with a positive family history for autoimmune polyglandular syndrome type II. Eur J Endocrinol. 2001;145:101-102.
  14. Maschio M, Marigliano M, Sabbion A, et al. A rare case of granuloma annulare in a 5-year-old child with type 1 diabetes and autoimmune thyroiditis. Am J Dermatopathol. 2013;35:385-387.
  15. Smith NP. AIDS, Kaposi’s sarcoma and the dermatologist. J R Soc Med. 1985;78:97-99.
  16. Huerter CJ, Bass J, Bergfeld WF, et al. Perforating granuloma annulare in a patient with acquired immunodeficiency syndrome. Immunohistologic evaluation of the cellular infiltrate. Arch Dermatol. 1987;123:1217-1220.
  17. Jones SK, Harman RR. Atypical granuloma annulare in patients with the acquired immunodeficiency syndrome. J Am Acad Dermatol. 1989;20(2 pt 1):299-300.
  18. Devesa Parente JA, Dores JA, Aranha JM. Generalized perforating granuloma annulare: case report. Acta Dermatovenerol Croat. 2012;20:260-262.
  19. Ghadially R, Sibbald RG, Walter JB, et al. Granuloma annulare in patients with human immunodeficiency virus infections. J Am Acad Dermatol. 1989;20(2, pt 1):232-235.
  20. Toro JR, Chu P, Yen TS, et al. Granuloma annulare and human immunodeficiency virus infection. Arch Dermatol. 1999;135:1341-1346.
  21. Cohen PR. Granuloma annulare: a mucocutaneous condition in human immunodeficiency virus-infected patients. Arch Dermatol. 1999;135:1404-1407.
  22. O’Moore EJ, Nandawni R, Uthayakumar S, et al. HIV-associated granuloma annulare (HAGA): a report of six cases. Br J Dermatol. 2000;142:1054-1056.
  23. Kapembwa MS, Goolamali SK, Price A, et al. Granuloma annulare masquerading as molluscum contagiosum-like eruption in an HIV-positive African woman. J Am Acad Dermatol. 2003;49(suppl 2):S184-S186.
  24. Morris SD, Cerio R, Paige DG. An unusual presentation of diffuse granuloma annulare in an HIV-positive patient—immunohistochemical evidence of predominant CD8 lymphocytes. Clin Exp Dermatol. 2002;27:205-208.
  25. Maahs DM, West NA, Lawrence JM, et al. Epidemiology of type 1 diabetes. Endocrinol Metab Clin North Am. 2010;39:481-497.
  26. Centers for Disease Control and Prevention. Viral hepatitis surveillance—United States, 2010. www.cdc.gov/hepatitis/statistics/2010surveillance/commentary.htm. Accessed November 10, 2018.
  27. Mozaffarian D, Benjamin EJ, Go AS, et al; American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2015 update: a report from the American Heart Association. Circulation. 2015;131:E29-E322.
  28. Wu W, Robinson-Bostom L, Kokkotou E, et al. Dyslipidemia in granuloma annulare: a case-control study. Arch Dermatol. 2012;148:1131-1136.
  29. Pratt LA, Brody DJ, Gu Q. Antidepressant Use in Persons Aged 12 and Over: United States, 2005-2008. NCHS Data Brief, No. 76. Hyattsville, MD: National Center for Health Statistics; 2011. http://www.cdc.gov/nchs/data/databriefs/db76.htm. Updated October 19, 2011. Accessed June 1, 2014.
References
  1. Muhlbauer JE. Granuloma annulare. J Am Acad Dermatol. 1980;3:217-230.
  2. Thornsberry LA, English JC 3rd. Etiology, diagnosis, and therapeutic management of granuloma annulare: an update. Am J Clin Dermatol. 2013;14:279-290.
  3. Wells RS, Smith MA. The natural history of granuloma annulare. Br J Dermatol. 1963;75:199-205.
  4. Wallet-Faber N, Farhi D, Gorin I, et al. Outcome of granuloma annulare: shorter duration is associated with younger age and recent onset. J Eur Acad Dermatol Venereol. 2010;24:103-104.
  5. Dahl MV. Granuloma annulare: long-term follow-up. Arch Dermatol. 2007;143:946-947.
  6. Yun JH, Lee JY, Kim MK, et al. Clinical and pathological features of generalized granuloma annulare with their correlation: a retrospective multicenter study in Korea. Ann Dermatol. 2009;21:113-119.
  7. Tan HH, Goh CL. Granuloma annulare: a review of 41 cases at the National Skin Centre. Ann Acad Med Singapore. 2000;29:714-718.
  8. Cyr PR. Diagnosis and management of granuloma annulare. Am Fam Physician. 2006;74:1729-1734.
  9. Smith MD, Downie JB, DiCostanzo D. Granuloma annulare. Int J Dermatol. 1997;36:326-333.
  10. Vanderpump MPJ. The epidemiology of thyroid diseases. In: Braverman LE, Utiger RD, eds. Werner and Ingbar’s The Thyroid: A Fundamental and Clinical Text. 9th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2005:398-496.
  11. Vázquez-López F, Pereiro M Jr, Manjón Haces JA, et al. Localized granuloma annulare and autoimmune thyroiditis in adult women: a case-control study. J Am Acad Dermatol. 2003;48:517-520.
  12. Vázquez-López F, González-López MA, Raya-Aguado C, et al. Localized granuloma annulare and autoimmune thyroiditis: a new case report. J Am Acad Dermatol. 2000;43(5, pt 2):943-945.
  13. Kappeler D, Troendle A, Mueller B. Localized granuloma annulare associated with autoimmune thyroid disease in a patient with a positive family history for autoimmune polyglandular syndrome type II. Eur J Endocrinol. 2001;145:101-102.
  14. Maschio M, Marigliano M, Sabbion A, et al. A rare case of granuloma annulare in a 5-year-old child with type 1 diabetes and autoimmune thyroiditis. Am J Dermatopathol. 2013;35:385-387.
  15. Smith NP. AIDS, Kaposi’s sarcoma and the dermatologist. J R Soc Med. 1985;78:97-99.
  16. Huerter CJ, Bass J, Bergfeld WF, et al. Perforating granuloma annulare in a patient with acquired immunodeficiency syndrome. Immunohistologic evaluation of the cellular infiltrate. Arch Dermatol. 1987;123:1217-1220.
  17. Jones SK, Harman RR. Atypical granuloma annulare in patients with the acquired immunodeficiency syndrome. J Am Acad Dermatol. 1989;20(2 pt 1):299-300.
  18. Devesa Parente JA, Dores JA, Aranha JM. Generalized perforating granuloma annulare: case report. Acta Dermatovenerol Croat. 2012;20:260-262.
  19. Ghadially R, Sibbald RG, Walter JB, et al. Granuloma annulare in patients with human immunodeficiency virus infections. J Am Acad Dermatol. 1989;20(2, pt 1):232-235.
  20. Toro JR, Chu P, Yen TS, et al. Granuloma annulare and human immunodeficiency virus infection. Arch Dermatol. 1999;135:1341-1346.
  21. Cohen PR. Granuloma annulare: a mucocutaneous condition in human immunodeficiency virus-infected patients. Arch Dermatol. 1999;135:1404-1407.
  22. O’Moore EJ, Nandawni R, Uthayakumar S, et al. HIV-associated granuloma annulare (HAGA): a report of six cases. Br J Dermatol. 2000;142:1054-1056.
  23. Kapembwa MS, Goolamali SK, Price A, et al. Granuloma annulare masquerading as molluscum contagiosum-like eruption in an HIV-positive African woman. J Am Acad Dermatol. 2003;49(suppl 2):S184-S186.
  24. Morris SD, Cerio R, Paige DG. An unusual presentation of diffuse granuloma annulare in an HIV-positive patient—immunohistochemical evidence of predominant CD8 lymphocytes. Clin Exp Dermatol. 2002;27:205-208.
  25. Maahs DM, West NA, Lawrence JM, et al. Epidemiology of type 1 diabetes. Endocrinol Metab Clin North Am. 2010;39:481-497.
  26. Centers for Disease Control and Prevention. Viral hepatitis surveillance—United States, 2010. www.cdc.gov/hepatitis/statistics/2010surveillance/commentary.htm. Accessed November 10, 2018.
  27. Mozaffarian D, Benjamin EJ, Go AS, et al; American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2015 update: a report from the American Heart Association. Circulation. 2015;131:E29-E322.
  28. Wu W, Robinson-Bostom L, Kokkotou E, et al. Dyslipidemia in granuloma annulare: a case-control study. Arch Dermatol. 2012;148:1131-1136.
  29. Pratt LA, Brody DJ, Gu Q. Antidepressant Use in Persons Aged 12 and Over: United States, 2005-2008. NCHS Data Brief, No. 76. Hyattsville, MD: National Center for Health Statistics; 2011. http://www.cdc.gov/nchs/data/databriefs/db76.htm. Updated October 19, 2011. Accessed June 1, 2014.
Issue
Cutis - 103(2)
Issue
Cutis - 103(2)
Page Number
102-106
Page Number
102-106
Publications
Publications
Topics
Article Type
Display Headline
Granuloma Annulare: A Retrospective Series of 133 Patients
Display Headline
Granuloma Annulare: A Retrospective Series of 133 Patients
Sections
Inside the Article

Practice Points

  • Although the pathogenesis of granuloma annulare (GA) is unknown, associations between the disorder and underlying systemic processes (eg, diabetes mellitus, hyperlipidemia, thyroid disease, human immunodeficiency virus) have been proposed.
  • This study elicited a period prevalence of GA of 0.22% to 0.27%.
  • The most commonly used treatments of GA were topical steroids and intralesional triamcinolone, followed by hydroxychloroquine.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Clearance of Psoriasis After Ischemic Stroke

Article Type
Changed
Thu, 12/15/2022 - 14:42
Display Headline
Clearance of Psoriasis After Ischemic Stroke

The etiology of psoriasis is multifactorial, and it is attributed to both genetic and environmental components.1 One of the lesser-studied aspects of psoriasis pathogenesis is the involvement of the nervous system. It is thought that the pathogenesis involves inflammation of the cutaneous nerves,2 and cutaneous denervation has been shown to improve acanthosis and IL-23 expression in mice with psoriasiform skin.3 There also have been reports of psoriasis remission following peripheral and central nervous system injury from surgical nerve resection4 as well as cerebrovascular accident.5 We present a case of total psoriasis clearance following ischemic stroke.

Case Report

A 52-year-old man with psoriasis presented to the dermatology clinic for follow-up. The patient had been using topical clobetasol and apremilast with limited success but had not previously tried biologics. On physical examination he was noted to have erythematous, scaly, indurated papules and plaques on the chest, abdomen, back, arms, and legs, consistent with psoriasis. Affected body surface area was approximately 10%. Ustekinumab was prescribed, but the patient did not pick it up from the pharmacy.

Approximately 1 month later, the patient presented to the emergency department with left-sided weakness and numbness. He was hospitalized for treatment of stroke. During hospitalization, the patient was started on lisinopril, aspirin, and atorvastatin. He also was given subcutaneous enoxaparin with plans to initiate warfarin as an outpatient. His psoriasis was not treated with topical or systemic medications during the course of his admission. He was discharged to a skilled nursing facility after 3 days.



Three months following discharge, the patient returned to the dermatology clinic for follow-up. After his stroke, he reported that his psoriasis had cleared and had not returned. On physical examination his skin was clear of psoriatic lesions.

Comment

The nervous system is thought to play an important role in the pathophysiology of psoriasis. Evidence for this involvement includes the exacerbation of psoriasis with stress and the often symmetric distribution of psoriatic lesions.6

 

 

Moreover, numerous neuropeptides have been identified in the pathophysiology of psoriasis. Farber et al7 first proposed that release of substance P (SP) from cutaneous sensory nerve fibers causes a local neurogenic response that triggers psoriasis in predisposed individuals. The role of SP in psoriasis is unclear, as there have been reports of both higher8 and lower9 levels in involved and noninvolved skin of psoriatic patients compared to skin in healthy individuals. It has been suggested that numerous other neuropeptides, including nerve growth factor (NGF), calcitonin gene-related peptide, and vasoactive intestinal peptide, play a part in psoriasis.2,10 Specifically, NGF prevents apoptosis of keratinocytes11 and is found in higher levels in psoriatic skin compared to controls.12 Calcitonin gene-related peptide has been shown to stimulate keratinocyte proliferation13 and has been found at increased levels in psoriatic skin.14 Vasoactive intestinal peptide-positive nerve fibers in the epidermis and dermis are found in higher quantities in psoriatic plaques compared to nonlesional and normal skin.8

Neuropeptides also might play a role in the itching and Köbner phenomenon that accompany psoriasis. Increased levels of NGF in nonlesional skin of patients with psoriasis is thought to contribute to the development of psoriatic plaques following trauma by inducing an inflammatory response that upregulates other neuropeptides, such as SP and calcitonin gene-related peptide. These neuropeptides induce keratinocyte proliferation, which further increases NGF expression, thus creating a cycle of inflammation and formation of psoriatic lesions.6 Moreover, there is a notable correlation between pruritus severity and density of NGF-immunoreactive keratinocytes, high-affinity NGF receptors, protein gene product 9.5–immunoreactive intraepidermal fibers, and immunoreactive vessels for E-selectin.15

Spontaneous remission of psoriasis after cerebrovascular accident was first reported in 1998.5 Moreover, there have been cases of protective effects from psoriasis and psoriatic arthritis in limbs affected by poliomyelitis.16,17 In cases in which patients regained neurologic function, Zhu et al10 found that recurrence of skin lesions in areas corresponding to nervous system injury also occurred. However, in cases of permanent nerve damage, psoriasis did not return,10 confirming the role of peripheral nerves in the pathogenesis of psoriasis. It is thought that peripheral nerve damage results in decreased secretion of neuropeptides3 and that central nervous system injury also can cause similar downstream effects.10

Other reasons for the patient’s remission also were considered. Although it is possible that the sudden change in the patient’s usual environment could have induced remission of psoriasis, it seems more likely that the stress of the situation would have worsened his symptoms. Medications used during the patient’s hospitalization also were considered as reasons for symptom improvement. One study using a case-control and case-crossover design found psoriasis to be associated with nonsteroidal anti-inflammatory drugs and angiotensin-converting enzyme inhibitors (odds ratio, 4.0 and 2.1, respectively).18 Atorvastatin has been investigated as a potential treatment of psoriasis, though no therapeutic benefit has been proven.19,20 Heparin has been shown in case reports to improve psoriasis symptoms but was used in addition to standard psoriasis therapies and not as monotherapy.21

A more thorough understanding of which neuropeptides are directly implicated in the neurologic-mediated clearance of psoriasis might contribute to better targeted therapies. For example, infusion of peptide T, a vasoactive intestinal peptide analogue, was shown to have some effect in clearing the skin in 14 psoriasis patients.22 Although this finding has not been replicated, it demonstrates the potential utility of therapies targeted toward the neurologic aspects of psoriasis. More research is needed to evaluate the potential of targeting other neuropeptides for treatment of psoriatic plaques.

References
  1. Boehncke WH. Etiology and pathogenesis of psoriasis. Rheum Dis Clin North Am. 2015;41:665-675.
  2. Saraceno R, Kleyn CE, Terenghi G, et al. The role of neuropeptides in psoriasis. Br J Dermatol. 2006;155:876-882.
  3. Ostrowski SM, Belkai A, Loyd CM, et al. Cutaneous denervation of psoriasiform mouse skin improves acanthosis and inflammation in a sensory neuropeptide-dependent manner. J Invest Dermatol. 2011;131:1530-1538.
  4. Dewing SB. Remission of psoriasis associated with cutaneous nerve section. Arch Dermatol. 1971;104:220-221.
  5. Stratigos AJ, Katoulis AK, Stavrianeas NG. Spontaneous clearing of psoriasis after stroke. J Am Acad Dermatol. 1998;38(5, pt 1):768-770.
  6. Raychaudhuri SP, Farber EM. Neuroimmunologic aspects of psoriasis. Cutis. 2000;66:357-362.
  7. Farber EM, Nickoloff BJ, Recht B, et al. Stress, symmetry, and psoriasis: possible role of neuropeptides. J Am Acad Dermatol. 1986;14(2, pt 1):305-311.
  8. Al’Abadie MS, Senior HJ, Bleehen SS, et al. Neuropeptides and general neuronal marker in psoriasis—an immunohistochemical study. Clin Exp Dermatol. 1995;20:384-389.
  9. Pincelli C, Fantini F, Romualdi P, et al. Substance P is diminished and vasoactive intestinal peptide is augmented in psoriatic lesions and these peptides exert disparate effects on the proliferation of cultured human keratinocytes. J Invest Dermatol. 1992;98:421-427.
  10. Zhu TH, Nakamura M, Farahnik B, et al. The role of the nervous system in the pathophysiology of psoriasis: a review of cases of psoriasis remission or improvement following denervation injury. Am J Clin Dermatol. 2016;17:257-263.
  11. Pincelli C. Nerve growth factor and keratinocytes: a role in psoriasis. Eur J Dermatol. 2000;10:85-90.
  12. Raychaudhuri SP, Jiang WY, Farber EM. Psoriatic keratinocytes express high levels of nerve growth factor. Acta Derm Venereol. 1998;78:84-86.
  13. He Y, Ding G, Wang X, et al. Calcitonin gene‐related peptide in Langerhans cells in psoriatic plaque lesions. Chin Med J (Engl). 2000;113:747-751.
  14. Chu DQ, Choy M, Foster P, et al. A comparative study of the ability of calcitonin gene‐related peptide and adrenomedullin13–52 to modulate microvascular but not thermal hyperalgesia responses. Br J Pharmacol. 2000;130:1589-1596.
  15. Nakamura M, Toyoda M, Morohashi M. Pruritogenic mediators in psoriasis vulgaris: comparative evaluation of itch-associated cutaneous factors. Br J Dermatol. 2003;149:718-730.
  16. Wang TS, Tsai TF. Psoriasis sparing the lower limb with postpoliomeylitis residual paralysis. Br J Dermatol. 2014;171:429-431.
  17. Weiner SR, Bassett LW, Reichman RP. Protective effect of poliomyelitis on psoriatic arthritis. Arthritis Rheum. 1985;28:703-706.
  18. Cohen AD, Bonneh DY, Reuveni H, et al. Drug exposure and psoriasis vulgaris: case control and case-crossover studies. Acta Derm Venereol. 2005;85:299-303.
  19. Faghihi T, Radfar M, Mehrabian Z, et al. Atorvastatin for the treatment of plaque-type psoriasis. Pharmacotherapy. 2011;31:1045-1050.
  20. Chua SHH, Tioleco GMS, Dayrit CAF, et al. Atorvastatin as adjunctive therapy for chronic plaque type psoriasis versus betamethasone valerate alone: a randomized, double-blind, placebo-controlled trial. Indian J Dermatol Venereol Leprol. 2017;83:441-447.
  21. Jekel LG. Use of heparin in treatment of psoriasis. AMA Arch Derm Syphilol. 1953;68:80-82.
  22. Farber EM, Cohen EN, Trozak DJ, et al. Peptide T improves psoriasis when infused into lesions in nanogram amounts. J Am Acad Dermatol. 1991;25:658-664.
Article PDF
Author and Disclosure Information

Ms. Lee is from the John A. Burns School of Medicine, University of Hawaii, Honolulu. Ms. Reynolds is from the College of Medicine, University of Cincinnati, Ohio. Ms. Pithadia is from the Medical College of Georgia, Augusta University. Dr. Thiyanaratnam is from the Department of Dermatology, Kaiser Permanente Los Angeles Medical Center, California. Dr. Wu is from the Dermatology Research and Education Foundation, Irvine, California.

Ms. Lee, Ms. Reynolds, Ms. Pithadia, and Dr. Thiyanaratnam report no conflict of interest. Dr. Wu is an investigator for AbbVie; Amgen Inc; Eli Lilly & Company; Janssen Biotech, Inc; and Novartis. He also is consultant for Almirall, SA; Amgen Inc; Bristol-Myers Squibb Company; Dermira, Inc; Dr. Reddy's Laboratories Ltd; Eli Lilly & Company; Janssen Biotech, Inc; LEO Pharma Inc; and Promius Pharma. He also is a consultant and speaker for AbbVie; Celgene Corporation; Novartis; Regeneron Pharmaceuticals, Inc; Sun Pharmaceutical Industries Ltd; UCB, Inc; and Valeant Pharmaceuticals North America LLC.

Correspondence: Jashin J. Wu, MD ([email protected]).

Issue
Cutis - 103(2)
Publications
Topics
Page Number
74-76
Sections
Author and Disclosure Information

Ms. Lee is from the John A. Burns School of Medicine, University of Hawaii, Honolulu. Ms. Reynolds is from the College of Medicine, University of Cincinnati, Ohio. Ms. Pithadia is from the Medical College of Georgia, Augusta University. Dr. Thiyanaratnam is from the Department of Dermatology, Kaiser Permanente Los Angeles Medical Center, California. Dr. Wu is from the Dermatology Research and Education Foundation, Irvine, California.

Ms. Lee, Ms. Reynolds, Ms. Pithadia, and Dr. Thiyanaratnam report no conflict of interest. Dr. Wu is an investigator for AbbVie; Amgen Inc; Eli Lilly & Company; Janssen Biotech, Inc; and Novartis. He also is consultant for Almirall, SA; Amgen Inc; Bristol-Myers Squibb Company; Dermira, Inc; Dr. Reddy's Laboratories Ltd; Eli Lilly & Company; Janssen Biotech, Inc; LEO Pharma Inc; and Promius Pharma. He also is a consultant and speaker for AbbVie; Celgene Corporation; Novartis; Regeneron Pharmaceuticals, Inc; Sun Pharmaceutical Industries Ltd; UCB, Inc; and Valeant Pharmaceuticals North America LLC.

Correspondence: Jashin J. Wu, MD ([email protected]).

Author and Disclosure Information

Ms. Lee is from the John A. Burns School of Medicine, University of Hawaii, Honolulu. Ms. Reynolds is from the College of Medicine, University of Cincinnati, Ohio. Ms. Pithadia is from the Medical College of Georgia, Augusta University. Dr. Thiyanaratnam is from the Department of Dermatology, Kaiser Permanente Los Angeles Medical Center, California. Dr. Wu is from the Dermatology Research and Education Foundation, Irvine, California.

Ms. Lee, Ms. Reynolds, Ms. Pithadia, and Dr. Thiyanaratnam report no conflict of interest. Dr. Wu is an investigator for AbbVie; Amgen Inc; Eli Lilly & Company; Janssen Biotech, Inc; and Novartis. He also is consultant for Almirall, SA; Amgen Inc; Bristol-Myers Squibb Company; Dermira, Inc; Dr. Reddy's Laboratories Ltd; Eli Lilly & Company; Janssen Biotech, Inc; LEO Pharma Inc; and Promius Pharma. He also is a consultant and speaker for AbbVie; Celgene Corporation; Novartis; Regeneron Pharmaceuticals, Inc; Sun Pharmaceutical Industries Ltd; UCB, Inc; and Valeant Pharmaceuticals North America LLC.

Correspondence: Jashin J. Wu, MD ([email protected]).

Article PDF
Article PDF

The etiology of psoriasis is multifactorial, and it is attributed to both genetic and environmental components.1 One of the lesser-studied aspects of psoriasis pathogenesis is the involvement of the nervous system. It is thought that the pathogenesis involves inflammation of the cutaneous nerves,2 and cutaneous denervation has been shown to improve acanthosis and IL-23 expression in mice with psoriasiform skin.3 There also have been reports of psoriasis remission following peripheral and central nervous system injury from surgical nerve resection4 as well as cerebrovascular accident.5 We present a case of total psoriasis clearance following ischemic stroke.

Case Report

A 52-year-old man with psoriasis presented to the dermatology clinic for follow-up. The patient had been using topical clobetasol and apremilast with limited success but had not previously tried biologics. On physical examination he was noted to have erythematous, scaly, indurated papules and plaques on the chest, abdomen, back, arms, and legs, consistent with psoriasis. Affected body surface area was approximately 10%. Ustekinumab was prescribed, but the patient did not pick it up from the pharmacy.

Approximately 1 month later, the patient presented to the emergency department with left-sided weakness and numbness. He was hospitalized for treatment of stroke. During hospitalization, the patient was started on lisinopril, aspirin, and atorvastatin. He also was given subcutaneous enoxaparin with plans to initiate warfarin as an outpatient. His psoriasis was not treated with topical or systemic medications during the course of his admission. He was discharged to a skilled nursing facility after 3 days.



Three months following discharge, the patient returned to the dermatology clinic for follow-up. After his stroke, he reported that his psoriasis had cleared and had not returned. On physical examination his skin was clear of psoriatic lesions.

Comment

The nervous system is thought to play an important role in the pathophysiology of psoriasis. Evidence for this involvement includes the exacerbation of psoriasis with stress and the often symmetric distribution of psoriatic lesions.6

 

 

Moreover, numerous neuropeptides have been identified in the pathophysiology of psoriasis. Farber et al7 first proposed that release of substance P (SP) from cutaneous sensory nerve fibers causes a local neurogenic response that triggers psoriasis in predisposed individuals. The role of SP in psoriasis is unclear, as there have been reports of both higher8 and lower9 levels in involved and noninvolved skin of psoriatic patients compared to skin in healthy individuals. It has been suggested that numerous other neuropeptides, including nerve growth factor (NGF), calcitonin gene-related peptide, and vasoactive intestinal peptide, play a part in psoriasis.2,10 Specifically, NGF prevents apoptosis of keratinocytes11 and is found in higher levels in psoriatic skin compared to controls.12 Calcitonin gene-related peptide has been shown to stimulate keratinocyte proliferation13 and has been found at increased levels in psoriatic skin.14 Vasoactive intestinal peptide-positive nerve fibers in the epidermis and dermis are found in higher quantities in psoriatic plaques compared to nonlesional and normal skin.8

Neuropeptides also might play a role in the itching and Köbner phenomenon that accompany psoriasis. Increased levels of NGF in nonlesional skin of patients with psoriasis is thought to contribute to the development of psoriatic plaques following trauma by inducing an inflammatory response that upregulates other neuropeptides, such as SP and calcitonin gene-related peptide. These neuropeptides induce keratinocyte proliferation, which further increases NGF expression, thus creating a cycle of inflammation and formation of psoriatic lesions.6 Moreover, there is a notable correlation between pruritus severity and density of NGF-immunoreactive keratinocytes, high-affinity NGF receptors, protein gene product 9.5–immunoreactive intraepidermal fibers, and immunoreactive vessels for E-selectin.15

Spontaneous remission of psoriasis after cerebrovascular accident was first reported in 1998.5 Moreover, there have been cases of protective effects from psoriasis and psoriatic arthritis in limbs affected by poliomyelitis.16,17 In cases in which patients regained neurologic function, Zhu et al10 found that recurrence of skin lesions in areas corresponding to nervous system injury also occurred. However, in cases of permanent nerve damage, psoriasis did not return,10 confirming the role of peripheral nerves in the pathogenesis of psoriasis. It is thought that peripheral nerve damage results in decreased secretion of neuropeptides3 and that central nervous system injury also can cause similar downstream effects.10

Other reasons for the patient’s remission also were considered. Although it is possible that the sudden change in the patient’s usual environment could have induced remission of psoriasis, it seems more likely that the stress of the situation would have worsened his symptoms. Medications used during the patient’s hospitalization also were considered as reasons for symptom improvement. One study using a case-control and case-crossover design found psoriasis to be associated with nonsteroidal anti-inflammatory drugs and angiotensin-converting enzyme inhibitors (odds ratio, 4.0 and 2.1, respectively).18 Atorvastatin has been investigated as a potential treatment of psoriasis, though no therapeutic benefit has been proven.19,20 Heparin has been shown in case reports to improve psoriasis symptoms but was used in addition to standard psoriasis therapies and not as monotherapy.21

A more thorough understanding of which neuropeptides are directly implicated in the neurologic-mediated clearance of psoriasis might contribute to better targeted therapies. For example, infusion of peptide T, a vasoactive intestinal peptide analogue, was shown to have some effect in clearing the skin in 14 psoriasis patients.22 Although this finding has not been replicated, it demonstrates the potential utility of therapies targeted toward the neurologic aspects of psoriasis. More research is needed to evaluate the potential of targeting other neuropeptides for treatment of psoriatic plaques.

The etiology of psoriasis is multifactorial, and it is attributed to both genetic and environmental components.1 One of the lesser-studied aspects of psoriasis pathogenesis is the involvement of the nervous system. It is thought that the pathogenesis involves inflammation of the cutaneous nerves,2 and cutaneous denervation has been shown to improve acanthosis and IL-23 expression in mice with psoriasiform skin.3 There also have been reports of psoriasis remission following peripheral and central nervous system injury from surgical nerve resection4 as well as cerebrovascular accident.5 We present a case of total psoriasis clearance following ischemic stroke.

Case Report

A 52-year-old man with psoriasis presented to the dermatology clinic for follow-up. The patient had been using topical clobetasol and apremilast with limited success but had not previously tried biologics. On physical examination he was noted to have erythematous, scaly, indurated papules and plaques on the chest, abdomen, back, arms, and legs, consistent with psoriasis. Affected body surface area was approximately 10%. Ustekinumab was prescribed, but the patient did not pick it up from the pharmacy.

Approximately 1 month later, the patient presented to the emergency department with left-sided weakness and numbness. He was hospitalized for treatment of stroke. During hospitalization, the patient was started on lisinopril, aspirin, and atorvastatin. He also was given subcutaneous enoxaparin with plans to initiate warfarin as an outpatient. His psoriasis was not treated with topical or systemic medications during the course of his admission. He was discharged to a skilled nursing facility after 3 days.



Three months following discharge, the patient returned to the dermatology clinic for follow-up. After his stroke, he reported that his psoriasis had cleared and had not returned. On physical examination his skin was clear of psoriatic lesions.

Comment

The nervous system is thought to play an important role in the pathophysiology of psoriasis. Evidence for this involvement includes the exacerbation of psoriasis with stress and the often symmetric distribution of psoriatic lesions.6

 

 

Moreover, numerous neuropeptides have been identified in the pathophysiology of psoriasis. Farber et al7 first proposed that release of substance P (SP) from cutaneous sensory nerve fibers causes a local neurogenic response that triggers psoriasis in predisposed individuals. The role of SP in psoriasis is unclear, as there have been reports of both higher8 and lower9 levels in involved and noninvolved skin of psoriatic patients compared to skin in healthy individuals. It has been suggested that numerous other neuropeptides, including nerve growth factor (NGF), calcitonin gene-related peptide, and vasoactive intestinal peptide, play a part in psoriasis.2,10 Specifically, NGF prevents apoptosis of keratinocytes11 and is found in higher levels in psoriatic skin compared to controls.12 Calcitonin gene-related peptide has been shown to stimulate keratinocyte proliferation13 and has been found at increased levels in psoriatic skin.14 Vasoactive intestinal peptide-positive nerve fibers in the epidermis and dermis are found in higher quantities in psoriatic plaques compared to nonlesional and normal skin.8

Neuropeptides also might play a role in the itching and Köbner phenomenon that accompany psoriasis. Increased levels of NGF in nonlesional skin of patients with psoriasis is thought to contribute to the development of psoriatic plaques following trauma by inducing an inflammatory response that upregulates other neuropeptides, such as SP and calcitonin gene-related peptide. These neuropeptides induce keratinocyte proliferation, which further increases NGF expression, thus creating a cycle of inflammation and formation of psoriatic lesions.6 Moreover, there is a notable correlation between pruritus severity and density of NGF-immunoreactive keratinocytes, high-affinity NGF receptors, protein gene product 9.5–immunoreactive intraepidermal fibers, and immunoreactive vessels for E-selectin.15

Spontaneous remission of psoriasis after cerebrovascular accident was first reported in 1998.5 Moreover, there have been cases of protective effects from psoriasis and psoriatic arthritis in limbs affected by poliomyelitis.16,17 In cases in which patients regained neurologic function, Zhu et al10 found that recurrence of skin lesions in areas corresponding to nervous system injury also occurred. However, in cases of permanent nerve damage, psoriasis did not return,10 confirming the role of peripheral nerves in the pathogenesis of psoriasis. It is thought that peripheral nerve damage results in decreased secretion of neuropeptides3 and that central nervous system injury also can cause similar downstream effects.10

Other reasons for the patient’s remission also were considered. Although it is possible that the sudden change in the patient’s usual environment could have induced remission of psoriasis, it seems more likely that the stress of the situation would have worsened his symptoms. Medications used during the patient’s hospitalization also were considered as reasons for symptom improvement. One study using a case-control and case-crossover design found psoriasis to be associated with nonsteroidal anti-inflammatory drugs and angiotensin-converting enzyme inhibitors (odds ratio, 4.0 and 2.1, respectively).18 Atorvastatin has been investigated as a potential treatment of psoriasis, though no therapeutic benefit has been proven.19,20 Heparin has been shown in case reports to improve psoriasis symptoms but was used in addition to standard psoriasis therapies and not as monotherapy.21

A more thorough understanding of which neuropeptides are directly implicated in the neurologic-mediated clearance of psoriasis might contribute to better targeted therapies. For example, infusion of peptide T, a vasoactive intestinal peptide analogue, was shown to have some effect in clearing the skin in 14 psoriasis patients.22 Although this finding has not been replicated, it demonstrates the potential utility of therapies targeted toward the neurologic aspects of psoriasis. More research is needed to evaluate the potential of targeting other neuropeptides for treatment of psoriatic plaques.

References
  1. Boehncke WH. Etiology and pathogenesis of psoriasis. Rheum Dis Clin North Am. 2015;41:665-675.
  2. Saraceno R, Kleyn CE, Terenghi G, et al. The role of neuropeptides in psoriasis. Br J Dermatol. 2006;155:876-882.
  3. Ostrowski SM, Belkai A, Loyd CM, et al. Cutaneous denervation of psoriasiform mouse skin improves acanthosis and inflammation in a sensory neuropeptide-dependent manner. J Invest Dermatol. 2011;131:1530-1538.
  4. Dewing SB. Remission of psoriasis associated with cutaneous nerve section. Arch Dermatol. 1971;104:220-221.
  5. Stratigos AJ, Katoulis AK, Stavrianeas NG. Spontaneous clearing of psoriasis after stroke. J Am Acad Dermatol. 1998;38(5, pt 1):768-770.
  6. Raychaudhuri SP, Farber EM. Neuroimmunologic aspects of psoriasis. Cutis. 2000;66:357-362.
  7. Farber EM, Nickoloff BJ, Recht B, et al. Stress, symmetry, and psoriasis: possible role of neuropeptides. J Am Acad Dermatol. 1986;14(2, pt 1):305-311.
  8. Al’Abadie MS, Senior HJ, Bleehen SS, et al. Neuropeptides and general neuronal marker in psoriasis—an immunohistochemical study. Clin Exp Dermatol. 1995;20:384-389.
  9. Pincelli C, Fantini F, Romualdi P, et al. Substance P is diminished and vasoactive intestinal peptide is augmented in psoriatic lesions and these peptides exert disparate effects on the proliferation of cultured human keratinocytes. J Invest Dermatol. 1992;98:421-427.
  10. Zhu TH, Nakamura M, Farahnik B, et al. The role of the nervous system in the pathophysiology of psoriasis: a review of cases of psoriasis remission or improvement following denervation injury. Am J Clin Dermatol. 2016;17:257-263.
  11. Pincelli C. Nerve growth factor and keratinocytes: a role in psoriasis. Eur J Dermatol. 2000;10:85-90.
  12. Raychaudhuri SP, Jiang WY, Farber EM. Psoriatic keratinocytes express high levels of nerve growth factor. Acta Derm Venereol. 1998;78:84-86.
  13. He Y, Ding G, Wang X, et al. Calcitonin gene‐related peptide in Langerhans cells in psoriatic plaque lesions. Chin Med J (Engl). 2000;113:747-751.
  14. Chu DQ, Choy M, Foster P, et al. A comparative study of the ability of calcitonin gene‐related peptide and adrenomedullin13–52 to modulate microvascular but not thermal hyperalgesia responses. Br J Pharmacol. 2000;130:1589-1596.
  15. Nakamura M, Toyoda M, Morohashi M. Pruritogenic mediators in psoriasis vulgaris: comparative evaluation of itch-associated cutaneous factors. Br J Dermatol. 2003;149:718-730.
  16. Wang TS, Tsai TF. Psoriasis sparing the lower limb with postpoliomeylitis residual paralysis. Br J Dermatol. 2014;171:429-431.
  17. Weiner SR, Bassett LW, Reichman RP. Protective effect of poliomyelitis on psoriatic arthritis. Arthritis Rheum. 1985;28:703-706.
  18. Cohen AD, Bonneh DY, Reuveni H, et al. Drug exposure and psoriasis vulgaris: case control and case-crossover studies. Acta Derm Venereol. 2005;85:299-303.
  19. Faghihi T, Radfar M, Mehrabian Z, et al. Atorvastatin for the treatment of plaque-type psoriasis. Pharmacotherapy. 2011;31:1045-1050.
  20. Chua SHH, Tioleco GMS, Dayrit CAF, et al. Atorvastatin as adjunctive therapy for chronic plaque type psoriasis versus betamethasone valerate alone: a randomized, double-blind, placebo-controlled trial. Indian J Dermatol Venereol Leprol. 2017;83:441-447.
  21. Jekel LG. Use of heparin in treatment of psoriasis. AMA Arch Derm Syphilol. 1953;68:80-82.
  22. Farber EM, Cohen EN, Trozak DJ, et al. Peptide T improves psoriasis when infused into lesions in nanogram amounts. J Am Acad Dermatol. 1991;25:658-664.
References
  1. Boehncke WH. Etiology and pathogenesis of psoriasis. Rheum Dis Clin North Am. 2015;41:665-675.
  2. Saraceno R, Kleyn CE, Terenghi G, et al. The role of neuropeptides in psoriasis. Br J Dermatol. 2006;155:876-882.
  3. Ostrowski SM, Belkai A, Loyd CM, et al. Cutaneous denervation of psoriasiform mouse skin improves acanthosis and inflammation in a sensory neuropeptide-dependent manner. J Invest Dermatol. 2011;131:1530-1538.
  4. Dewing SB. Remission of psoriasis associated with cutaneous nerve section. Arch Dermatol. 1971;104:220-221.
  5. Stratigos AJ, Katoulis AK, Stavrianeas NG. Spontaneous clearing of psoriasis after stroke. J Am Acad Dermatol. 1998;38(5, pt 1):768-770.
  6. Raychaudhuri SP, Farber EM. Neuroimmunologic aspects of psoriasis. Cutis. 2000;66:357-362.
  7. Farber EM, Nickoloff BJ, Recht B, et al. Stress, symmetry, and psoriasis: possible role of neuropeptides. J Am Acad Dermatol. 1986;14(2, pt 1):305-311.
  8. Al’Abadie MS, Senior HJ, Bleehen SS, et al. Neuropeptides and general neuronal marker in psoriasis—an immunohistochemical study. Clin Exp Dermatol. 1995;20:384-389.
  9. Pincelli C, Fantini F, Romualdi P, et al. Substance P is diminished and vasoactive intestinal peptide is augmented in psoriatic lesions and these peptides exert disparate effects on the proliferation of cultured human keratinocytes. J Invest Dermatol. 1992;98:421-427.
  10. Zhu TH, Nakamura M, Farahnik B, et al. The role of the nervous system in the pathophysiology of psoriasis: a review of cases of psoriasis remission or improvement following denervation injury. Am J Clin Dermatol. 2016;17:257-263.
  11. Pincelli C. Nerve growth factor and keratinocytes: a role in psoriasis. Eur J Dermatol. 2000;10:85-90.
  12. Raychaudhuri SP, Jiang WY, Farber EM. Psoriatic keratinocytes express high levels of nerve growth factor. Acta Derm Venereol. 1998;78:84-86.
  13. He Y, Ding G, Wang X, et al. Calcitonin gene‐related peptide in Langerhans cells in psoriatic plaque lesions. Chin Med J (Engl). 2000;113:747-751.
  14. Chu DQ, Choy M, Foster P, et al. A comparative study of the ability of calcitonin gene‐related peptide and adrenomedullin13–52 to modulate microvascular but not thermal hyperalgesia responses. Br J Pharmacol. 2000;130:1589-1596.
  15. Nakamura M, Toyoda M, Morohashi M. Pruritogenic mediators in psoriasis vulgaris: comparative evaluation of itch-associated cutaneous factors. Br J Dermatol. 2003;149:718-730.
  16. Wang TS, Tsai TF. Psoriasis sparing the lower limb with postpoliomeylitis residual paralysis. Br J Dermatol. 2014;171:429-431.
  17. Weiner SR, Bassett LW, Reichman RP. Protective effect of poliomyelitis on psoriatic arthritis. Arthritis Rheum. 1985;28:703-706.
  18. Cohen AD, Bonneh DY, Reuveni H, et al. Drug exposure and psoriasis vulgaris: case control and case-crossover studies. Acta Derm Venereol. 2005;85:299-303.
  19. Faghihi T, Radfar M, Mehrabian Z, et al. Atorvastatin for the treatment of plaque-type psoriasis. Pharmacotherapy. 2011;31:1045-1050.
  20. Chua SHH, Tioleco GMS, Dayrit CAF, et al. Atorvastatin as adjunctive therapy for chronic plaque type psoriasis versus betamethasone valerate alone: a randomized, double-blind, placebo-controlled trial. Indian J Dermatol Venereol Leprol. 2017;83:441-447.
  21. Jekel LG. Use of heparin in treatment of psoriasis. AMA Arch Derm Syphilol. 1953;68:80-82.
  22. Farber EM, Cohen EN, Trozak DJ, et al. Peptide T improves psoriasis when infused into lesions in nanogram amounts. J Am Acad Dermatol. 1991;25:658-664.
Issue
Cutis - 103(2)
Issue
Cutis - 103(2)
Page Number
74-76
Page Number
74-76
Publications
Publications
Topics
Article Type
Display Headline
Clearance of Psoriasis After Ischemic Stroke
Display Headline
Clearance of Psoriasis After Ischemic Stroke
Sections
Inside the Article

Practice Points

  • Psoriasis is exacerbated in the presence of stress, and psoriatic lesions often have a symmetric distribution, which is evidence that the nervous system is involved in the pathophysiology of the condition.
  • Various neuropeptides are involved in the pathophysiology of psoriasis, including substance P, nerve growth factor, calcitonin gene-related peptide, and vasoactive intestinal peptide.
  • Peripheral nerve damage results in decreased secretion of neuropeptides, which can lead to remission of psoriasis.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Large Hemorrhagic Plaque With Central Crusting

Article Type
Changed
Mon, 02/11/2019 - 10:50
Display Headline
Large Hemorrhagic Plaque With Central Crusting

The Diagnosis: Bullous/Hemorrhagic Lichen Sclerosus et Atrophicus

Histopathologic examination revealed hyperkeratosis of the stratum corneum and thinning of the epidermis (Figure). Subepidermal edema and hemorrhage in the papillary dermis were seen. There were dilated vessels beneath the edema in the reticular dermis, as well as perivascular, perifollicular, and interstitial lymphocytic inflammation. No cytologic atypia characteristic of squamous cell carcinoma (SCC) and angiosarcoma or large lymphatic channels characteristic of lymphangioma were noted. Based on clinicopathologic correlation, the diagnosis of the bullous/hemorrhagic form of lichen sclerosus et atrophicus (LS&A) was made. The patient was treated with high-potency topical steroids with notable symptomatic improvement and rapid resolution of the hemorrhagic lesion.

A, Histologic analysis showed hyperkeratosis of the stratum corneum, papillary dermal sclerosis and edema, zone of lymphoid inflammation, and copious erythrocyte extravasation (H&E, original magnification ×100). B, Copious erythrocyte extravasation also was evident in the superficial dermis (H&E, original magnification ×40).

Lichen sclerosus et atrophicus is a chronic inflammatory condition with a predilection for the anogenital region, though rare cases of extragenital involvement have been reported. It is seen in both sexes and across all age groups, with notably higher prevalence in females in the fifth and sixth decades of life.1,2 Lichen sclerosus et atrophicus can be difficult to diagnose, as these patients may present to a variety of specialists, may be embarrassed by the condition and reluctant for full evaluation, or may have asymptomatic lesions.2,3 Rare cases of isolated extragenital involvement and hemorrhagic or bullous lesions further complicate the diagnosis.1,2 Despite these difficulties, diagnosis is essential, as there is potential for cosmetically and functionally detrimental scarring as well as atrophy and development of overlying malignancies. Lichen sclerosus et atrophicus is not curable and rarely remits spontaneously, but appropriate treatment strategies can help control the symptoms of the condition as well as its most devastating sequelae.3

For females, classic LS&A is most common in theprepubertal, perimenopausal, or postmenopausal periods, commonly involving the vulva or perineum. Symptoms include pruritus, burning sensation, dysuria, dyspareunia, and labial stenosis, among others. For males, most cases involve the glans penis in prepubertal boys or middleaged men, and symptoms include pruritus, new-onset phimosis, decreased sensation, painful erections, dysuria, and urinary obstruction.1-3 An estimated 97% of patients have some form of genital involvement with only 2.5% showing isolated extragenital involvement, though the latter may be underdiagnosed, as this area is more likely to be asymptomatic.3-6 Extragenital LS&A most often involves the neck and shoulders. The classic appearance of LS&A includes shiny, white-red macules and papules that ultimately coalesce into atrophic plaques and can be accompanied by fissuring or scarring, especially in the genital area.2 There is an increased risk for SCC associated with genital LS&A.1

Bullous/hemorrhagic LS&A has been described as a rare phenotype. One case report cited an increased incidence of this subtype in patients with exclusively extragenital lesions, and the authors considered blister formation to be a characteristic feature of extragenital LS&A. The pathogenesis of blister formation and hemorrhage in LS&A is not completely understood, but trauma is thought to play a role due to decreased stress tolerance from atrophic skin.4 Furthermore, distortion of blood vessel architecture in LS&A has been described with loss of the capillary network and enlargement of vessels along  the dermoepidermal junction, which also could play a role in hemorrhage. Differential diagnosis of the bullous/hemorrhagic type of LS&A includes bullous pemphigoid, bullous lichen planus, or bullous scleroderma.7 In our more exophytic hemorrhagic case, malignancies such as SCC or angiosarcoma also had to be considered. Unlike genital LS&A, extragenital LS&A including the bullous/hemorrhagic variant has not been linked to an increasedrisk for malignancy.1,5

The mainstay of treatment of all forms of LS&A is high-potency topical steroids, but topical retinoids, tacrolimus, and UVA phototherapy also have been used. Bullous/hemorrhagic lesions often resolve quickly with topical steroids, leaving behind more classic plaques in their place, which can be more refractory to treatment.5,7

References
  1. Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. J Am Acad Dermatol. 1995;32:393-416.
  2. Pugliese JM, Morey AF, Peterson AC. Lichen sclerosus: review of the literature and current recommendations for management. J Urol. 2007;178:2268-2276.
  3. Fistarol SK, Itin PH. Diagnosis and treatment of lichen sclerosus: an update. Am J Clin Dermatol. 2013;14:27-47.
  4. Kimura A, Kambe N, Satoh T, et al. Follicular keratosis and bullous formation are typical signs of extragenital lichen sclerosus. J Dermatol. 2011;38:834-836.
  5. Khatu S, Vasani R. Isolated, localised extragenital bullous lichen sclerosus et atrophicus: a rare entity. Indian J Dermatol. 2013;58:409.
  6. Luzar B, Neil SM, Calonje E. Angiokeratoma-like changes in extragenital and genital lichen sclerosus. J Cutan Pathol. 2009;36:540-542.
  7. Lima RS, Maquine GA, Schettini AP, et al. Bullous and hemorrhagic lichen sclerosus—case report. An Bras Dermatol. 2015;90 (3 suppl 1):118-120.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Henry Ford Hospital, Detroit, Michigan.

The authors report no conflict of interest.

Correspondence: Caitlin Farmer, MD, 3031 W Grand Blvd, Ste 800, Detroit, MI 48202 ([email protected]).

Issue
Cutis - 103(2)
Publications
Topics
Page Number
68,79-80
Sections
Author and Disclosure Information

From the Department of Dermatology, Henry Ford Hospital, Detroit, Michigan.

The authors report no conflict of interest.

Correspondence: Caitlin Farmer, MD, 3031 W Grand Blvd, Ste 800, Detroit, MI 48202 ([email protected]).

Author and Disclosure Information

From the Department of Dermatology, Henry Ford Hospital, Detroit, Michigan.

The authors report no conflict of interest.

Correspondence: Caitlin Farmer, MD, 3031 W Grand Blvd, Ste 800, Detroit, MI 48202 ([email protected]).

Article PDF
Article PDF
Related Articles

The Diagnosis: Bullous/Hemorrhagic Lichen Sclerosus et Atrophicus

Histopathologic examination revealed hyperkeratosis of the stratum corneum and thinning of the epidermis (Figure). Subepidermal edema and hemorrhage in the papillary dermis were seen. There were dilated vessels beneath the edema in the reticular dermis, as well as perivascular, perifollicular, and interstitial lymphocytic inflammation. No cytologic atypia characteristic of squamous cell carcinoma (SCC) and angiosarcoma or large lymphatic channels characteristic of lymphangioma were noted. Based on clinicopathologic correlation, the diagnosis of the bullous/hemorrhagic form of lichen sclerosus et atrophicus (LS&A) was made. The patient was treated with high-potency topical steroids with notable symptomatic improvement and rapid resolution of the hemorrhagic lesion.

A, Histologic analysis showed hyperkeratosis of the stratum corneum, papillary dermal sclerosis and edema, zone of lymphoid inflammation, and copious erythrocyte extravasation (H&E, original magnification ×100). B, Copious erythrocyte extravasation also was evident in the superficial dermis (H&E, original magnification ×40).

Lichen sclerosus et atrophicus is a chronic inflammatory condition with a predilection for the anogenital region, though rare cases of extragenital involvement have been reported. It is seen in both sexes and across all age groups, with notably higher prevalence in females in the fifth and sixth decades of life.1,2 Lichen sclerosus et atrophicus can be difficult to diagnose, as these patients may present to a variety of specialists, may be embarrassed by the condition and reluctant for full evaluation, or may have asymptomatic lesions.2,3 Rare cases of isolated extragenital involvement and hemorrhagic or bullous lesions further complicate the diagnosis.1,2 Despite these difficulties, diagnosis is essential, as there is potential for cosmetically and functionally detrimental scarring as well as atrophy and development of overlying malignancies. Lichen sclerosus et atrophicus is not curable and rarely remits spontaneously, but appropriate treatment strategies can help control the symptoms of the condition as well as its most devastating sequelae.3

For females, classic LS&A is most common in theprepubertal, perimenopausal, or postmenopausal periods, commonly involving the vulva or perineum. Symptoms include pruritus, burning sensation, dysuria, dyspareunia, and labial stenosis, among others. For males, most cases involve the glans penis in prepubertal boys or middleaged men, and symptoms include pruritus, new-onset phimosis, decreased sensation, painful erections, dysuria, and urinary obstruction.1-3 An estimated 97% of patients have some form of genital involvement with only 2.5% showing isolated extragenital involvement, though the latter may be underdiagnosed, as this area is more likely to be asymptomatic.3-6 Extragenital LS&A most often involves the neck and shoulders. The classic appearance of LS&A includes shiny, white-red macules and papules that ultimately coalesce into atrophic plaques and can be accompanied by fissuring or scarring, especially in the genital area.2 There is an increased risk for SCC associated with genital LS&A.1

Bullous/hemorrhagic LS&A has been described as a rare phenotype. One case report cited an increased incidence of this subtype in patients with exclusively extragenital lesions, and the authors considered blister formation to be a characteristic feature of extragenital LS&A. The pathogenesis of blister formation and hemorrhage in LS&A is not completely understood, but trauma is thought to play a role due to decreased stress tolerance from atrophic skin.4 Furthermore, distortion of blood vessel architecture in LS&A has been described with loss of the capillary network and enlargement of vessels along  the dermoepidermal junction, which also could play a role in hemorrhage. Differential diagnosis of the bullous/hemorrhagic type of LS&A includes bullous pemphigoid, bullous lichen planus, or bullous scleroderma.7 In our more exophytic hemorrhagic case, malignancies such as SCC or angiosarcoma also had to be considered. Unlike genital LS&A, extragenital LS&A including the bullous/hemorrhagic variant has not been linked to an increasedrisk for malignancy.1,5

The mainstay of treatment of all forms of LS&A is high-potency topical steroids, but topical retinoids, tacrolimus, and UVA phototherapy also have been used. Bullous/hemorrhagic lesions often resolve quickly with topical steroids, leaving behind more classic plaques in their place, which can be more refractory to treatment.5,7

The Diagnosis: Bullous/Hemorrhagic Lichen Sclerosus et Atrophicus

Histopathologic examination revealed hyperkeratosis of the stratum corneum and thinning of the epidermis (Figure). Subepidermal edema and hemorrhage in the papillary dermis were seen. There were dilated vessels beneath the edema in the reticular dermis, as well as perivascular, perifollicular, and interstitial lymphocytic inflammation. No cytologic atypia characteristic of squamous cell carcinoma (SCC) and angiosarcoma or large lymphatic channels characteristic of lymphangioma were noted. Based on clinicopathologic correlation, the diagnosis of the bullous/hemorrhagic form of lichen sclerosus et atrophicus (LS&A) was made. The patient was treated with high-potency topical steroids with notable symptomatic improvement and rapid resolution of the hemorrhagic lesion.

A, Histologic analysis showed hyperkeratosis of the stratum corneum, papillary dermal sclerosis and edema, zone of lymphoid inflammation, and copious erythrocyte extravasation (H&E, original magnification ×100). B, Copious erythrocyte extravasation also was evident in the superficial dermis (H&E, original magnification ×40).

Lichen sclerosus et atrophicus is a chronic inflammatory condition with a predilection for the anogenital region, though rare cases of extragenital involvement have been reported. It is seen in both sexes and across all age groups, with notably higher prevalence in females in the fifth and sixth decades of life.1,2 Lichen sclerosus et atrophicus can be difficult to diagnose, as these patients may present to a variety of specialists, may be embarrassed by the condition and reluctant for full evaluation, or may have asymptomatic lesions.2,3 Rare cases of isolated extragenital involvement and hemorrhagic or bullous lesions further complicate the diagnosis.1,2 Despite these difficulties, diagnosis is essential, as there is potential for cosmetically and functionally detrimental scarring as well as atrophy and development of overlying malignancies. Lichen sclerosus et atrophicus is not curable and rarely remits spontaneously, but appropriate treatment strategies can help control the symptoms of the condition as well as its most devastating sequelae.3

For females, classic LS&A is most common in theprepubertal, perimenopausal, or postmenopausal periods, commonly involving the vulva or perineum. Symptoms include pruritus, burning sensation, dysuria, dyspareunia, and labial stenosis, among others. For males, most cases involve the glans penis in prepubertal boys or middleaged men, and symptoms include pruritus, new-onset phimosis, decreased sensation, painful erections, dysuria, and urinary obstruction.1-3 An estimated 97% of patients have some form of genital involvement with only 2.5% showing isolated extragenital involvement, though the latter may be underdiagnosed, as this area is more likely to be asymptomatic.3-6 Extragenital LS&A most often involves the neck and shoulders. The classic appearance of LS&A includes shiny, white-red macules and papules that ultimately coalesce into atrophic plaques and can be accompanied by fissuring or scarring, especially in the genital area.2 There is an increased risk for SCC associated with genital LS&A.1

Bullous/hemorrhagic LS&A has been described as a rare phenotype. One case report cited an increased incidence of this subtype in patients with exclusively extragenital lesions, and the authors considered blister formation to be a characteristic feature of extragenital LS&A. The pathogenesis of blister formation and hemorrhage in LS&A is not completely understood, but trauma is thought to play a role due to decreased stress tolerance from atrophic skin.4 Furthermore, distortion of blood vessel architecture in LS&A has been described with loss of the capillary network and enlargement of vessels along  the dermoepidermal junction, which also could play a role in hemorrhage. Differential diagnosis of the bullous/hemorrhagic type of LS&A includes bullous pemphigoid, bullous lichen planus, or bullous scleroderma.7 In our more exophytic hemorrhagic case, malignancies such as SCC or angiosarcoma also had to be considered. Unlike genital LS&A, extragenital LS&A including the bullous/hemorrhagic variant has not been linked to an increasedrisk for malignancy.1,5

The mainstay of treatment of all forms of LS&A is high-potency topical steroids, but topical retinoids, tacrolimus, and UVA phototherapy also have been used. Bullous/hemorrhagic lesions often resolve quickly with topical steroids, leaving behind more classic plaques in their place, which can be more refractory to treatment.5,7

References
  1. Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. J Am Acad Dermatol. 1995;32:393-416.
  2. Pugliese JM, Morey AF, Peterson AC. Lichen sclerosus: review of the literature and current recommendations for management. J Urol. 2007;178:2268-2276.
  3. Fistarol SK, Itin PH. Diagnosis and treatment of lichen sclerosus: an update. Am J Clin Dermatol. 2013;14:27-47.
  4. Kimura A, Kambe N, Satoh T, et al. Follicular keratosis and bullous formation are typical signs of extragenital lichen sclerosus. J Dermatol. 2011;38:834-836.
  5. Khatu S, Vasani R. Isolated, localised extragenital bullous lichen sclerosus et atrophicus: a rare entity. Indian J Dermatol. 2013;58:409.
  6. Luzar B, Neil SM, Calonje E. Angiokeratoma-like changes in extragenital and genital lichen sclerosus. J Cutan Pathol. 2009;36:540-542.
  7. Lima RS, Maquine GA, Schettini AP, et al. Bullous and hemorrhagic lichen sclerosus—case report. An Bras Dermatol. 2015;90 (3 suppl 1):118-120.
References
  1. Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. J Am Acad Dermatol. 1995;32:393-416.
  2. Pugliese JM, Morey AF, Peterson AC. Lichen sclerosus: review of the literature and current recommendations for management. J Urol. 2007;178:2268-2276.
  3. Fistarol SK, Itin PH. Diagnosis and treatment of lichen sclerosus: an update. Am J Clin Dermatol. 2013;14:27-47.
  4. Kimura A, Kambe N, Satoh T, et al. Follicular keratosis and bullous formation are typical signs of extragenital lichen sclerosus. J Dermatol. 2011;38:834-836.
  5. Khatu S, Vasani R. Isolated, localised extragenital bullous lichen sclerosus et atrophicus: a rare entity. Indian J Dermatol. 2013;58:409.
  6. Luzar B, Neil SM, Calonje E. Angiokeratoma-like changes in extragenital and genital lichen sclerosus. J Cutan Pathol. 2009;36:540-542.
  7. Lima RS, Maquine GA, Schettini AP, et al. Bullous and hemorrhagic lichen sclerosus—case report. An Bras Dermatol. 2015;90 (3 suppl 1):118-120.
Issue
Cutis - 103(2)
Issue
Cutis - 103(2)
Page Number
68,79-80
Page Number
68,79-80
Publications
Publications
Topics
Article Type
Display Headline
Large Hemorrhagic Plaque With Central Crusting
Display Headline
Large Hemorrhagic Plaque With Central Crusting
Sections
Questionnaire Body

A 54-year-old woman with no notable medical history was referred to dermatology by her primary care provider for evaluation of a hematoma on the posterior neck that had developed gradually over 5 months. The lesion initially was asymptomatic but more recently had started to be painful and bleed intermittently. The patient denied any personal or family history of skin cancer. Physical examination revealed a large hemorrhagic plaque on the left side of the posterior neck with central brown-yellow crusting. There were few smaller, white, thin, sclerotic plaques with crinkling atrophy at the periphery of and inferolateral to the lesion. A punch biopsy specimen was obtained from the hemorrhagic plaque.

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 01/08/2019 - 11:00
Un-Gate On Date
Tue, 01/08/2019 - 11:00
Use ProPublica
CFC Schedule Remove Status
Tue, 01/08/2019 - 11:00
Article PDF Media

Managing Postinflammatory Hyperpigmentation in Pediatric Patients With Skin of Color

Article Type
Changed
Thu, 10/29/2020 - 14:50
Display Headline
Managing Postinflammatory Hyperpigmentation in Pediatric Patients With Skin of Color
In Collaboration With the Skin of Color Society

Postnflammatory hyperpigmentation (PIH) is an acquired hypermelanosis that can occur in children and adults following an inflammatory cutaneous disease or trauma. Postinflammatory hyperpigmentation may last for months to even years. Although PIH may occur in all skin types, it is more common and presents with greater severity and intensity in individuals with skin of color. By the year 2050, 1 in 3 US residents is projected to be Hispanic.1 It is projected that by 2044, non-Hispanic white individuals (all ages) will make up less than 50% of the US population.2 Currently, the majority of the US residents younger than 18 years are minorities. The majority minority population in the United States already exists in those younger than 18 years and is predicted to occur in the adult population by 2044.2

Effective treatment options and management strategies for PIH in adults with skin of color have been described in the literature.3 Due to a paucity of research, the approach to management of PIH in children with skin of color has been based on clinical experience and lessons learned from adult patients. This article focuses on management of PIH in pediatric patients with skin of color, which includes black/African American, African-Caribbean, Hispanic, Asian, Pacific Islander, and American Indian individuals.

Underlying Inflammatory Dermatoses Resulting in PIH

There are numerous conditions that may result in PIH, including but not limited to atopic dermatitis (AD), acne, arthropod bites, and injuries to the skin. Postinflammatory hyperpigmentation may have more of a psychological impact than the inciting disease or injury itself. The most important step in the approach to managing PIH is treating the underlying inflammatory condition that caused the pigmentation.

Parents/guardians may report a chief concern of dark spots, manchas (stains), blemishes, or stains on the skin, often with no mention of a coexisting inflammatory dermatosis. Parents/guardians of children with skin of color often have personally experienced PIH and may be determined to shield their children from similar angst associated with the condition. Although physicians may see just another pediatric patient with PIH, the child’s parents/guardians may see a condition that will be readily perceptible during major life events, such as the child’s prom or even his/her wedding day. Promptly diagnosing and instituting early treatment of inflammatory conditions associated with PIH may accelerate resolution and prevent worsening of the pigmentation.3

Select inflammatory dermatoses that are common in children with skin of color and may lead to PIH are highlighted below. Although this list is not comprehensive, the approach and management strategies should prompt creation of plans that keep PIH in mind when treating primary inflammatory skin diseases.

Atopic Dermatitis
Atopic dermatitis may induce PIH or hypopigmentation of the skin in children with skin of color. Developing a plan for AD flare prevention, as well as management of mild, moderate, and severe AD flares, is imperative in pediatric patients. Prevention plans should include gentle skin care, twice-daily application of emollients to the full body, and reduction of Staphylococcus aureus loads on the skin. The treatment action plan for mild to moderate flares may include topical corticosteroids, immunomodulators, and nonsteroidal agents. Treatment options for severe AD or patients who were unsuccessfully treated with other therapies may include phototherapy, biologics, and methotrexate, among others.4 Creating action plans for AD flares is a vital step in the prevention of PIH in patients with skin of color. Additionally, PIH should not be considered a sign of AD treatment failure.

 

 

Acne
Acne is a common skin disorder seen in patients with skin of color.5 A prospective observational study found that 34.3% of 683 children aged 9 to 14 years in a pediatric ambulatory clinic had acne.6 The number of preadolescents with acne is growing. Most cases are not associated with underlying endocrinopathy.7 With the growing population of children with skin of color in the United States along with the increasing childhood acne rate and subsequent inherent risk for hyperpigmentation, early acne interventions should be considered in pediatric acne patients with skin of color to reduce the impact of PIH in those at risk.

In a survey study of 313 adult acne patients with skin of color, 37.2% reported the presence of dark marks lasting 4 months or longer.5 Regardless of the severity of the acne, treatment should be initiated as tolerated in those with PIH. Adolescent acne patients with skin of color may develop PIH that is more severe and longer lasting than the acne itself.

The foundation for treatment of acne in adolescent skin of color patients is the same as those without skin of color, including topical retinoids, topical antibiotics, oral antibiotics, and isotretinoin when needed. Topical tretinoin, adapalene, azelaic acid, and tazarotene not only treat acne but also are a valuable part of the treatment armamentarium for PIH. Several studies in adults with skin of color have demonstrated improvement of PIH from the use of topical retinoids alone.8-10 Despite wanting to treat the acne aggressively, special guidance should be given to prevent retinoid dermatitis, which may lead to PIH.10 Demonstrating the application of the topical acne medications, discussing how to avoid potential side effects, and giving permission to skip applications, if needed, may empower families to make adjustments between visits to limit irritation that might prompt further PIH. Incorporating α-hydroxy acid–based cleansers, α-hydroxy acid–based chemical peels, or salicylic acid chemical peels may be warranted in the setting of intense PIH. Selecting treatments that not only help the inflammatory disease leading to the PIH but also can help improve the pigmentation are preferred; however, the risks and benefits have to be weighed because many treatments that work well for PIH also may cause irritation, leading to new or worsening PIH.

Arthropod Bites
Arthropod bites cause inflamed pruritic papules and nodules, and the resulting PIH in those with darker skin types may be quite dramatic. Parents/guardians should be instructed to have a low-potency topical corticosteroid on hand to use on bites for a few days when they appear, which will not only help with the inflammation associated with the bite but also will help decrease pruritus and subsequently skin injury from scratching. In homes with pets, checking animals routinely for fleas and other infestations is helpful. In the setting of repeated arthropod bites in the spring and summer, applying bug repellant with 10% to 30% DEET (N,N-diethyl-meta-toluamide) on the child’s clothing and exposed body areas before playing outside or in the morning before school or camp may prevent some bites. There are DEET alternatives, such as picaridin, that may be used. Product instructions should be followed when using insect repellants in the pediatric population.11

PIH Management Strategies

Gentle Skin Care Routine
There are misconceptions that areas of hyperpigmentation on the skin are caused by dirt and that scrubbing the skin harder may help to lighten the affected areas. Parents/guardians may report that the child’s skin looks dirty or, in the setting of acne, view dirt as the cause of the skin condition, which may prompt the patient to scrub the skin and the friction further worsens the PIH. Use of daily gentle cleansers and moisturizers is advised to keep the skin moisturized and free of further potential irritation and dryness that may prompt scratching or flares of the underlying condition.

Photoprotection
During the treatment course for PIH, using sun protection is helpful to prevent further darkening of the PIH areas. Sun protection may be in the form of broad-spectrum sunscreen, hats, or sun-protective clothing. Patients should be encouraged to apply sunscreen daily and to reapply every 2 hours and after water-based activities.12 For pediatric and adolescent populations, practicing sun-protective behaviors before school or outdoor activities also should be advised, as many families only think about sun protection in the setting of sunny vacation activities. Research has demonstrated that individuals with skin of color may not realize that they can be affected by skin cancer,13 thus they may not have any experience selecting, applying, or regularly using sunscreens. Products that do not leave a white hue on the skin are suggested for adolescents who may be sensitive about their appearance following sunscreen application.

 

 

Skin Lightening Treatments

Although the most important therapy for PIH is to treat the underlying inflammatory conditions, some parents/guardians may desire additional options due to the extent of involvement of the PIH, its psychological impact on the child, or adverse effect on the child’s quality of life.14 In adolescents, incorporating an α-hydroxy acid–based cleanser, glycolic acid chemical peels, salicylic acid chemical peels, and topical cosmeceuticals may be warranted in the setting of intense PIH and acne. However, irritation may lead to further dyspigmentation.

Topical ammonium lactate 12% is lactic acid neutralized with ammonium hydroxide that is formulated as a lotion or a cream. It is used to hydrate dry skin and may decrease corneocyte cohesion.15 Topical ammonium lactate also has been used anecdotally for PIH on the body during periods of watchful waiting.

Topical hydroquinone, the gold standard for treating hyperpigmentation,3,16 is not approved in children, but some parents/guardians elect to utilize hydroquinone off label to accelerate the clearing of distressing PIH in adolescents. Careful consideration including a discussion of potential risks and alternatives (eg, watchful waiting) should be highlighted.

In the setting of chronic inflammatory conditions that recur and remit, potentially irritating topical treatments should be used only during periods when symptoms of inflammation such as itching or erythema are absent.

Conclusion

Despite the best management efforts, PIH in some patients with skin of color may be present for months to years. In the pediatric skin of color population, treatment of the underlying inflammatory condition, gentle skin care, use of photoprotection, and time may be all that is needed for PIH resolution. With their parent/guardians’ consent, adolescents distressed by PIH may decide to pursue more aggressive, potentially irritating treatments. Above all, the most important management in the setting of PIH is to treat the underlying inflammatory condition causing the PIH and set reasonable expectations. For challenging cases, pediatric dermatologists with special expertise in treating pediatric and adolescent patients with skin of color may be consulted.

References
  1. Broughton A. Minorities expected to be majority in 2050. CNN. August 13, 2008.  Accessed January 2, 2019.
  2. Colby SL, Ortman JM. Projections of the Size and Composition of the US Population: 2014 to 2060. Washington, DC: US Census Bureau; 2014. Current Population Reports, P25-1143. Published March 2015. Accessed January 23, 2019.
  3. Davis EC, Callender VD. Postinflammatory hyperpigmentation: a review of the epidemiology, clinical features, and treatment options in skin of color. J Clin Aesthet Dermatol2010;3:20-31.
  4. Eichenfield LF, Ahluwalia J, Waldman A, et al. Current guidelines for the evaluation and management of atopic dermatitis: a comparison of the Joint Task Force Practice Parameter and American Academy of Dermatology guidelines. J Allergy Clin Immunol. 2017;139(4S):S49-S57.
  5. Taylor SC, Cook-Bolden F, Rahman Z, et al. Acne vulgaris in skin of color. J Am Acad Dermatol. 2002;46(2 suppl):S98-S106.
  6. Napolitano M, Ruggiero G, Monfrecola G, et al. Acne prevalence in 9 to 14-year-old patients attending pediatric ambulatory clinics in Italy. Int J Dermatol. 2018;57:1320-1323.
  7. Mancini AJ, Baldwin HE, Eichenfield LF. Acne life cycle: the spectrum of pediatric disease. Semin Cutan Med Surg 2011;30:2-5.
  8. Lowe NJ, Rizk D, Grimes P, et al. Azelaic acid 20% cream in the treatment of facial hyperpigmentation in darker-skinned patients. Clin Ther. 1998;20:945-959.
  9. Grimes P, Callender V. Tazarotene cream for postinflammatory hyperpigmentation and acne vulgaris in darker skin: a double-blind, randomized, vehicle-controlled study. Cutis2006;77:45-50.
  10. Bulengo-Ransby SM, Griffiths CE, Kimbrough-Green CK, et al. Topical tretinoin (retinoid acid) therapy for hyperpigmented lesions caused by inflammation of the skin in black patients. N Engl J Med. 1993;328:1438-1443.
  11. American Academy of Pediatrics. Choosing an insect repellent for your child. Healthy Children website. Updated July 18, 2018. Accessed January 8, 2019.
  12. Agbai ON, Buster K, Sanchez M, et al. Skin cancer and photoprotection in people of color: a review and recommendations for physicians and the public. J Am Acad Dermatol. 2014;70:312-317.
  13. Buster KJ, You Z, Fouad M, et al. Skin cancer risk perceptions: a comparison across ethnicity, age, education, gender, and income. J Am Acad Dermatol. 2012;66:771-779.
  14. Downie J. Help prevent and reverse post-inflammatory hyperpigmentation. Pract Dermatol Pediatr. May/June 2011:12-14. Accessed January 18, 2019.
  15. Ammonium lactate lotion 12% [package insert]. Bronx, New York: Perrigo New York, Inc; 2006.
  16. Grimes PE. Management of hyperpigmentation in darker racial ethnic groups. Semin Cutan Med Surg. 2009;28:77-85.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Lewis Katz School of Medicine, Temple University Hospital, Philadelphia, Pennsylvania.

Dr. Heath is a consultant for Unilever, a former advisory board member and speaker for Pfizer Inc, and owner of Heath Health.

Correspondence: Candrice R. Heath, MD, 1316 W Ontario St, Jones Hall, Philadelphia, PA 19140 ([email protected]).

Issue
Cutis - 103(2)
Publications
Topics
Page Number
71-73
Sections
Author and Disclosure Information

From the Department of Dermatology, Lewis Katz School of Medicine, Temple University Hospital, Philadelphia, Pennsylvania.

Dr. Heath is a consultant for Unilever, a former advisory board member and speaker for Pfizer Inc, and owner of Heath Health.

Correspondence: Candrice R. Heath, MD, 1316 W Ontario St, Jones Hall, Philadelphia, PA 19140 ([email protected]).

Author and Disclosure Information

From the Department of Dermatology, Lewis Katz School of Medicine, Temple University Hospital, Philadelphia, Pennsylvania.

Dr. Heath is a consultant for Unilever, a former advisory board member and speaker for Pfizer Inc, and owner of Heath Health.

Correspondence: Candrice R. Heath, MD, 1316 W Ontario St, Jones Hall, Philadelphia, PA 19140 ([email protected]).

Article PDF
Article PDF
In Collaboration With the Skin of Color Society
In Collaboration With the Skin of Color Society

Postnflammatory hyperpigmentation (PIH) is an acquired hypermelanosis that can occur in children and adults following an inflammatory cutaneous disease or trauma. Postinflammatory hyperpigmentation may last for months to even years. Although PIH may occur in all skin types, it is more common and presents with greater severity and intensity in individuals with skin of color. By the year 2050, 1 in 3 US residents is projected to be Hispanic.1 It is projected that by 2044, non-Hispanic white individuals (all ages) will make up less than 50% of the US population.2 Currently, the majority of the US residents younger than 18 years are minorities. The majority minority population in the United States already exists in those younger than 18 years and is predicted to occur in the adult population by 2044.2

Effective treatment options and management strategies for PIH in adults with skin of color have been described in the literature.3 Due to a paucity of research, the approach to management of PIH in children with skin of color has been based on clinical experience and lessons learned from adult patients. This article focuses on management of PIH in pediatric patients with skin of color, which includes black/African American, African-Caribbean, Hispanic, Asian, Pacific Islander, and American Indian individuals.

Underlying Inflammatory Dermatoses Resulting in PIH

There are numerous conditions that may result in PIH, including but not limited to atopic dermatitis (AD), acne, arthropod bites, and injuries to the skin. Postinflammatory hyperpigmentation may have more of a psychological impact than the inciting disease or injury itself. The most important step in the approach to managing PIH is treating the underlying inflammatory condition that caused the pigmentation.

Parents/guardians may report a chief concern of dark spots, manchas (stains), blemishes, or stains on the skin, often with no mention of a coexisting inflammatory dermatosis. Parents/guardians of children with skin of color often have personally experienced PIH and may be determined to shield their children from similar angst associated with the condition. Although physicians may see just another pediatric patient with PIH, the child’s parents/guardians may see a condition that will be readily perceptible during major life events, such as the child’s prom or even his/her wedding day. Promptly diagnosing and instituting early treatment of inflammatory conditions associated with PIH may accelerate resolution and prevent worsening of the pigmentation.3

Select inflammatory dermatoses that are common in children with skin of color and may lead to PIH are highlighted below. Although this list is not comprehensive, the approach and management strategies should prompt creation of plans that keep PIH in mind when treating primary inflammatory skin diseases.

Atopic Dermatitis
Atopic dermatitis may induce PIH or hypopigmentation of the skin in children with skin of color. Developing a plan for AD flare prevention, as well as management of mild, moderate, and severe AD flares, is imperative in pediatric patients. Prevention plans should include gentle skin care, twice-daily application of emollients to the full body, and reduction of Staphylococcus aureus loads on the skin. The treatment action plan for mild to moderate flares may include topical corticosteroids, immunomodulators, and nonsteroidal agents. Treatment options for severe AD or patients who were unsuccessfully treated with other therapies may include phototherapy, biologics, and methotrexate, among others.4 Creating action plans for AD flares is a vital step in the prevention of PIH in patients with skin of color. Additionally, PIH should not be considered a sign of AD treatment failure.

 

 

Acne
Acne is a common skin disorder seen in patients with skin of color.5 A prospective observational study found that 34.3% of 683 children aged 9 to 14 years in a pediatric ambulatory clinic had acne.6 The number of preadolescents with acne is growing. Most cases are not associated with underlying endocrinopathy.7 With the growing population of children with skin of color in the United States along with the increasing childhood acne rate and subsequent inherent risk for hyperpigmentation, early acne interventions should be considered in pediatric acne patients with skin of color to reduce the impact of PIH in those at risk.

In a survey study of 313 adult acne patients with skin of color, 37.2% reported the presence of dark marks lasting 4 months or longer.5 Regardless of the severity of the acne, treatment should be initiated as tolerated in those with PIH. Adolescent acne patients with skin of color may develop PIH that is more severe and longer lasting than the acne itself.

The foundation for treatment of acne in adolescent skin of color patients is the same as those without skin of color, including topical retinoids, topical antibiotics, oral antibiotics, and isotretinoin when needed. Topical tretinoin, adapalene, azelaic acid, and tazarotene not only treat acne but also are a valuable part of the treatment armamentarium for PIH. Several studies in adults with skin of color have demonstrated improvement of PIH from the use of topical retinoids alone.8-10 Despite wanting to treat the acne aggressively, special guidance should be given to prevent retinoid dermatitis, which may lead to PIH.10 Demonstrating the application of the topical acne medications, discussing how to avoid potential side effects, and giving permission to skip applications, if needed, may empower families to make adjustments between visits to limit irritation that might prompt further PIH. Incorporating α-hydroxy acid–based cleansers, α-hydroxy acid–based chemical peels, or salicylic acid chemical peels may be warranted in the setting of intense PIH. Selecting treatments that not only help the inflammatory disease leading to the PIH but also can help improve the pigmentation are preferred; however, the risks and benefits have to be weighed because many treatments that work well for PIH also may cause irritation, leading to new or worsening PIH.

Arthropod Bites
Arthropod bites cause inflamed pruritic papules and nodules, and the resulting PIH in those with darker skin types may be quite dramatic. Parents/guardians should be instructed to have a low-potency topical corticosteroid on hand to use on bites for a few days when they appear, which will not only help with the inflammation associated with the bite but also will help decrease pruritus and subsequently skin injury from scratching. In homes with pets, checking animals routinely for fleas and other infestations is helpful. In the setting of repeated arthropod bites in the spring and summer, applying bug repellant with 10% to 30% DEET (N,N-diethyl-meta-toluamide) on the child’s clothing and exposed body areas before playing outside or in the morning before school or camp may prevent some bites. There are DEET alternatives, such as picaridin, that may be used. Product instructions should be followed when using insect repellants in the pediatric population.11

PIH Management Strategies

Gentle Skin Care Routine
There are misconceptions that areas of hyperpigmentation on the skin are caused by dirt and that scrubbing the skin harder may help to lighten the affected areas. Parents/guardians may report that the child’s skin looks dirty or, in the setting of acne, view dirt as the cause of the skin condition, which may prompt the patient to scrub the skin and the friction further worsens the PIH. Use of daily gentle cleansers and moisturizers is advised to keep the skin moisturized and free of further potential irritation and dryness that may prompt scratching or flares of the underlying condition.

Photoprotection
During the treatment course for PIH, using sun protection is helpful to prevent further darkening of the PIH areas. Sun protection may be in the form of broad-spectrum sunscreen, hats, or sun-protective clothing. Patients should be encouraged to apply sunscreen daily and to reapply every 2 hours and after water-based activities.12 For pediatric and adolescent populations, practicing sun-protective behaviors before school or outdoor activities also should be advised, as many families only think about sun protection in the setting of sunny vacation activities. Research has demonstrated that individuals with skin of color may not realize that they can be affected by skin cancer,13 thus they may not have any experience selecting, applying, or regularly using sunscreens. Products that do not leave a white hue on the skin are suggested for adolescents who may be sensitive about their appearance following sunscreen application.

 

 

Skin Lightening Treatments

Although the most important therapy for PIH is to treat the underlying inflammatory conditions, some parents/guardians may desire additional options due to the extent of involvement of the PIH, its psychological impact on the child, or adverse effect on the child’s quality of life.14 In adolescents, incorporating an α-hydroxy acid–based cleanser, glycolic acid chemical peels, salicylic acid chemical peels, and topical cosmeceuticals may be warranted in the setting of intense PIH and acne. However, irritation may lead to further dyspigmentation.

Topical ammonium lactate 12% is lactic acid neutralized with ammonium hydroxide that is formulated as a lotion or a cream. It is used to hydrate dry skin and may decrease corneocyte cohesion.15 Topical ammonium lactate also has been used anecdotally for PIH on the body during periods of watchful waiting.

Topical hydroquinone, the gold standard for treating hyperpigmentation,3,16 is not approved in children, but some parents/guardians elect to utilize hydroquinone off label to accelerate the clearing of distressing PIH in adolescents. Careful consideration including a discussion of potential risks and alternatives (eg, watchful waiting) should be highlighted.

In the setting of chronic inflammatory conditions that recur and remit, potentially irritating topical treatments should be used only during periods when symptoms of inflammation such as itching or erythema are absent.

Conclusion

Despite the best management efforts, PIH in some patients with skin of color may be present for months to years. In the pediatric skin of color population, treatment of the underlying inflammatory condition, gentle skin care, use of photoprotection, and time may be all that is needed for PIH resolution. With their parent/guardians’ consent, adolescents distressed by PIH may decide to pursue more aggressive, potentially irritating treatments. Above all, the most important management in the setting of PIH is to treat the underlying inflammatory condition causing the PIH and set reasonable expectations. For challenging cases, pediatric dermatologists with special expertise in treating pediatric and adolescent patients with skin of color may be consulted.

Postnflammatory hyperpigmentation (PIH) is an acquired hypermelanosis that can occur in children and adults following an inflammatory cutaneous disease or trauma. Postinflammatory hyperpigmentation may last for months to even years. Although PIH may occur in all skin types, it is more common and presents with greater severity and intensity in individuals with skin of color. By the year 2050, 1 in 3 US residents is projected to be Hispanic.1 It is projected that by 2044, non-Hispanic white individuals (all ages) will make up less than 50% of the US population.2 Currently, the majority of the US residents younger than 18 years are minorities. The majority minority population in the United States already exists in those younger than 18 years and is predicted to occur in the adult population by 2044.2

Effective treatment options and management strategies for PIH in adults with skin of color have been described in the literature.3 Due to a paucity of research, the approach to management of PIH in children with skin of color has been based on clinical experience and lessons learned from adult patients. This article focuses on management of PIH in pediatric patients with skin of color, which includes black/African American, African-Caribbean, Hispanic, Asian, Pacific Islander, and American Indian individuals.

Underlying Inflammatory Dermatoses Resulting in PIH

There are numerous conditions that may result in PIH, including but not limited to atopic dermatitis (AD), acne, arthropod bites, and injuries to the skin. Postinflammatory hyperpigmentation may have more of a psychological impact than the inciting disease or injury itself. The most important step in the approach to managing PIH is treating the underlying inflammatory condition that caused the pigmentation.

Parents/guardians may report a chief concern of dark spots, manchas (stains), blemishes, or stains on the skin, often with no mention of a coexisting inflammatory dermatosis. Parents/guardians of children with skin of color often have personally experienced PIH and may be determined to shield their children from similar angst associated with the condition. Although physicians may see just another pediatric patient with PIH, the child’s parents/guardians may see a condition that will be readily perceptible during major life events, such as the child’s prom or even his/her wedding day. Promptly diagnosing and instituting early treatment of inflammatory conditions associated with PIH may accelerate resolution and prevent worsening of the pigmentation.3

Select inflammatory dermatoses that are common in children with skin of color and may lead to PIH are highlighted below. Although this list is not comprehensive, the approach and management strategies should prompt creation of plans that keep PIH in mind when treating primary inflammatory skin diseases.

Atopic Dermatitis
Atopic dermatitis may induce PIH or hypopigmentation of the skin in children with skin of color. Developing a plan for AD flare prevention, as well as management of mild, moderate, and severe AD flares, is imperative in pediatric patients. Prevention plans should include gentle skin care, twice-daily application of emollients to the full body, and reduction of Staphylococcus aureus loads on the skin. The treatment action plan for mild to moderate flares may include topical corticosteroids, immunomodulators, and nonsteroidal agents. Treatment options for severe AD or patients who were unsuccessfully treated with other therapies may include phototherapy, biologics, and methotrexate, among others.4 Creating action plans for AD flares is a vital step in the prevention of PIH in patients with skin of color. Additionally, PIH should not be considered a sign of AD treatment failure.

 

 

Acne
Acne is a common skin disorder seen in patients with skin of color.5 A prospective observational study found that 34.3% of 683 children aged 9 to 14 years in a pediatric ambulatory clinic had acne.6 The number of preadolescents with acne is growing. Most cases are not associated with underlying endocrinopathy.7 With the growing population of children with skin of color in the United States along with the increasing childhood acne rate and subsequent inherent risk for hyperpigmentation, early acne interventions should be considered in pediatric acne patients with skin of color to reduce the impact of PIH in those at risk.

In a survey study of 313 adult acne patients with skin of color, 37.2% reported the presence of dark marks lasting 4 months or longer.5 Regardless of the severity of the acne, treatment should be initiated as tolerated in those with PIH. Adolescent acne patients with skin of color may develop PIH that is more severe and longer lasting than the acne itself.

The foundation for treatment of acne in adolescent skin of color patients is the same as those without skin of color, including topical retinoids, topical antibiotics, oral antibiotics, and isotretinoin when needed. Topical tretinoin, adapalene, azelaic acid, and tazarotene not only treat acne but also are a valuable part of the treatment armamentarium for PIH. Several studies in adults with skin of color have demonstrated improvement of PIH from the use of topical retinoids alone.8-10 Despite wanting to treat the acne aggressively, special guidance should be given to prevent retinoid dermatitis, which may lead to PIH.10 Demonstrating the application of the topical acne medications, discussing how to avoid potential side effects, and giving permission to skip applications, if needed, may empower families to make adjustments between visits to limit irritation that might prompt further PIH. Incorporating α-hydroxy acid–based cleansers, α-hydroxy acid–based chemical peels, or salicylic acid chemical peels may be warranted in the setting of intense PIH. Selecting treatments that not only help the inflammatory disease leading to the PIH but also can help improve the pigmentation are preferred; however, the risks and benefits have to be weighed because many treatments that work well for PIH also may cause irritation, leading to new or worsening PIH.

Arthropod Bites
Arthropod bites cause inflamed pruritic papules and nodules, and the resulting PIH in those with darker skin types may be quite dramatic. Parents/guardians should be instructed to have a low-potency topical corticosteroid on hand to use on bites for a few days when they appear, which will not only help with the inflammation associated with the bite but also will help decrease pruritus and subsequently skin injury from scratching. In homes with pets, checking animals routinely for fleas and other infestations is helpful. In the setting of repeated arthropod bites in the spring and summer, applying bug repellant with 10% to 30% DEET (N,N-diethyl-meta-toluamide) on the child’s clothing and exposed body areas before playing outside or in the morning before school or camp may prevent some bites. There are DEET alternatives, such as picaridin, that may be used. Product instructions should be followed when using insect repellants in the pediatric population.11

PIH Management Strategies

Gentle Skin Care Routine
There are misconceptions that areas of hyperpigmentation on the skin are caused by dirt and that scrubbing the skin harder may help to lighten the affected areas. Parents/guardians may report that the child’s skin looks dirty or, in the setting of acne, view dirt as the cause of the skin condition, which may prompt the patient to scrub the skin and the friction further worsens the PIH. Use of daily gentle cleansers and moisturizers is advised to keep the skin moisturized and free of further potential irritation and dryness that may prompt scratching or flares of the underlying condition.

Photoprotection
During the treatment course for PIH, using sun protection is helpful to prevent further darkening of the PIH areas. Sun protection may be in the form of broad-spectrum sunscreen, hats, or sun-protective clothing. Patients should be encouraged to apply sunscreen daily and to reapply every 2 hours and after water-based activities.12 For pediatric and adolescent populations, practicing sun-protective behaviors before school or outdoor activities also should be advised, as many families only think about sun protection in the setting of sunny vacation activities. Research has demonstrated that individuals with skin of color may not realize that they can be affected by skin cancer,13 thus they may not have any experience selecting, applying, or regularly using sunscreens. Products that do not leave a white hue on the skin are suggested for adolescents who may be sensitive about their appearance following sunscreen application.

 

 

Skin Lightening Treatments

Although the most important therapy for PIH is to treat the underlying inflammatory conditions, some parents/guardians may desire additional options due to the extent of involvement of the PIH, its psychological impact on the child, or adverse effect on the child’s quality of life.14 In adolescents, incorporating an α-hydroxy acid–based cleanser, glycolic acid chemical peels, salicylic acid chemical peels, and topical cosmeceuticals may be warranted in the setting of intense PIH and acne. However, irritation may lead to further dyspigmentation.

Topical ammonium lactate 12% is lactic acid neutralized with ammonium hydroxide that is formulated as a lotion or a cream. It is used to hydrate dry skin and may decrease corneocyte cohesion.15 Topical ammonium lactate also has been used anecdotally for PIH on the body during periods of watchful waiting.

Topical hydroquinone, the gold standard for treating hyperpigmentation,3,16 is not approved in children, but some parents/guardians elect to utilize hydroquinone off label to accelerate the clearing of distressing PIH in adolescents. Careful consideration including a discussion of potential risks and alternatives (eg, watchful waiting) should be highlighted.

In the setting of chronic inflammatory conditions that recur and remit, potentially irritating topical treatments should be used only during periods when symptoms of inflammation such as itching or erythema are absent.

Conclusion

Despite the best management efforts, PIH in some patients with skin of color may be present for months to years. In the pediatric skin of color population, treatment of the underlying inflammatory condition, gentle skin care, use of photoprotection, and time may be all that is needed for PIH resolution. With their parent/guardians’ consent, adolescents distressed by PIH may decide to pursue more aggressive, potentially irritating treatments. Above all, the most important management in the setting of PIH is to treat the underlying inflammatory condition causing the PIH and set reasonable expectations. For challenging cases, pediatric dermatologists with special expertise in treating pediatric and adolescent patients with skin of color may be consulted.

References
  1. Broughton A. Minorities expected to be majority in 2050. CNN. August 13, 2008.  Accessed January 2, 2019.
  2. Colby SL, Ortman JM. Projections of the Size and Composition of the US Population: 2014 to 2060. Washington, DC: US Census Bureau; 2014. Current Population Reports, P25-1143. Published March 2015. Accessed January 23, 2019.
  3. Davis EC, Callender VD. Postinflammatory hyperpigmentation: a review of the epidemiology, clinical features, and treatment options in skin of color. J Clin Aesthet Dermatol2010;3:20-31.
  4. Eichenfield LF, Ahluwalia J, Waldman A, et al. Current guidelines for the evaluation and management of atopic dermatitis: a comparison of the Joint Task Force Practice Parameter and American Academy of Dermatology guidelines. J Allergy Clin Immunol. 2017;139(4S):S49-S57.
  5. Taylor SC, Cook-Bolden F, Rahman Z, et al. Acne vulgaris in skin of color. J Am Acad Dermatol. 2002;46(2 suppl):S98-S106.
  6. Napolitano M, Ruggiero G, Monfrecola G, et al. Acne prevalence in 9 to 14-year-old patients attending pediatric ambulatory clinics in Italy. Int J Dermatol. 2018;57:1320-1323.
  7. Mancini AJ, Baldwin HE, Eichenfield LF. Acne life cycle: the spectrum of pediatric disease. Semin Cutan Med Surg 2011;30:2-5.
  8. Lowe NJ, Rizk D, Grimes P, et al. Azelaic acid 20% cream in the treatment of facial hyperpigmentation in darker-skinned patients. Clin Ther. 1998;20:945-959.
  9. Grimes P, Callender V. Tazarotene cream for postinflammatory hyperpigmentation and acne vulgaris in darker skin: a double-blind, randomized, vehicle-controlled study. Cutis2006;77:45-50.
  10. Bulengo-Ransby SM, Griffiths CE, Kimbrough-Green CK, et al. Topical tretinoin (retinoid acid) therapy for hyperpigmented lesions caused by inflammation of the skin in black patients. N Engl J Med. 1993;328:1438-1443.
  11. American Academy of Pediatrics. Choosing an insect repellent for your child. Healthy Children website. Updated July 18, 2018. Accessed January 8, 2019.
  12. Agbai ON, Buster K, Sanchez M, et al. Skin cancer and photoprotection in people of color: a review and recommendations for physicians and the public. J Am Acad Dermatol. 2014;70:312-317.
  13. Buster KJ, You Z, Fouad M, et al. Skin cancer risk perceptions: a comparison across ethnicity, age, education, gender, and income. J Am Acad Dermatol. 2012;66:771-779.
  14. Downie J. Help prevent and reverse post-inflammatory hyperpigmentation. Pract Dermatol Pediatr. May/June 2011:12-14. Accessed January 18, 2019.
  15. Ammonium lactate lotion 12% [package insert]. Bronx, New York: Perrigo New York, Inc; 2006.
  16. Grimes PE. Management of hyperpigmentation in darker racial ethnic groups. Semin Cutan Med Surg. 2009;28:77-85.
References
  1. Broughton A. Minorities expected to be majority in 2050. CNN. August 13, 2008.  Accessed January 2, 2019.
  2. Colby SL, Ortman JM. Projections of the Size and Composition of the US Population: 2014 to 2060. Washington, DC: US Census Bureau; 2014. Current Population Reports, P25-1143. Published March 2015. Accessed January 23, 2019.
  3. Davis EC, Callender VD. Postinflammatory hyperpigmentation: a review of the epidemiology, clinical features, and treatment options in skin of color. J Clin Aesthet Dermatol2010;3:20-31.
  4. Eichenfield LF, Ahluwalia J, Waldman A, et al. Current guidelines for the evaluation and management of atopic dermatitis: a comparison of the Joint Task Force Practice Parameter and American Academy of Dermatology guidelines. J Allergy Clin Immunol. 2017;139(4S):S49-S57.
  5. Taylor SC, Cook-Bolden F, Rahman Z, et al. Acne vulgaris in skin of color. J Am Acad Dermatol. 2002;46(2 suppl):S98-S106.
  6. Napolitano M, Ruggiero G, Monfrecola G, et al. Acne prevalence in 9 to 14-year-old patients attending pediatric ambulatory clinics in Italy. Int J Dermatol. 2018;57:1320-1323.
  7. Mancini AJ, Baldwin HE, Eichenfield LF. Acne life cycle: the spectrum of pediatric disease. Semin Cutan Med Surg 2011;30:2-5.
  8. Lowe NJ, Rizk D, Grimes P, et al. Azelaic acid 20% cream in the treatment of facial hyperpigmentation in darker-skinned patients. Clin Ther. 1998;20:945-959.
  9. Grimes P, Callender V. Tazarotene cream for postinflammatory hyperpigmentation and acne vulgaris in darker skin: a double-blind, randomized, vehicle-controlled study. Cutis2006;77:45-50.
  10. Bulengo-Ransby SM, Griffiths CE, Kimbrough-Green CK, et al. Topical tretinoin (retinoid acid) therapy for hyperpigmented lesions caused by inflammation of the skin in black patients. N Engl J Med. 1993;328:1438-1443.
  11. American Academy of Pediatrics. Choosing an insect repellent for your child. Healthy Children website. Updated July 18, 2018. Accessed January 8, 2019.
  12. Agbai ON, Buster K, Sanchez M, et al. Skin cancer and photoprotection in people of color: a review and recommendations for physicians and the public. J Am Acad Dermatol. 2014;70:312-317.
  13. Buster KJ, You Z, Fouad M, et al. Skin cancer risk perceptions: a comparison across ethnicity, age, education, gender, and income. J Am Acad Dermatol. 2012;66:771-779.
  14. Downie J. Help prevent and reverse post-inflammatory hyperpigmentation. Pract Dermatol Pediatr. May/June 2011:12-14. Accessed January 18, 2019.
  15. Ammonium lactate lotion 12% [package insert]. Bronx, New York: Perrigo New York, Inc; 2006.
  16. Grimes PE. Management of hyperpigmentation in darker racial ethnic groups. Semin Cutan Med Surg. 2009;28:77-85.
Issue
Cutis - 103(2)
Issue
Cutis - 103(2)
Page Number
71-73
Page Number
71-73
Publications
Publications
Topics
Article Type
Display Headline
Managing Postinflammatory Hyperpigmentation in Pediatric Patients With Skin of Color
Display Headline
Managing Postinflammatory Hyperpigmentation in Pediatric Patients With Skin of Color
Sections
Inside the Article

Practice Points

  • The US population of children with skin of color is growing rapidly.
  • Treating the underlying inflammatory dermatosis is the most important step in managing postinflammatory hyperpigmentation (PIH); however, many pediatric PIH patients and their parents/guardians presenting with a chief concern of pigmentary changes are unaware of the associated inflammatory condition.
  • When appropriate, choose treatments for the underlying inflammatory condition that can simultaneously improve any existing PIH. Gentle skin care, avoidance of rubbing and scrubbing the skin, and photoprotection are essential to halt worsening of PIH.
  • Patients’ parents/guardians may consent to more aggressive PIH treatment in select cases (eg, emotional distress in adolescents).
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
Article PDF Media

Safety and Efficacy of Halobetasol Propionate Lotion 0.01% in the Treatment of Moderate to Severe Plaque Psoriasis: A Pooled Analysis of 2 Phase 3 Studies

Article Type
Changed
Thu, 12/15/2022 - 14:42
Display Headline
Safety and Efficacy of Halobetasol Propionate Lotion 0.01% in the Treatment of Moderate to Severe Plaque Psoriasis: A Pooled Analysis of 2 Phase 3 Studies

Psoriasis is a chronic, immune-mediated, inflammatory disease affecting almost 2% of the population.1-3 It is characterized by patches of raised reddish skin covered by silvery-white scales. Most patients have limited disease (<5% body surface area [BSA] involvement) that can be managed with topical agents.4 Topical corticosteroids (TCSs) are considered first-line therapy for mild to moderate disease because of the inflammatory nature of the condition and often are used in conjunction with systemic agents in more severe psoriasis.4

As many as 20% to 30% of patients with moderate to severe plaque psoriasis have inadequate disease control.5 Several factors may affect patient outcomes; however, drug selection and patient adherence are important given the chronic nature of the disease. A survey of 1200 patients with psoriasis reported nonadherence rates of 73% with topical therapy.6 In addition, patients tend to apply less than the recommended dose or abandon treatment altogether if rapid improvement does not occur7,8; it is not uncommon for patients with psoriasis to mistakenly believe treatment will improve their condition within 1 to 2 weeks.9 Patient satisfaction with topical treatments is low, partly because of these false expectations and formulation issues. Treatments can be greasy and sticky, with unpleasant odors and the potential to stain clothes and linens.7,10 Safety concerns with TCSs also limit their consecutive use beyond 2 to 4 weeks, which is not ideal for a disease that requires a long-term management strategy.

A potent/superpotent TCS that is administered once daily and has a safety profile that affords longer-term, once-daily treatment in an aesthetically pleasing formulation would seem ideal. Herein, we investigate the safety and tolerability of a novel low-concentration (0.01%) lotion formulation of halobetasol propionate (HP), reporting on the pooled data from 2 phase 3 clinical studies in participants with moderate to severe psoriasis.

METHODS
Study Design

We conducted 2 multicenter, double-blind, randomized, parallel-group phase 3 studies to assess the safety, tolerability, and efficacy of HP lotion 0.01% in participants with a clinical diagnosis of moderate to severe psoriasis with an investigator global assessment (IGA) score of 3 or 4 and an affected BSA of 3% to 12%. Participants were randomized (2:1) to receive HP lotion or vehicle applied topically to the affected area once daily for 8 weeks.

 

 

Inclusion and Exclusion Criteria
The studies included individuals of either sex aged 18 years or older. A target lesion was defined primarily to assess signs of psoriasis, measuring 16 to 100 cm2, with a score of 3 (moderate) or higher for 2 of 3 different psoriasis signs—erythema, plaque elevation, and scaling—and summed score of 8 or higher, with no sign scoring less than 2. Participants who had pustular psoriasis or used phototherapy, photochemotherapy, or systemic psoriasis therapy within the prior 4 weeks or biologics within the prior 3 months, or those who were diagnosed with skin conditions that would interfere with the interpretation of results were excluded from the studies.

Study Oversight
Participants provided written informed consent before study-related procedures were performed, and the protocol and consent were approved by institutional review boards or ethics committees at all investigational sites. The study was conducted in accordance with the principles of Good Clinical Practice and the Declaration of Helsinki.

Efficacy Assessment

A 5-point scale ranging from 0 (clear) to 4 (severe) was used by the investigator at each study visit to assess the overall psoriasis severity of the treatable areas. Treatment success (the percentage of participants with at least a 2-grade improvement in baseline IGA score and a score of 0 [clear] or 1 [almost clear]) was evaluated at weeks 2, 4, 6, and 8, with a posttreatment follow-up at week 12.

Signs of psoriasis at the target lesion were assessed at each visit using individual 5-point scales ranging from 0 (clear) to 4 (severe). Treatment success was defined as at least a 2-grade improvement from baseline score for each of the key signs—erythema, plaque elevation, and scaling—and reported at weeks 2, 4, 6, and 8, with a posttreatment follow-up at week 12.

Affected BSA also was evaluated at each visit. In addition, an IGA×BSA composite score was calculated by multiplying the IGA by the BSA (range, 9–48 [eg, maximum IGA=4 and maximum BSA=12]) at each time point. The mean percentage change in IGA×BSA from baseline was calculated for each study visit. Additional end points included the achievement of a 50%, 75%, and 90% or greater reduction from baseline IGA×BSA score—IGA×BSA-50, IGA×BSA-75, and IGA×BSA-90—at week 8.

Safety Assessment

Safety evaluations including adverse events (AEs), local skin reactions (LSRs), vital signs, laboratory evaluations, and physical examinations were performed. Information on reported and observed AEs was obtained at each visit. Routine safety laboratory tests were performed at screening, week 4, and week 8. An abbreviated physical examination was performed at baseline, week 8 (end of treatment), and week 12 (end of study). Treatment areas also were examined by the investigator at baseline and each subsequent visit for the presence or absence of marked known drug-related AEs including skin atrophy, striae, telangiectasia, and folliculitis.

LSR Assessment
Local skin reactions such as itching, dryness, and burning/stinging were evaluated at each study visit using 4-point scales ranging from 0 (clear) to 3 (severe). Given the nature of the disease, the presence of LSRs and symptoms at baseline is commonplace, and as such, these evaluations identified both improvement and any emergent issues.

 

 

Statistical Analysis

The primary study goal was to assess differences in treatment efficacy between HP lotion and vehicle with respect to IGA. All statistical processing was performed using SAS unless otherwise stated; statistical tests were 2-sided and performed at the 0.05 level of significance. Markov Chain Monte Carlo multiple imputation was the primary method used to handle missing efficacy data. No imputations were made for missing safety data. All participants were randomized, and the dispensed study drug was included in the intention-to-treat analysis set. This analysis was considered primary for the evaluation of efficacy. Data were analyzed using Cochran-Mantel-Haenszel tests, stratified by analysis center.

Body surface area data were analyzed in a post hoc analysis of covariance with factors of treatment and analysis center and baseline BSA as a covariate. P values for comparisons of percentage change in IGA×BSA were derived from a Wilcoxon rank sum test. For IGA×BSA-50, IGA×BSA-75, and IGA×BSA-90, P values were derived from a Cochran-Mantel-Haenszel test. Last observation carried forward was used to impute data for IGA and BSA through week 8 prior to analysis.

The primary safety analysis was conducted at week 8 using the safety analysis set, which included all participants who were randomized, received at least 1 confirmed dose of the study drug, and had at least 1 postbaseline safety assessment. Adverse events were recorded and classified using the Medical Dictionary for Regulatory Activities (MedDRA, Version 18.0). A post hoc Wilcoxon rank sum test was conducted to compare itching, dryness, and burning/stinging scores at week 8 for HP lotion versus vehicle.

RESULTS
Participant Disposition

Overall, 430 participants were randomized (2:1) to HP lotion (n=285) or vehicle (n=145)(eFigure 1) and included in the intention-to-treat population. Across the 2 studies, 93.3% (n=266) of participants treated with HP lotion and 89.7% (n=130) of participants treated with vehicle completed treatment. The main reasons for study discontinuation with HP lotion were lost to follow-up (3.2%; n=9), participant request (1.8%; n=5), and AEs (1.4%; n=4). Participant request (4.8%; n=7), lost to follow-up (4.1%; n=6), and AEs (1.4%; n=2) also were the main reasons for treatment discontinuation in the vehicle arm.

eFigure 1. Summary of participant disposition in the 2 phase 3 studies (all randomized participants, pooled data [N=430]).

A total of 426 participants were included in the safety population, with no postbaseline safety evaluation in 4 participants.

Baseline Participant Demographics
Demographic data were comparable across the 2 studies. The mean age (SD) was 52.6 (14.13) years. Overall, the majority of participants were male (58.8%; n=253) and white (86.5%; n=372)(eTable 1).



Baseline disease characteristics also were comparable across the treatment groups. Participants had moderate (86.3%; n=371) or severe (13.7%; n=59) disease, with a mean BSA (SD) of 6.1% (2.83) and mean size of target lesion (SD) of 40.4 cm2 (24.14). The majority of participants had moderate (erythema, 84.0%; plaque elevation, 76.0%; and scaling, 74.9%) or severe (erythema, 9.1%; plaque elevation, 13.0%; and scaling, 15.6%) signs of psoriasis at the target lesion site (eTable 2).

Efficacy Evaluation

IGA of Disease Severity
Halobetasol propionate lotion was consistently more effective than its vehicle in achieving treatment success (at least a 2-grade improvement in baseline IGA score and a score of 0 [clear] or 1 [almost clear]). Halobetasol propionate lotion demonstrated statistically significant superiority over vehicle as early as week 2 (P=.003). By week 8, 37.43% of participants in the HP lotion group achieved treatment success compared with 10.03% in the vehicle group (P<.001)(Figure 1).

Figure 1. Investigator global assessment (IGA) of disease severity at each study visit: participants categorized as treatment successes (intention-totreat population pooled study data). Treatment success was defined as at least a 2-grade improvement in baseline IGA score and a score of 0 (clear) or 1 (almost clear). Asterisk indicates P=.003; dagger, P<.001.

 

 

Overall, 39% of participants who had moderate disease (IGA score, 3) at baseline were treatment successes with HP lotion at week 8 compared with 11.53% of participants treated with vehicle; 27.97% of participants with severe disease (IGA score, 4) were treatment successes, with at least a 3-grade improvement in IGA. No participants with severe psoriasis who were treated with vehicle achieved treatment success at week 8. Efficacy was similar in female and male participants, allowing for vehicle effects.

Severity of Signs of Psoriasis (Erythema, Plaque Elevation, and Scaling) at Target Lesion Site
Halobetasol propionate lotion was statistically superior to vehicle in reducing the psoriasis signs of erythema, plaque elevation, and scaling at the target lesion from week 2. At week 8, treatment success (at least a 2-grade improvement from baseline) was achieved by 51.48% (erythema), 57.64% (plaque elevation), and 58.98% (scaling) of participants compared with 17.85%, 23.61%, and 22.82%, respectively, with vehicle (all P<.001)(Figure 2).

Figure 2. Improvement in psoriasis signs of erythema, plaque elevation, and scaling at each study visit: participants categorized as treatment successes (intention-to-treat population pooled study data). Treatment success was defined as at least a 2-grade improvement from baseline. P<.001 at all time points for erythema and scaling. P<.001 at weeks 4, 6, and 8, and P=.056 at week 2 for plaque elevation.


BSA Assessment
Halobetasol propionate lotion was statistically superior to vehicle in reducing BSA from week 2. At week 8 there was a 35.20% reduction in mean BSA for HP lotion compared to 5.85% for vehicle (P<.001)(eFigure 2).

eFigure 2. Percentage reduction in mean body surface area (BSA) from baseline to week 8 (intention-totreat population pooled study data). Asterisk indicates P<.001 vs vehicle.


IGA×BSA Composite Score
At baseline, the mean IGA×BSA scores for HP lotion and vehicle were similar: 19.3 and 18.8, respectively. By week 8, the percentage change in mean IGA×BSA score with HP lotion was 49.44% compared to 13.35% with vehicle (P<.001). Differences were significant from week 2 (P<.001)(Figure 3).

Figure 3. Percentage reduction in IGA×BSA composite tool from baseline to week 8 (intention-to-treat population pooled study data). Asterisk indicates P<.001 vs vehicle. IGA indicates investigator global assessment; BSA, body surface area.


By week 8, 56.8% of participants (n=162) treated with HP lotion had achieved a 50% or greater reduction in baseline IGA×BSA compared to 17.2% of participants treated with vehicle (P<.001). Reductions of IGA×BSA-75 and IGA×BSA-90 were achieved in 39.3% and 19.3% of participants treated with HP lotion, respectively, compared with 9.7% and 2.8% of participants treated with vehicle (both P<.001)(eFigure 3).

eFigure 3. Achievement of 50% (IGA×BSA-50), 75% (IGA×BSA-75), and 90% (IGA×BSA-90) reduction in mean IGA×BSA by week 8 (intent-totreat population pooled study data). Asterisk indicates P<.001 vs vehicle. IGA indicates investigator global assessment; BSA, body surface area.

Safety Evaluation

Adverse event reports were low and similar between the active and vehicle groups. Overall, 61 participants (21.5%) treated with HP lotion reported AEs compared with 34 participants (23.9%) treated with vehicle (Table). The majority of participants treated with HP lotion (90.2%) had AEs that were mild or moderate. There was 1 AE of telangiectasia, not considered treatment related. There were 5 treatment-related AEs for HP lotion, all at the application site: dermatitis (0.7%; n=2), infection (0.4%; n=1), pruritus (0.4%; n=1), and discoloration (0.4%; n=1). There were no AE reports of skin atrophy or folliculitis.

Local Skin Reactions
Most LSRs at baseline were mild to moderate in severity. Itching was the most common, present in 76.8% of participants. Participant-reported burning/stinging was less common, reported by 40.6% of participants. Investigator-reported dryness was noted in 65.7% of participants. There was a rapid improvement in participant-reported itching as early as week 2 that was sustained to the end of the studies, with more gradual improvements in skin dryness and burning/stinging.

COMMENT

Plaque psoriasis is a chronic condition. The rationale behind the development of HP lotion 0.01% was to provide optimal topical treatment of moderate to severe psoriasis, allowing for the potential of prolonged use beyond the 2-week consecutive use normally applied to HP cream 0.05% in a light, once-daily, aesthetically pleasing lotion formulation that patients would prefer.

 

 

Treatment success was rapid and achieved in more than 37% of participants by week 8, with significant improvements in psoriasis signs and symptoms (erythema, plaque elevation, and scaling) compared with vehicle. However, IGA does not consider BSA involvement, a key aspect of disease severity,11,12 and improvements in psoriasis signs of erythema, plaque elevation, and scaling were only assessed at the target lesion. Recently, the product of the IGA and BSA involvement (IGA×BSA) has been proposed as a simple alternative for assessing response to therapy that has been consistently shown to be highly correlated with the psoriasis area and severity index.13-19 Halobetasol propionate lotion 0.01% achieved a 50% reduction in IGA×BSA score by week 8. This efficacy compares well with results reported with apremilast in patients with moderate plaque psoriasis.20

Achieving clinically meaningful outcomes is an important aspect of disease management, especially in psoriasis with its disease burden and detriment to quality of life. It has been suggested that achieving a 75% or greater reduction from baseline IGA×BSA score (IGA×BSA-75) is an appropriate clinical goal.20 In our investigation, IGA×BSA-75 was achieved by 39% of participants treated with HP lotion by week 8, which again compares favorably with 35% of participants in the apremilast study who achieved IGA×BSA-75 at week 16.20

Physicians continue to have long-term safety concerns with TCSs,4,11,12 participants remain concerned about the risk for skin thinning,13 and product labelling restricts HP cream 0.05% consecutive use to 2 weeks. In clinical experience, HP cream 0.05% is well tolerated, with potential local AEs similar to those experienced with other superpotent TCSs. In short-term clinical trials, local AEs at the site of application were reported in up to 13% of patients21-26; itching, burning, or stinging were the most common local AEs (reported in 4.4% of patients).27

There were minimal safety concerns in our 2 studies using an 8-week, once-daily treatment regimen with HP lotion 0.01%. Local AEs at the application site were reported in less than 1% of participants. Baseline itching, dryness, and burning/stinging all improved with treatment.

CONCLUSION

Halobetasol propionate lotion 0.01% provides rapid improvement in disease severity. Halobetasol propionate lotion was consistently more effective than vehicle in achieving treatment success; reducing the BSA affected by the disease; reducing erythema, plaque elevation, and scaling at the target lesion; and improving IGA×BSA score over 8 weeks, which is a realistic time frame to see improvement in psoriasis with a topical steroid. There were minimal safety concerns with prolonged use. Halobetasol propionate lotion may provide an effective and reasonable treatment option in patients with moderate to severe plaque psoriasis.

Acknowledgment
We thank Brian Bulley, MSc (Konic Limited, United Kingdom), for assistance with the preparation of this article. Ortho Dermatologics funded Mr. Bulley’s activities pertaining to this article.

References
  1. Gudjonsson JE, Elder JT. Psoriasis: epidemiology. Clin Dermatol. 2007;25:535-546.
  2. Liu Y, Krueger JG, Bowcock AM. Psoriasis: genetic associations and immune system changes. Genes Immun. 2007;8:1-12.
  3. Nestle FO, Kaplan DH, Barker J. Psoriasis. N Engl J Med. 2009;361:496-509.
  4. Menter A, Korman NJ, Elmets CA, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis. section 3. guidelines of care for the management and treatment of psoriasis with topical therapies. J Am Acad Dermatol. 2009;60:643-659.
  5. Alinia H, Moradi Tuchayi S, Smith JA, et al. Long-term adherence to topical psoriasis treatment can be abysmal: a 1-year randomized intervention study using objective electronic adherence monitoring. Br J Dermatol. 2017;176:759-764.
  6. Young M, Aldredge L, Parker P. Psoriasis for the primary care practitioner. J Am Assoc Nurse Pract. 2017;29:157-178.
  7. Devaux S, Castela A, Archier E, et al. Adherence to topical treatment in psoriasis: a systematic literature review. J Eur Acad Dermatol Venereol. 2012;26(suppl 3):61-67.
  8. Ersser SJ, Cowdell FC, Latter SM, et al. Self-management experiences in adults with mild-moderate psoriasis: an exploratory study and implications for improved support. Br J Dermatol. 2010;163:1044-1049.
  9. Choi CW, Kim BR, Ohn J, et al. The advantage of cyclosporine A and methotrexate rotational therapy in long-term systemic treatment for chronic plaque psoriasis in a real world practice. Ann Dermatol. 2017;29:55-60.
  10. Callis Duffin K, Yeung H, Takeshita J, et al. Patient satisfaction with treatments for moderate-to-severe plaque psoriasis in clinical practice. Br J Dermatol. 2014;170:672-680.
  11. Spuls PI, Lecluse LL, Poulsen ML, et al. How good are clinical severity and outcome measures for psoriasis? quantitative evaluation in a systematic review. J Invest Dermatol. 2010;130:933-943.
  12. Menter A, Gottlieb A, Feldman SR, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: section 1. overview of psoriasis and guidelines of care for the treatment of psoriasis with biologics. J Am Acad Dermatol. 2008;58:826-850.
  13. Bozek A, Reich A. The reliability of three psoriasis assessment tools: psoriasis area severity index, body surface area and physician global assessment. Adv Clin Exp Med. 2017;26:851-856.
  14. Walsh JA, McFadden M, Woodcock J, et al. Product of the Physician Global Assessment and body surface area: a simple static measure of psoriasis severity in a longitudinal cohort. J Am Acad Dermatol. 2013;69:931-937.
  15. Paul C, Cather J, Gooderham M, et al. Efficacy and safety of apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate to severe plaque psoriasis over 52 weeks: a phase III, randomized, controlled trial (ESTEEM 2). Br J Dermatol. 2015;173:1387-1399.
  16. Duffin KC, Papp KA, Bagel J, et al. Evaluation of the Physician Global Assessment and body surface area composite tool for assessing psoriasis response to apremilast therapy: results from ESTEEM 1 and ESTEEM 2. J Drugs Dermatol. 2017;16:147-153.
  17. Chiesa Fuxench ZC, Callis DK, Siegel M, et al. Validity of the Simple Measure for Assessing Psoriasis Activity (S-MAPA) for objectively evaluating disease severity in patients with plaque psoriasis. J Am Acad Dermatol. 2015;73:868-870.
  18. Walsh J. Comparative assessment of PASI and variations of PGA×BSA as measures of psoriasis severity in a clinical trial of moderate to severe psoriasis [poster 1830]. Presented at: Annual Meeting of the American Academy of Dermatology; March 20-24, 2015; San Francisco, CA.
  19. Gottlieb AB, Merola JF, Chen R, et al. Assessing clinical response and defining minimal disease activity in plaque psoriasis with the Physician Global Assessment and body surface area (PGA×BSA) composite tool: An analysis of apremilast phase 3 ESTEEM data. J Am Acad Dermatol. 2017;77:1178-1180.
  20. Strober B, Bagel J, Lebwohl M, et al. Efficacy and safety of apremilast in patients with moderate plaque psoriasis with lower BSA: week 16 results from the UNVEIL study. J Drugs Dermatol. 2017;16:801-808.
  21. Bernhard J, Whitmore C, Guzzo C, et al. Evaluation of halobetasol propionate ointment in the treatment of plaque psoriasis: report on two double-blind, vehicle-controlled studies. J Am Acad Dermatol. 1991;25:1170-1174.
  22. Katz HI, Gross E, Buxman M, et al. A double-blind, vehicle-controlled paired comparison of halobetasol propionate cream on patients with plaque psoriasis. J Am Acad Dermatol. 1991;25:1175-1178.
  23. Blum G, Yawalkar S. A comparative, multicenter, double blind trial of 0.05% halobetasol propionate ointment and 0.1% betamethasone valerate ointment in the treatment of patients with chronic, localized plaque psoriasis. J Am Acad Dermatol. 1991;25:1153-1156.
  24. Goldberg B, Hartdegen R, Presbury D, et al. A double-blind, multicenter comparison of 0.05% halobetasol propionate ointment and 0.05% clobetasol propionate ointment in patients with chronic, localized plaque psoriasis. J Am Acad Dermatol. 1991;25:1145-1148.
  25. Mensing H, Korsukewitz G, Yawalkar S. A double-blind, multicenter comparison between 0.05% halobetasol propionate ointment and 0.05% betamethasone dipropionate ointment in chronic plaque psoriasis. J Am Acad Dermatol. 1991;25:1149-1152.
  26. Herz G, Blum G, Yawalkar S. Halobetasol propionate cream by day and halobetasol propionate ointment at night for the treatment of pediatric patients with chronic, localized psoriasis and atopic dermatitis. J Am Acad Dermatol. 1991;25:1166-1169.
  27. Ultravate [package insert]. Jacksonville, FL: Ranbaxy; 2012.
Article PDF
Author and Disclosure Information

Dr. Sugarman is from the University of California, San Francisco. Dr. Weiss is from Gwinnett Dermatology, PC, and Gwinnett Clinical Research Center, Inc, Snellville, Georgia. Dr. Tanghetti is from the Center for Dermatology and Laser Surgery, Sacramento, California. Dr. Soung is from Southern California Dermatology, Santa Ana. Dr. Yamauchi is from the David Geffen School of Medicine at UCLA, Los Angeles, California. Dr. Lin is from Ortho Dermatologics, Bridgewater, New Jersey. Ms. Harris is from Bausch Health, Bridgewater. Ms. Martin and Dr. Pillai are from Bausch Health, Petaluma, California.

Dr. Sugarman is an advisor and investigator for Bausch Health. Dr. Weiss is a consultant and investigator for Ortho Dermatologics. Dr. Tanghetti is a speaker for AbbVie; Eli Lilly and Company; Galderma Laboratories, LP; LEO Pharma; Ortho Dermatologics; Novartis; and Sun Pharmaceutical Industries, Ltd. He also is an advisory board member for Galderma Laboratories, LP; an advisory board member and investigator for Ortho Dermatologics; and an investigator for LEO Pharma and Novartis. Dr. Soung has received honoraria and/or research grants from and is on the speakers bureau for AbbVie, Amgen Inc, Celgene Corporation, Eli Lilly and Company, Novartis, and Ortho Dermatologics; on the advisory board for Eli Lilly and Company; an investigator for AbbVie and Ortho Dermatologics; and an investigator and consultant for Novartis. She also is an investigator and has received research grants and/or honoraria from Boehringer Ingelheim; GlaxoSmithKline; Janssen Biotech, Inc; Kadmon Corporation; Pfizer Inc; and UCB. Dr. Yamauchi is a consultant, investigator, and speaker for Ortho Dermatologics. Dr. Lin is an employee and stockholder of Ortho Dermatologics. Ms. Harris and Ms. Martin are employees of Bausch Health. Dr. Pillai holds patents from Bausch Health.

These studies were registered at ClinicalTrials.gov with the identifiers NCT02514577 and NCT02515097.

The eFigures and eTables are available in the Appendix.

Correspondence: Jeffrey L. Sugarman, MD, PhD, Redwood Dermatology Research, 2725 Mendocino Ave, Santa Rosa, CA 95403 ([email protected]).

Issue
Cutis - 103(2)
Publications
Topics
Page Number
111-116, E1-E3
Sections
Author and Disclosure Information

Dr. Sugarman is from the University of California, San Francisco. Dr. Weiss is from Gwinnett Dermatology, PC, and Gwinnett Clinical Research Center, Inc, Snellville, Georgia. Dr. Tanghetti is from the Center for Dermatology and Laser Surgery, Sacramento, California. Dr. Soung is from Southern California Dermatology, Santa Ana. Dr. Yamauchi is from the David Geffen School of Medicine at UCLA, Los Angeles, California. Dr. Lin is from Ortho Dermatologics, Bridgewater, New Jersey. Ms. Harris is from Bausch Health, Bridgewater. Ms. Martin and Dr. Pillai are from Bausch Health, Petaluma, California.

Dr. Sugarman is an advisor and investigator for Bausch Health. Dr. Weiss is a consultant and investigator for Ortho Dermatologics. Dr. Tanghetti is a speaker for AbbVie; Eli Lilly and Company; Galderma Laboratories, LP; LEO Pharma; Ortho Dermatologics; Novartis; and Sun Pharmaceutical Industries, Ltd. He also is an advisory board member for Galderma Laboratories, LP; an advisory board member and investigator for Ortho Dermatologics; and an investigator for LEO Pharma and Novartis. Dr. Soung has received honoraria and/or research grants from and is on the speakers bureau for AbbVie, Amgen Inc, Celgene Corporation, Eli Lilly and Company, Novartis, and Ortho Dermatologics; on the advisory board for Eli Lilly and Company; an investigator for AbbVie and Ortho Dermatologics; and an investigator and consultant for Novartis. She also is an investigator and has received research grants and/or honoraria from Boehringer Ingelheim; GlaxoSmithKline; Janssen Biotech, Inc; Kadmon Corporation; Pfizer Inc; and UCB. Dr. Yamauchi is a consultant, investigator, and speaker for Ortho Dermatologics. Dr. Lin is an employee and stockholder of Ortho Dermatologics. Ms. Harris and Ms. Martin are employees of Bausch Health. Dr. Pillai holds patents from Bausch Health.

These studies were registered at ClinicalTrials.gov with the identifiers NCT02514577 and NCT02515097.

The eFigures and eTables are available in the Appendix.

Correspondence: Jeffrey L. Sugarman, MD, PhD, Redwood Dermatology Research, 2725 Mendocino Ave, Santa Rosa, CA 95403 ([email protected]).

Author and Disclosure Information

Dr. Sugarman is from the University of California, San Francisco. Dr. Weiss is from Gwinnett Dermatology, PC, and Gwinnett Clinical Research Center, Inc, Snellville, Georgia. Dr. Tanghetti is from the Center for Dermatology and Laser Surgery, Sacramento, California. Dr. Soung is from Southern California Dermatology, Santa Ana. Dr. Yamauchi is from the David Geffen School of Medicine at UCLA, Los Angeles, California. Dr. Lin is from Ortho Dermatologics, Bridgewater, New Jersey. Ms. Harris is from Bausch Health, Bridgewater. Ms. Martin and Dr. Pillai are from Bausch Health, Petaluma, California.

Dr. Sugarman is an advisor and investigator for Bausch Health. Dr. Weiss is a consultant and investigator for Ortho Dermatologics. Dr. Tanghetti is a speaker for AbbVie; Eli Lilly and Company; Galderma Laboratories, LP; LEO Pharma; Ortho Dermatologics; Novartis; and Sun Pharmaceutical Industries, Ltd. He also is an advisory board member for Galderma Laboratories, LP; an advisory board member and investigator for Ortho Dermatologics; and an investigator for LEO Pharma and Novartis. Dr. Soung has received honoraria and/or research grants from and is on the speakers bureau for AbbVie, Amgen Inc, Celgene Corporation, Eli Lilly and Company, Novartis, and Ortho Dermatologics; on the advisory board for Eli Lilly and Company; an investigator for AbbVie and Ortho Dermatologics; and an investigator and consultant for Novartis. She also is an investigator and has received research grants and/or honoraria from Boehringer Ingelheim; GlaxoSmithKline; Janssen Biotech, Inc; Kadmon Corporation; Pfizer Inc; and UCB. Dr. Yamauchi is a consultant, investigator, and speaker for Ortho Dermatologics. Dr. Lin is an employee and stockholder of Ortho Dermatologics. Ms. Harris and Ms. Martin are employees of Bausch Health. Dr. Pillai holds patents from Bausch Health.

These studies were registered at ClinicalTrials.gov with the identifiers NCT02514577 and NCT02515097.

The eFigures and eTables are available in the Appendix.

Correspondence: Jeffrey L. Sugarman, MD, PhD, Redwood Dermatology Research, 2725 Mendocino Ave, Santa Rosa, CA 95403 ([email protected]).

Article PDF
Article PDF

Psoriasis is a chronic, immune-mediated, inflammatory disease affecting almost 2% of the population.1-3 It is characterized by patches of raised reddish skin covered by silvery-white scales. Most patients have limited disease (<5% body surface area [BSA] involvement) that can be managed with topical agents.4 Topical corticosteroids (TCSs) are considered first-line therapy for mild to moderate disease because of the inflammatory nature of the condition and often are used in conjunction with systemic agents in more severe psoriasis.4

As many as 20% to 30% of patients with moderate to severe plaque psoriasis have inadequate disease control.5 Several factors may affect patient outcomes; however, drug selection and patient adherence are important given the chronic nature of the disease. A survey of 1200 patients with psoriasis reported nonadherence rates of 73% with topical therapy.6 In addition, patients tend to apply less than the recommended dose or abandon treatment altogether if rapid improvement does not occur7,8; it is not uncommon for patients with psoriasis to mistakenly believe treatment will improve their condition within 1 to 2 weeks.9 Patient satisfaction with topical treatments is low, partly because of these false expectations and formulation issues. Treatments can be greasy and sticky, with unpleasant odors and the potential to stain clothes and linens.7,10 Safety concerns with TCSs also limit their consecutive use beyond 2 to 4 weeks, which is not ideal for a disease that requires a long-term management strategy.

A potent/superpotent TCS that is administered once daily and has a safety profile that affords longer-term, once-daily treatment in an aesthetically pleasing formulation would seem ideal. Herein, we investigate the safety and tolerability of a novel low-concentration (0.01%) lotion formulation of halobetasol propionate (HP), reporting on the pooled data from 2 phase 3 clinical studies in participants with moderate to severe psoriasis.

METHODS
Study Design

We conducted 2 multicenter, double-blind, randomized, parallel-group phase 3 studies to assess the safety, tolerability, and efficacy of HP lotion 0.01% in participants with a clinical diagnosis of moderate to severe psoriasis with an investigator global assessment (IGA) score of 3 or 4 and an affected BSA of 3% to 12%. Participants were randomized (2:1) to receive HP lotion or vehicle applied topically to the affected area once daily for 8 weeks.

 

 

Inclusion and Exclusion Criteria
The studies included individuals of either sex aged 18 years or older. A target lesion was defined primarily to assess signs of psoriasis, measuring 16 to 100 cm2, with a score of 3 (moderate) or higher for 2 of 3 different psoriasis signs—erythema, plaque elevation, and scaling—and summed score of 8 or higher, with no sign scoring less than 2. Participants who had pustular psoriasis or used phototherapy, photochemotherapy, or systemic psoriasis therapy within the prior 4 weeks or biologics within the prior 3 months, or those who were diagnosed with skin conditions that would interfere with the interpretation of results were excluded from the studies.

Study Oversight
Participants provided written informed consent before study-related procedures were performed, and the protocol and consent were approved by institutional review boards or ethics committees at all investigational sites. The study was conducted in accordance with the principles of Good Clinical Practice and the Declaration of Helsinki.

Efficacy Assessment

A 5-point scale ranging from 0 (clear) to 4 (severe) was used by the investigator at each study visit to assess the overall psoriasis severity of the treatable areas. Treatment success (the percentage of participants with at least a 2-grade improvement in baseline IGA score and a score of 0 [clear] or 1 [almost clear]) was evaluated at weeks 2, 4, 6, and 8, with a posttreatment follow-up at week 12.

Signs of psoriasis at the target lesion were assessed at each visit using individual 5-point scales ranging from 0 (clear) to 4 (severe). Treatment success was defined as at least a 2-grade improvement from baseline score for each of the key signs—erythema, plaque elevation, and scaling—and reported at weeks 2, 4, 6, and 8, with a posttreatment follow-up at week 12.

Affected BSA also was evaluated at each visit. In addition, an IGA×BSA composite score was calculated by multiplying the IGA by the BSA (range, 9–48 [eg, maximum IGA=4 and maximum BSA=12]) at each time point. The mean percentage change in IGA×BSA from baseline was calculated for each study visit. Additional end points included the achievement of a 50%, 75%, and 90% or greater reduction from baseline IGA×BSA score—IGA×BSA-50, IGA×BSA-75, and IGA×BSA-90—at week 8.

Safety Assessment

Safety evaluations including adverse events (AEs), local skin reactions (LSRs), vital signs, laboratory evaluations, and physical examinations were performed. Information on reported and observed AEs was obtained at each visit. Routine safety laboratory tests were performed at screening, week 4, and week 8. An abbreviated physical examination was performed at baseline, week 8 (end of treatment), and week 12 (end of study). Treatment areas also were examined by the investigator at baseline and each subsequent visit for the presence or absence of marked known drug-related AEs including skin atrophy, striae, telangiectasia, and folliculitis.

LSR Assessment
Local skin reactions such as itching, dryness, and burning/stinging were evaluated at each study visit using 4-point scales ranging from 0 (clear) to 3 (severe). Given the nature of the disease, the presence of LSRs and symptoms at baseline is commonplace, and as such, these evaluations identified both improvement and any emergent issues.

 

 

Statistical Analysis

The primary study goal was to assess differences in treatment efficacy between HP lotion and vehicle with respect to IGA. All statistical processing was performed using SAS unless otherwise stated; statistical tests were 2-sided and performed at the 0.05 level of significance. Markov Chain Monte Carlo multiple imputation was the primary method used to handle missing efficacy data. No imputations were made for missing safety data. All participants were randomized, and the dispensed study drug was included in the intention-to-treat analysis set. This analysis was considered primary for the evaluation of efficacy. Data were analyzed using Cochran-Mantel-Haenszel tests, stratified by analysis center.

Body surface area data were analyzed in a post hoc analysis of covariance with factors of treatment and analysis center and baseline BSA as a covariate. P values for comparisons of percentage change in IGA×BSA were derived from a Wilcoxon rank sum test. For IGA×BSA-50, IGA×BSA-75, and IGA×BSA-90, P values were derived from a Cochran-Mantel-Haenszel test. Last observation carried forward was used to impute data for IGA and BSA through week 8 prior to analysis.

The primary safety analysis was conducted at week 8 using the safety analysis set, which included all participants who were randomized, received at least 1 confirmed dose of the study drug, and had at least 1 postbaseline safety assessment. Adverse events were recorded and classified using the Medical Dictionary for Regulatory Activities (MedDRA, Version 18.0). A post hoc Wilcoxon rank sum test was conducted to compare itching, dryness, and burning/stinging scores at week 8 for HP lotion versus vehicle.

RESULTS
Participant Disposition

Overall, 430 participants were randomized (2:1) to HP lotion (n=285) or vehicle (n=145)(eFigure 1) and included in the intention-to-treat population. Across the 2 studies, 93.3% (n=266) of participants treated with HP lotion and 89.7% (n=130) of participants treated with vehicle completed treatment. The main reasons for study discontinuation with HP lotion were lost to follow-up (3.2%; n=9), participant request (1.8%; n=5), and AEs (1.4%; n=4). Participant request (4.8%; n=7), lost to follow-up (4.1%; n=6), and AEs (1.4%; n=2) also were the main reasons for treatment discontinuation in the vehicle arm.

eFigure 1. Summary of participant disposition in the 2 phase 3 studies (all randomized participants, pooled data [N=430]).

A total of 426 participants were included in the safety population, with no postbaseline safety evaluation in 4 participants.

Baseline Participant Demographics
Demographic data were comparable across the 2 studies. The mean age (SD) was 52.6 (14.13) years. Overall, the majority of participants were male (58.8%; n=253) and white (86.5%; n=372)(eTable 1).



Baseline disease characteristics also were comparable across the treatment groups. Participants had moderate (86.3%; n=371) or severe (13.7%; n=59) disease, with a mean BSA (SD) of 6.1% (2.83) and mean size of target lesion (SD) of 40.4 cm2 (24.14). The majority of participants had moderate (erythema, 84.0%; plaque elevation, 76.0%; and scaling, 74.9%) or severe (erythema, 9.1%; plaque elevation, 13.0%; and scaling, 15.6%) signs of psoriasis at the target lesion site (eTable 2).

Efficacy Evaluation

IGA of Disease Severity
Halobetasol propionate lotion was consistently more effective than its vehicle in achieving treatment success (at least a 2-grade improvement in baseline IGA score and a score of 0 [clear] or 1 [almost clear]). Halobetasol propionate lotion demonstrated statistically significant superiority over vehicle as early as week 2 (P=.003). By week 8, 37.43% of participants in the HP lotion group achieved treatment success compared with 10.03% in the vehicle group (P<.001)(Figure 1).

Figure 1. Investigator global assessment (IGA) of disease severity at each study visit: participants categorized as treatment successes (intention-totreat population pooled study data). Treatment success was defined as at least a 2-grade improvement in baseline IGA score and a score of 0 (clear) or 1 (almost clear). Asterisk indicates P=.003; dagger, P<.001.

 

 

Overall, 39% of participants who had moderate disease (IGA score, 3) at baseline were treatment successes with HP lotion at week 8 compared with 11.53% of participants treated with vehicle; 27.97% of participants with severe disease (IGA score, 4) were treatment successes, with at least a 3-grade improvement in IGA. No participants with severe psoriasis who were treated with vehicle achieved treatment success at week 8. Efficacy was similar in female and male participants, allowing for vehicle effects.

Severity of Signs of Psoriasis (Erythema, Plaque Elevation, and Scaling) at Target Lesion Site
Halobetasol propionate lotion was statistically superior to vehicle in reducing the psoriasis signs of erythema, plaque elevation, and scaling at the target lesion from week 2. At week 8, treatment success (at least a 2-grade improvement from baseline) was achieved by 51.48% (erythema), 57.64% (plaque elevation), and 58.98% (scaling) of participants compared with 17.85%, 23.61%, and 22.82%, respectively, with vehicle (all P<.001)(Figure 2).

Figure 2. Improvement in psoriasis signs of erythema, plaque elevation, and scaling at each study visit: participants categorized as treatment successes (intention-to-treat population pooled study data). Treatment success was defined as at least a 2-grade improvement from baseline. P<.001 at all time points for erythema and scaling. P<.001 at weeks 4, 6, and 8, and P=.056 at week 2 for plaque elevation.


BSA Assessment
Halobetasol propionate lotion was statistically superior to vehicle in reducing BSA from week 2. At week 8 there was a 35.20% reduction in mean BSA for HP lotion compared to 5.85% for vehicle (P<.001)(eFigure 2).

eFigure 2. Percentage reduction in mean body surface area (BSA) from baseline to week 8 (intention-totreat population pooled study data). Asterisk indicates P<.001 vs vehicle.


IGA×BSA Composite Score
At baseline, the mean IGA×BSA scores for HP lotion and vehicle were similar: 19.3 and 18.8, respectively. By week 8, the percentage change in mean IGA×BSA score with HP lotion was 49.44% compared to 13.35% with vehicle (P<.001). Differences were significant from week 2 (P<.001)(Figure 3).

Figure 3. Percentage reduction in IGA×BSA composite tool from baseline to week 8 (intention-to-treat population pooled study data). Asterisk indicates P<.001 vs vehicle. IGA indicates investigator global assessment; BSA, body surface area.


By week 8, 56.8% of participants (n=162) treated with HP lotion had achieved a 50% or greater reduction in baseline IGA×BSA compared to 17.2% of participants treated with vehicle (P<.001). Reductions of IGA×BSA-75 and IGA×BSA-90 were achieved in 39.3% and 19.3% of participants treated with HP lotion, respectively, compared with 9.7% and 2.8% of participants treated with vehicle (both P<.001)(eFigure 3).

eFigure 3. Achievement of 50% (IGA×BSA-50), 75% (IGA×BSA-75), and 90% (IGA×BSA-90) reduction in mean IGA×BSA by week 8 (intent-totreat population pooled study data). Asterisk indicates P<.001 vs vehicle. IGA indicates investigator global assessment; BSA, body surface area.

Safety Evaluation

Adverse event reports were low and similar between the active and vehicle groups. Overall, 61 participants (21.5%) treated with HP lotion reported AEs compared with 34 participants (23.9%) treated with vehicle (Table). The majority of participants treated with HP lotion (90.2%) had AEs that were mild or moderate. There was 1 AE of telangiectasia, not considered treatment related. There were 5 treatment-related AEs for HP lotion, all at the application site: dermatitis (0.7%; n=2), infection (0.4%; n=1), pruritus (0.4%; n=1), and discoloration (0.4%; n=1). There were no AE reports of skin atrophy or folliculitis.

Local Skin Reactions
Most LSRs at baseline were mild to moderate in severity. Itching was the most common, present in 76.8% of participants. Participant-reported burning/stinging was less common, reported by 40.6% of participants. Investigator-reported dryness was noted in 65.7% of participants. There was a rapid improvement in participant-reported itching as early as week 2 that was sustained to the end of the studies, with more gradual improvements in skin dryness and burning/stinging.

COMMENT

Plaque psoriasis is a chronic condition. The rationale behind the development of HP lotion 0.01% was to provide optimal topical treatment of moderate to severe psoriasis, allowing for the potential of prolonged use beyond the 2-week consecutive use normally applied to HP cream 0.05% in a light, once-daily, aesthetically pleasing lotion formulation that patients would prefer.

 

 

Treatment success was rapid and achieved in more than 37% of participants by week 8, with significant improvements in psoriasis signs and symptoms (erythema, plaque elevation, and scaling) compared with vehicle. However, IGA does not consider BSA involvement, a key aspect of disease severity,11,12 and improvements in psoriasis signs of erythema, plaque elevation, and scaling were only assessed at the target lesion. Recently, the product of the IGA and BSA involvement (IGA×BSA) has been proposed as a simple alternative for assessing response to therapy that has been consistently shown to be highly correlated with the psoriasis area and severity index.13-19 Halobetasol propionate lotion 0.01% achieved a 50% reduction in IGA×BSA score by week 8. This efficacy compares well with results reported with apremilast in patients with moderate plaque psoriasis.20

Achieving clinically meaningful outcomes is an important aspect of disease management, especially in psoriasis with its disease burden and detriment to quality of life. It has been suggested that achieving a 75% or greater reduction from baseline IGA×BSA score (IGA×BSA-75) is an appropriate clinical goal.20 In our investigation, IGA×BSA-75 was achieved by 39% of participants treated with HP lotion by week 8, which again compares favorably with 35% of participants in the apremilast study who achieved IGA×BSA-75 at week 16.20

Physicians continue to have long-term safety concerns with TCSs,4,11,12 participants remain concerned about the risk for skin thinning,13 and product labelling restricts HP cream 0.05% consecutive use to 2 weeks. In clinical experience, HP cream 0.05% is well tolerated, with potential local AEs similar to those experienced with other superpotent TCSs. In short-term clinical trials, local AEs at the site of application were reported in up to 13% of patients21-26; itching, burning, or stinging were the most common local AEs (reported in 4.4% of patients).27

There were minimal safety concerns in our 2 studies using an 8-week, once-daily treatment regimen with HP lotion 0.01%. Local AEs at the application site were reported in less than 1% of participants. Baseline itching, dryness, and burning/stinging all improved with treatment.

CONCLUSION

Halobetasol propionate lotion 0.01% provides rapid improvement in disease severity. Halobetasol propionate lotion was consistently more effective than vehicle in achieving treatment success; reducing the BSA affected by the disease; reducing erythema, plaque elevation, and scaling at the target lesion; and improving IGA×BSA score over 8 weeks, which is a realistic time frame to see improvement in psoriasis with a topical steroid. There were minimal safety concerns with prolonged use. Halobetasol propionate lotion may provide an effective and reasonable treatment option in patients with moderate to severe plaque psoriasis.

Acknowledgment
We thank Brian Bulley, MSc (Konic Limited, United Kingdom), for assistance with the preparation of this article. Ortho Dermatologics funded Mr. Bulley’s activities pertaining to this article.

Psoriasis is a chronic, immune-mediated, inflammatory disease affecting almost 2% of the population.1-3 It is characterized by patches of raised reddish skin covered by silvery-white scales. Most patients have limited disease (<5% body surface area [BSA] involvement) that can be managed with topical agents.4 Topical corticosteroids (TCSs) are considered first-line therapy for mild to moderate disease because of the inflammatory nature of the condition and often are used in conjunction with systemic agents in more severe psoriasis.4

As many as 20% to 30% of patients with moderate to severe plaque psoriasis have inadequate disease control.5 Several factors may affect patient outcomes; however, drug selection and patient adherence are important given the chronic nature of the disease. A survey of 1200 patients with psoriasis reported nonadherence rates of 73% with topical therapy.6 In addition, patients tend to apply less than the recommended dose or abandon treatment altogether if rapid improvement does not occur7,8; it is not uncommon for patients with psoriasis to mistakenly believe treatment will improve their condition within 1 to 2 weeks.9 Patient satisfaction with topical treatments is low, partly because of these false expectations and formulation issues. Treatments can be greasy and sticky, with unpleasant odors and the potential to stain clothes and linens.7,10 Safety concerns with TCSs also limit their consecutive use beyond 2 to 4 weeks, which is not ideal for a disease that requires a long-term management strategy.

A potent/superpotent TCS that is administered once daily and has a safety profile that affords longer-term, once-daily treatment in an aesthetically pleasing formulation would seem ideal. Herein, we investigate the safety and tolerability of a novel low-concentration (0.01%) lotion formulation of halobetasol propionate (HP), reporting on the pooled data from 2 phase 3 clinical studies in participants with moderate to severe psoriasis.

METHODS
Study Design

We conducted 2 multicenter, double-blind, randomized, parallel-group phase 3 studies to assess the safety, tolerability, and efficacy of HP lotion 0.01% in participants with a clinical diagnosis of moderate to severe psoriasis with an investigator global assessment (IGA) score of 3 or 4 and an affected BSA of 3% to 12%. Participants were randomized (2:1) to receive HP lotion or vehicle applied topically to the affected area once daily for 8 weeks.

 

 

Inclusion and Exclusion Criteria
The studies included individuals of either sex aged 18 years or older. A target lesion was defined primarily to assess signs of psoriasis, measuring 16 to 100 cm2, with a score of 3 (moderate) or higher for 2 of 3 different psoriasis signs—erythema, plaque elevation, and scaling—and summed score of 8 or higher, with no sign scoring less than 2. Participants who had pustular psoriasis or used phototherapy, photochemotherapy, or systemic psoriasis therapy within the prior 4 weeks or biologics within the prior 3 months, or those who were diagnosed with skin conditions that would interfere with the interpretation of results were excluded from the studies.

Study Oversight
Participants provided written informed consent before study-related procedures were performed, and the protocol and consent were approved by institutional review boards or ethics committees at all investigational sites. The study was conducted in accordance with the principles of Good Clinical Practice and the Declaration of Helsinki.

Efficacy Assessment

A 5-point scale ranging from 0 (clear) to 4 (severe) was used by the investigator at each study visit to assess the overall psoriasis severity of the treatable areas. Treatment success (the percentage of participants with at least a 2-grade improvement in baseline IGA score and a score of 0 [clear] or 1 [almost clear]) was evaluated at weeks 2, 4, 6, and 8, with a posttreatment follow-up at week 12.

Signs of psoriasis at the target lesion were assessed at each visit using individual 5-point scales ranging from 0 (clear) to 4 (severe). Treatment success was defined as at least a 2-grade improvement from baseline score for each of the key signs—erythema, plaque elevation, and scaling—and reported at weeks 2, 4, 6, and 8, with a posttreatment follow-up at week 12.

Affected BSA also was evaluated at each visit. In addition, an IGA×BSA composite score was calculated by multiplying the IGA by the BSA (range, 9–48 [eg, maximum IGA=4 and maximum BSA=12]) at each time point. The mean percentage change in IGA×BSA from baseline was calculated for each study visit. Additional end points included the achievement of a 50%, 75%, and 90% or greater reduction from baseline IGA×BSA score—IGA×BSA-50, IGA×BSA-75, and IGA×BSA-90—at week 8.

Safety Assessment

Safety evaluations including adverse events (AEs), local skin reactions (LSRs), vital signs, laboratory evaluations, and physical examinations were performed. Information on reported and observed AEs was obtained at each visit. Routine safety laboratory tests were performed at screening, week 4, and week 8. An abbreviated physical examination was performed at baseline, week 8 (end of treatment), and week 12 (end of study). Treatment areas also were examined by the investigator at baseline and each subsequent visit for the presence or absence of marked known drug-related AEs including skin atrophy, striae, telangiectasia, and folliculitis.

LSR Assessment
Local skin reactions such as itching, dryness, and burning/stinging were evaluated at each study visit using 4-point scales ranging from 0 (clear) to 3 (severe). Given the nature of the disease, the presence of LSRs and symptoms at baseline is commonplace, and as such, these evaluations identified both improvement and any emergent issues.

 

 

Statistical Analysis

The primary study goal was to assess differences in treatment efficacy between HP lotion and vehicle with respect to IGA. All statistical processing was performed using SAS unless otherwise stated; statistical tests were 2-sided and performed at the 0.05 level of significance. Markov Chain Monte Carlo multiple imputation was the primary method used to handle missing efficacy data. No imputations were made for missing safety data. All participants were randomized, and the dispensed study drug was included in the intention-to-treat analysis set. This analysis was considered primary for the evaluation of efficacy. Data were analyzed using Cochran-Mantel-Haenszel tests, stratified by analysis center.

Body surface area data were analyzed in a post hoc analysis of covariance with factors of treatment and analysis center and baseline BSA as a covariate. P values for comparisons of percentage change in IGA×BSA were derived from a Wilcoxon rank sum test. For IGA×BSA-50, IGA×BSA-75, and IGA×BSA-90, P values were derived from a Cochran-Mantel-Haenszel test. Last observation carried forward was used to impute data for IGA and BSA through week 8 prior to analysis.

The primary safety analysis was conducted at week 8 using the safety analysis set, which included all participants who were randomized, received at least 1 confirmed dose of the study drug, and had at least 1 postbaseline safety assessment. Adverse events were recorded and classified using the Medical Dictionary for Regulatory Activities (MedDRA, Version 18.0). A post hoc Wilcoxon rank sum test was conducted to compare itching, dryness, and burning/stinging scores at week 8 for HP lotion versus vehicle.

RESULTS
Participant Disposition

Overall, 430 participants were randomized (2:1) to HP lotion (n=285) or vehicle (n=145)(eFigure 1) and included in the intention-to-treat population. Across the 2 studies, 93.3% (n=266) of participants treated with HP lotion and 89.7% (n=130) of participants treated with vehicle completed treatment. The main reasons for study discontinuation with HP lotion were lost to follow-up (3.2%; n=9), participant request (1.8%; n=5), and AEs (1.4%; n=4). Participant request (4.8%; n=7), lost to follow-up (4.1%; n=6), and AEs (1.4%; n=2) also were the main reasons for treatment discontinuation in the vehicle arm.

eFigure 1. Summary of participant disposition in the 2 phase 3 studies (all randomized participants, pooled data [N=430]).

A total of 426 participants were included in the safety population, with no postbaseline safety evaluation in 4 participants.

Baseline Participant Demographics
Demographic data were comparable across the 2 studies. The mean age (SD) was 52.6 (14.13) years. Overall, the majority of participants were male (58.8%; n=253) and white (86.5%; n=372)(eTable 1).



Baseline disease characteristics also were comparable across the treatment groups. Participants had moderate (86.3%; n=371) or severe (13.7%; n=59) disease, with a mean BSA (SD) of 6.1% (2.83) and mean size of target lesion (SD) of 40.4 cm2 (24.14). The majority of participants had moderate (erythema, 84.0%; plaque elevation, 76.0%; and scaling, 74.9%) or severe (erythema, 9.1%; plaque elevation, 13.0%; and scaling, 15.6%) signs of psoriasis at the target lesion site (eTable 2).

Efficacy Evaluation

IGA of Disease Severity
Halobetasol propionate lotion was consistently more effective than its vehicle in achieving treatment success (at least a 2-grade improvement in baseline IGA score and a score of 0 [clear] or 1 [almost clear]). Halobetasol propionate lotion demonstrated statistically significant superiority over vehicle as early as week 2 (P=.003). By week 8, 37.43% of participants in the HP lotion group achieved treatment success compared with 10.03% in the vehicle group (P<.001)(Figure 1).

Figure 1. Investigator global assessment (IGA) of disease severity at each study visit: participants categorized as treatment successes (intention-totreat population pooled study data). Treatment success was defined as at least a 2-grade improvement in baseline IGA score and a score of 0 (clear) or 1 (almost clear). Asterisk indicates P=.003; dagger, P<.001.

 

 

Overall, 39% of participants who had moderate disease (IGA score, 3) at baseline were treatment successes with HP lotion at week 8 compared with 11.53% of participants treated with vehicle; 27.97% of participants with severe disease (IGA score, 4) were treatment successes, with at least a 3-grade improvement in IGA. No participants with severe psoriasis who were treated with vehicle achieved treatment success at week 8. Efficacy was similar in female and male participants, allowing for vehicle effects.

Severity of Signs of Psoriasis (Erythema, Plaque Elevation, and Scaling) at Target Lesion Site
Halobetasol propionate lotion was statistically superior to vehicle in reducing the psoriasis signs of erythema, plaque elevation, and scaling at the target lesion from week 2. At week 8, treatment success (at least a 2-grade improvement from baseline) was achieved by 51.48% (erythema), 57.64% (plaque elevation), and 58.98% (scaling) of participants compared with 17.85%, 23.61%, and 22.82%, respectively, with vehicle (all P<.001)(Figure 2).

Figure 2. Improvement in psoriasis signs of erythema, plaque elevation, and scaling at each study visit: participants categorized as treatment successes (intention-to-treat population pooled study data). Treatment success was defined as at least a 2-grade improvement from baseline. P<.001 at all time points for erythema and scaling. P<.001 at weeks 4, 6, and 8, and P=.056 at week 2 for plaque elevation.


BSA Assessment
Halobetasol propionate lotion was statistically superior to vehicle in reducing BSA from week 2. At week 8 there was a 35.20% reduction in mean BSA for HP lotion compared to 5.85% for vehicle (P<.001)(eFigure 2).

eFigure 2. Percentage reduction in mean body surface area (BSA) from baseline to week 8 (intention-totreat population pooled study data). Asterisk indicates P<.001 vs vehicle.


IGA×BSA Composite Score
At baseline, the mean IGA×BSA scores for HP lotion and vehicle were similar: 19.3 and 18.8, respectively. By week 8, the percentage change in mean IGA×BSA score with HP lotion was 49.44% compared to 13.35% with vehicle (P<.001). Differences were significant from week 2 (P<.001)(Figure 3).

Figure 3. Percentage reduction in IGA×BSA composite tool from baseline to week 8 (intention-to-treat population pooled study data). Asterisk indicates P<.001 vs vehicle. IGA indicates investigator global assessment; BSA, body surface area.


By week 8, 56.8% of participants (n=162) treated with HP lotion had achieved a 50% or greater reduction in baseline IGA×BSA compared to 17.2% of participants treated with vehicle (P<.001). Reductions of IGA×BSA-75 and IGA×BSA-90 were achieved in 39.3% and 19.3% of participants treated with HP lotion, respectively, compared with 9.7% and 2.8% of participants treated with vehicle (both P<.001)(eFigure 3).

eFigure 3. Achievement of 50% (IGA×BSA-50), 75% (IGA×BSA-75), and 90% (IGA×BSA-90) reduction in mean IGA×BSA by week 8 (intent-totreat population pooled study data). Asterisk indicates P<.001 vs vehicle. IGA indicates investigator global assessment; BSA, body surface area.

Safety Evaluation

Adverse event reports were low and similar between the active and vehicle groups. Overall, 61 participants (21.5%) treated with HP lotion reported AEs compared with 34 participants (23.9%) treated with vehicle (Table). The majority of participants treated with HP lotion (90.2%) had AEs that were mild or moderate. There was 1 AE of telangiectasia, not considered treatment related. There were 5 treatment-related AEs for HP lotion, all at the application site: dermatitis (0.7%; n=2), infection (0.4%; n=1), pruritus (0.4%; n=1), and discoloration (0.4%; n=1). There were no AE reports of skin atrophy or folliculitis.

Local Skin Reactions
Most LSRs at baseline were mild to moderate in severity. Itching was the most common, present in 76.8% of participants. Participant-reported burning/stinging was less common, reported by 40.6% of participants. Investigator-reported dryness was noted in 65.7% of participants. There was a rapid improvement in participant-reported itching as early as week 2 that was sustained to the end of the studies, with more gradual improvements in skin dryness and burning/stinging.

COMMENT

Plaque psoriasis is a chronic condition. The rationale behind the development of HP lotion 0.01% was to provide optimal topical treatment of moderate to severe psoriasis, allowing for the potential of prolonged use beyond the 2-week consecutive use normally applied to HP cream 0.05% in a light, once-daily, aesthetically pleasing lotion formulation that patients would prefer.

 

 

Treatment success was rapid and achieved in more than 37% of participants by week 8, with significant improvements in psoriasis signs and symptoms (erythema, plaque elevation, and scaling) compared with vehicle. However, IGA does not consider BSA involvement, a key aspect of disease severity,11,12 and improvements in psoriasis signs of erythema, plaque elevation, and scaling were only assessed at the target lesion. Recently, the product of the IGA and BSA involvement (IGA×BSA) has been proposed as a simple alternative for assessing response to therapy that has been consistently shown to be highly correlated with the psoriasis area and severity index.13-19 Halobetasol propionate lotion 0.01% achieved a 50% reduction in IGA×BSA score by week 8. This efficacy compares well with results reported with apremilast in patients with moderate plaque psoriasis.20

Achieving clinically meaningful outcomes is an important aspect of disease management, especially in psoriasis with its disease burden and detriment to quality of life. It has been suggested that achieving a 75% or greater reduction from baseline IGA×BSA score (IGA×BSA-75) is an appropriate clinical goal.20 In our investigation, IGA×BSA-75 was achieved by 39% of participants treated with HP lotion by week 8, which again compares favorably with 35% of participants in the apremilast study who achieved IGA×BSA-75 at week 16.20

Physicians continue to have long-term safety concerns with TCSs,4,11,12 participants remain concerned about the risk for skin thinning,13 and product labelling restricts HP cream 0.05% consecutive use to 2 weeks. In clinical experience, HP cream 0.05% is well tolerated, with potential local AEs similar to those experienced with other superpotent TCSs. In short-term clinical trials, local AEs at the site of application were reported in up to 13% of patients21-26; itching, burning, or stinging were the most common local AEs (reported in 4.4% of patients).27

There were minimal safety concerns in our 2 studies using an 8-week, once-daily treatment regimen with HP lotion 0.01%. Local AEs at the application site were reported in less than 1% of participants. Baseline itching, dryness, and burning/stinging all improved with treatment.

CONCLUSION

Halobetasol propionate lotion 0.01% provides rapid improvement in disease severity. Halobetasol propionate lotion was consistently more effective than vehicle in achieving treatment success; reducing the BSA affected by the disease; reducing erythema, plaque elevation, and scaling at the target lesion; and improving IGA×BSA score over 8 weeks, which is a realistic time frame to see improvement in psoriasis with a topical steroid. There were minimal safety concerns with prolonged use. Halobetasol propionate lotion may provide an effective and reasonable treatment option in patients with moderate to severe plaque psoriasis.

Acknowledgment
We thank Brian Bulley, MSc (Konic Limited, United Kingdom), for assistance with the preparation of this article. Ortho Dermatologics funded Mr. Bulley’s activities pertaining to this article.

References
  1. Gudjonsson JE, Elder JT. Psoriasis: epidemiology. Clin Dermatol. 2007;25:535-546.
  2. Liu Y, Krueger JG, Bowcock AM. Psoriasis: genetic associations and immune system changes. Genes Immun. 2007;8:1-12.
  3. Nestle FO, Kaplan DH, Barker J. Psoriasis. N Engl J Med. 2009;361:496-509.
  4. Menter A, Korman NJ, Elmets CA, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis. section 3. guidelines of care for the management and treatment of psoriasis with topical therapies. J Am Acad Dermatol. 2009;60:643-659.
  5. Alinia H, Moradi Tuchayi S, Smith JA, et al. Long-term adherence to topical psoriasis treatment can be abysmal: a 1-year randomized intervention study using objective electronic adherence monitoring. Br J Dermatol. 2017;176:759-764.
  6. Young M, Aldredge L, Parker P. Psoriasis for the primary care practitioner. J Am Assoc Nurse Pract. 2017;29:157-178.
  7. Devaux S, Castela A, Archier E, et al. Adherence to topical treatment in psoriasis: a systematic literature review. J Eur Acad Dermatol Venereol. 2012;26(suppl 3):61-67.
  8. Ersser SJ, Cowdell FC, Latter SM, et al. Self-management experiences in adults with mild-moderate psoriasis: an exploratory study and implications for improved support. Br J Dermatol. 2010;163:1044-1049.
  9. Choi CW, Kim BR, Ohn J, et al. The advantage of cyclosporine A and methotrexate rotational therapy in long-term systemic treatment for chronic plaque psoriasis in a real world practice. Ann Dermatol. 2017;29:55-60.
  10. Callis Duffin K, Yeung H, Takeshita J, et al. Patient satisfaction with treatments for moderate-to-severe plaque psoriasis in clinical practice. Br J Dermatol. 2014;170:672-680.
  11. Spuls PI, Lecluse LL, Poulsen ML, et al. How good are clinical severity and outcome measures for psoriasis? quantitative evaluation in a systematic review. J Invest Dermatol. 2010;130:933-943.
  12. Menter A, Gottlieb A, Feldman SR, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: section 1. overview of psoriasis and guidelines of care for the treatment of psoriasis with biologics. J Am Acad Dermatol. 2008;58:826-850.
  13. Bozek A, Reich A. The reliability of three psoriasis assessment tools: psoriasis area severity index, body surface area and physician global assessment. Adv Clin Exp Med. 2017;26:851-856.
  14. Walsh JA, McFadden M, Woodcock J, et al. Product of the Physician Global Assessment and body surface area: a simple static measure of psoriasis severity in a longitudinal cohort. J Am Acad Dermatol. 2013;69:931-937.
  15. Paul C, Cather J, Gooderham M, et al. Efficacy and safety of apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate to severe plaque psoriasis over 52 weeks: a phase III, randomized, controlled trial (ESTEEM 2). Br J Dermatol. 2015;173:1387-1399.
  16. Duffin KC, Papp KA, Bagel J, et al. Evaluation of the Physician Global Assessment and body surface area composite tool for assessing psoriasis response to apremilast therapy: results from ESTEEM 1 and ESTEEM 2. J Drugs Dermatol. 2017;16:147-153.
  17. Chiesa Fuxench ZC, Callis DK, Siegel M, et al. Validity of the Simple Measure for Assessing Psoriasis Activity (S-MAPA) for objectively evaluating disease severity in patients with plaque psoriasis. J Am Acad Dermatol. 2015;73:868-870.
  18. Walsh J. Comparative assessment of PASI and variations of PGA×BSA as measures of psoriasis severity in a clinical trial of moderate to severe psoriasis [poster 1830]. Presented at: Annual Meeting of the American Academy of Dermatology; March 20-24, 2015; San Francisco, CA.
  19. Gottlieb AB, Merola JF, Chen R, et al. Assessing clinical response and defining minimal disease activity in plaque psoriasis with the Physician Global Assessment and body surface area (PGA×BSA) composite tool: An analysis of apremilast phase 3 ESTEEM data. J Am Acad Dermatol. 2017;77:1178-1180.
  20. Strober B, Bagel J, Lebwohl M, et al. Efficacy and safety of apremilast in patients with moderate plaque psoriasis with lower BSA: week 16 results from the UNVEIL study. J Drugs Dermatol. 2017;16:801-808.
  21. Bernhard J, Whitmore C, Guzzo C, et al. Evaluation of halobetasol propionate ointment in the treatment of plaque psoriasis: report on two double-blind, vehicle-controlled studies. J Am Acad Dermatol. 1991;25:1170-1174.
  22. Katz HI, Gross E, Buxman M, et al. A double-blind, vehicle-controlled paired comparison of halobetasol propionate cream on patients with plaque psoriasis. J Am Acad Dermatol. 1991;25:1175-1178.
  23. Blum G, Yawalkar S. A comparative, multicenter, double blind trial of 0.05% halobetasol propionate ointment and 0.1% betamethasone valerate ointment in the treatment of patients with chronic, localized plaque psoriasis. J Am Acad Dermatol. 1991;25:1153-1156.
  24. Goldberg B, Hartdegen R, Presbury D, et al. A double-blind, multicenter comparison of 0.05% halobetasol propionate ointment and 0.05% clobetasol propionate ointment in patients with chronic, localized plaque psoriasis. J Am Acad Dermatol. 1991;25:1145-1148.
  25. Mensing H, Korsukewitz G, Yawalkar S. A double-blind, multicenter comparison between 0.05% halobetasol propionate ointment and 0.05% betamethasone dipropionate ointment in chronic plaque psoriasis. J Am Acad Dermatol. 1991;25:1149-1152.
  26. Herz G, Blum G, Yawalkar S. Halobetasol propionate cream by day and halobetasol propionate ointment at night for the treatment of pediatric patients with chronic, localized psoriasis and atopic dermatitis. J Am Acad Dermatol. 1991;25:1166-1169.
  27. Ultravate [package insert]. Jacksonville, FL: Ranbaxy; 2012.
References
  1. Gudjonsson JE, Elder JT. Psoriasis: epidemiology. Clin Dermatol. 2007;25:535-546.
  2. Liu Y, Krueger JG, Bowcock AM. Psoriasis: genetic associations and immune system changes. Genes Immun. 2007;8:1-12.
  3. Nestle FO, Kaplan DH, Barker J. Psoriasis. N Engl J Med. 2009;361:496-509.
  4. Menter A, Korman NJ, Elmets CA, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis. section 3. guidelines of care for the management and treatment of psoriasis with topical therapies. J Am Acad Dermatol. 2009;60:643-659.
  5. Alinia H, Moradi Tuchayi S, Smith JA, et al. Long-term adherence to topical psoriasis treatment can be abysmal: a 1-year randomized intervention study using objective electronic adherence monitoring. Br J Dermatol. 2017;176:759-764.
  6. Young M, Aldredge L, Parker P. Psoriasis for the primary care practitioner. J Am Assoc Nurse Pract. 2017;29:157-178.
  7. Devaux S, Castela A, Archier E, et al. Adherence to topical treatment in psoriasis: a systematic literature review. J Eur Acad Dermatol Venereol. 2012;26(suppl 3):61-67.
  8. Ersser SJ, Cowdell FC, Latter SM, et al. Self-management experiences in adults with mild-moderate psoriasis: an exploratory study and implications for improved support. Br J Dermatol. 2010;163:1044-1049.
  9. Choi CW, Kim BR, Ohn J, et al. The advantage of cyclosporine A and methotrexate rotational therapy in long-term systemic treatment for chronic plaque psoriasis in a real world practice. Ann Dermatol. 2017;29:55-60.
  10. Callis Duffin K, Yeung H, Takeshita J, et al. Patient satisfaction with treatments for moderate-to-severe plaque psoriasis in clinical practice. Br J Dermatol. 2014;170:672-680.
  11. Spuls PI, Lecluse LL, Poulsen ML, et al. How good are clinical severity and outcome measures for psoriasis? quantitative evaluation in a systematic review. J Invest Dermatol. 2010;130:933-943.
  12. Menter A, Gottlieb A, Feldman SR, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: section 1. overview of psoriasis and guidelines of care for the treatment of psoriasis with biologics. J Am Acad Dermatol. 2008;58:826-850.
  13. Bozek A, Reich A. The reliability of three psoriasis assessment tools: psoriasis area severity index, body surface area and physician global assessment. Adv Clin Exp Med. 2017;26:851-856.
  14. Walsh JA, McFadden M, Woodcock J, et al. Product of the Physician Global Assessment and body surface area: a simple static measure of psoriasis severity in a longitudinal cohort. J Am Acad Dermatol. 2013;69:931-937.
  15. Paul C, Cather J, Gooderham M, et al. Efficacy and safety of apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate to severe plaque psoriasis over 52 weeks: a phase III, randomized, controlled trial (ESTEEM 2). Br J Dermatol. 2015;173:1387-1399.
  16. Duffin KC, Papp KA, Bagel J, et al. Evaluation of the Physician Global Assessment and body surface area composite tool for assessing psoriasis response to apremilast therapy: results from ESTEEM 1 and ESTEEM 2. J Drugs Dermatol. 2017;16:147-153.
  17. Chiesa Fuxench ZC, Callis DK, Siegel M, et al. Validity of the Simple Measure for Assessing Psoriasis Activity (S-MAPA) for objectively evaluating disease severity in patients with plaque psoriasis. J Am Acad Dermatol. 2015;73:868-870.
  18. Walsh J. Comparative assessment of PASI and variations of PGA×BSA as measures of psoriasis severity in a clinical trial of moderate to severe psoriasis [poster 1830]. Presented at: Annual Meeting of the American Academy of Dermatology; March 20-24, 2015; San Francisco, CA.
  19. Gottlieb AB, Merola JF, Chen R, et al. Assessing clinical response and defining minimal disease activity in plaque psoriasis with the Physician Global Assessment and body surface area (PGA×BSA) composite tool: An analysis of apremilast phase 3 ESTEEM data. J Am Acad Dermatol. 2017;77:1178-1180.
  20. Strober B, Bagel J, Lebwohl M, et al. Efficacy and safety of apremilast in patients with moderate plaque psoriasis with lower BSA: week 16 results from the UNVEIL study. J Drugs Dermatol. 2017;16:801-808.
  21. Bernhard J, Whitmore C, Guzzo C, et al. Evaluation of halobetasol propionate ointment in the treatment of plaque psoriasis: report on two double-blind, vehicle-controlled studies. J Am Acad Dermatol. 1991;25:1170-1174.
  22. Katz HI, Gross E, Buxman M, et al. A double-blind, vehicle-controlled paired comparison of halobetasol propionate cream on patients with plaque psoriasis. J Am Acad Dermatol. 1991;25:1175-1178.
  23. Blum G, Yawalkar S. A comparative, multicenter, double blind trial of 0.05% halobetasol propionate ointment and 0.1% betamethasone valerate ointment in the treatment of patients with chronic, localized plaque psoriasis. J Am Acad Dermatol. 1991;25:1153-1156.
  24. Goldberg B, Hartdegen R, Presbury D, et al. A double-blind, multicenter comparison of 0.05% halobetasol propionate ointment and 0.05% clobetasol propionate ointment in patients with chronic, localized plaque psoriasis. J Am Acad Dermatol. 1991;25:1145-1148.
  25. Mensing H, Korsukewitz G, Yawalkar S. A double-blind, multicenter comparison between 0.05% halobetasol propionate ointment and 0.05% betamethasone dipropionate ointment in chronic plaque psoriasis. J Am Acad Dermatol. 1991;25:1149-1152.
  26. Herz G, Blum G, Yawalkar S. Halobetasol propionate cream by day and halobetasol propionate ointment at night for the treatment of pediatric patients with chronic, localized psoriasis and atopic dermatitis. J Am Acad Dermatol. 1991;25:1166-1169.
  27. Ultravate [package insert]. Jacksonville, FL: Ranbaxy; 2012.
Issue
Cutis - 103(2)
Issue
Cutis - 103(2)
Page Number
111-116, E1-E3
Page Number
111-116, E1-E3
Publications
Publications
Topics
Article Type
Display Headline
Safety and Efficacy of Halobetasol Propionate Lotion 0.01% in the Treatment of Moderate to Severe Plaque Psoriasis: A Pooled Analysis of 2 Phase 3 Studies
Display Headline
Safety and Efficacy of Halobetasol Propionate Lotion 0.01% in the Treatment of Moderate to Severe Plaque Psoriasis: A Pooled Analysis of 2 Phase 3 Studies
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media