Allowed Publications
Slot System
Featured Buckets
Featured Buckets Admin

Miliarial Gout in an Immunocompromised Patient

Article Type
Changed
Fri, 05/05/2023 - 09:06
Display Headline
Miliarial Gout in an Immunocompromised Patient

To the Editor:

Miliarial gout is a rare intradermal manifestation of tophaceous gout. It was first described in 2007 when a patient presented with multiple small papules with a red base containing a white- to cream-colored substance,1 which has rarely been reported,1-6 according to a PubMed search of articles indexed for MEDLINE from 2007 to 2023 using the term miliarial gout. We describe a case of miliarial gout in a patient with a history of gout, uric acid levels within reference range, and immunocompromised status due to a prior orthotopic heart transplant.

Miliarial gout. Multiple subcutaneous, firm, popcornlike papules on the right posterior upper arm.
FIGURE 1. Miliarial gout. Multiple subcutaneous, firm, popcornlike papules on the right posterior upper arm.

A 59-year-old man presented with innumerable subcutaneous, firm, popcornlike clustered papules on the posterior surfaces of the upper arms and thighs of 5 years’ duration (Figure 1). The involved areas were sometimes painful on manipulation, but the patient was otherwise asymptomatic. His medical history was notable for tophaceous gout of more than 10 years’ duration, calcinosis cutis, adrenal insufficiency, essential hypertension, and an orthotopic heart transplant 2 years prior to the current presentation. At the current presentation he was taking tacrolimus, colchicine, febuxostat, and low-dose prednisone. The patient denied any other skin changes such as ulceration or bullae. In addition to the innumerable subcutaneous papules, he had much larger firm deep nodules bilaterally on the elbow (Figure 2). A complete blood cell count with differential and comprehensive metabolic panel results were within reference range. A 4-mm punch biopsy of the right posterior arm revealed dermal deposits consistent with gout on hematoxylin and eosin staining (Figure 3) but no calcium deposits on von Kossa staining, consistent with miliarial gout.

Firm nodules consistent with miliarial gout on the right elbow.
FIGURE 2. Firm nodules consistent with miliarial gout on the right elbow.

He was treated with 0.6 mg of colchicine daily, 80 mg of febuxostat twice daily, and 2.5 mg of prednisone daily. Unfortunately, the patient had difficulty affording his medications and therefore experienced frequent flares.

A, Low-power histopathology revealed nodular aggregates of acellular material with areas varying in color from pale to eosinophilic (H&E, original magnification ×2).
FIGURE 3. A, Low-power histopathology revealed nodular aggregates of acellular material with areas varying in color from pale to eosinophilic (H&E, original magnification ×2). B, On closer inspection, the acellular material showed a feathery appearance with prominent clefts and empty spaces (H&E, original magnification ×10). C, There was a multinucleated (foreign body–type) giant cell reaction around the amorphous material (H&E, original magnification ×40).

Gout is caused by inflammation that occurs from deposition of monosodium urate crystals in tissues, most commonly occurring in the skin and joints. Gout affects8.3 million individuals and is one of the most common rheumatic diseases of adulthood. The classic presentation of the acute form is monoarticular with associated swelling, erythema, and pain. The chronic form (also known as tophaceous gout) affects soft tissue and presents with smooth or multilobulated nodules.2 Miliarial gout is a rare variant of chronic tophaceous gout, and the diagnosis is based on atypical location, size, and distribution of tophi deposition.

In the updated American College of Rheumatology criteria for gout published in 2020, tophi are defined as draining or chalklike subcutaneous nodules that typically are located in joints, ears, olecranon bursae, finger pads, and tendons.3 The term miliarial gout, which is not universally defined, is used to describe the morphology and distribution of tophi deposition in areas outside of the typical locations defined by the American College of Rheumatology criteria. Miliarial refers to the small, multilobulated, and disseminated presentation of tophi. The involvement of atypical locations distinguishes miliarial gout from chronic tophaceous gout.

The cause of tophi deposition in atypical locations is unknown. It is thought that patients with a history of sustained hyperuricemia have a much greater burden of urate crystal deposition, which can lead to involvement of atypical locations. Our patient had innumerable, discrete, 1- to 5-mm, multilobulated tophi located on the posterior upper arms and thighs even though his uric acid levels were within reference range over the last 5 years.

Miliarial gout is a rare entity.1 In 2007, Shukla et al1 coined the term miliarial gout when reporting the first known presentation of a patient with multiple tiny papules containing a white or creamlike substance scattered on an erythematous base. Other cases of miliarial gout have commonly involved the metacarpophalangeal joints of the hands, knees, abdomen, extensor forearms, and thighs.5 Similarly, our patient had disease involvement of the posterior upper arms and thighs. Furthermore, miliarial gout has been associated with carpal tunnel syndrome; monosodium urate crystal deposition in this space can lead to a clinical diagnosis of this condition.6

With a history of orthotopic heart transplant, it is possible that our patient’s immunocompromised status could have increased his susceptibility for the miliarial form of chronic tophaceous gout. Gout reportedly is the most common inflammatory arthritis in transplant recipients, with the highest prevalence following renal and heart transplantation.7 Pretransplant hyperuricemia is correlated with higher probabilities of posttransplant gout.8 In patients with a heart transplant, hyperuricemia may be due to diuretic use. Additionally, the presence of a gout diagnosis before transplant nearly triples the likelihood of posttransplant gout, which often is more severe than de novo gout, as seen in our patient. Calcineurin inhibitors, including tacrolimus, also can predispose patients to hyperuricemia and more severe forms of gout in the posttransplant phase by limiting fractional urate excretion within the first 3 months of therapy.7 Treatment with oral steroids, as in our patient, also has been identified as a potential inciting factor for the development of cutaneous tophaceous gout.9

Treatment with allopurinol and colchicine has been effective in patients with miliarial gout. Obesity and long-term treatment with furosemide (which our patient was not taking) are considered risk factors for the deposition of dermal and hypodermal urates.9 Our patient had a body mass index of 35 (≥30 indicates obesity); therefore, he also should be counseled on lifestyle modifications for optimal disease control.

References
  1. Shukla R, Vender RB, Alhabeeb A, et al. Miliarial gout (a new entity). J Cutan Med Surg. 2007;11:31-34.
  2. Zhu Y, Pandya BJ, Choi HK. Prevalence of gout and hyperuricemia in the US general population: the National Health and Nutrition Examination Survey 2007-2008. Arthritis Rheum. 2011;63:3136-3141.
  3. Neogi T, Jansen, TL, Dalbeth N, et al. 2015 gout classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheumatol. 2015;67:2557-2568.
  4. Hung TL, Wang WM, Chiang CP. Miliarial gout: a rare presentation of extensive cutaneous tophi. QJM. 2016;109:811-812.
  5. Mireku KA, Burgy JR, Davis LS. Miliarial gout: a rare clinical presentation. J Am Acad Dermatol. 2014;71:E17-E18.
  6. Sadovici-Bobeica V, Mazur-Nicorici L, Nicorici A, et al. Chronic miliarial gout associated with carpal tunnel syndrome: a very rare clinical presentation. Eur J Case Rep Intern Med. 2018;5:000926.
  7. Schwab P, Lipton S, Kerr GS. Rheumatologic sequelae and challenges in organ transplantation. Best Pract Res Clin Rheumatol. 2010;24:329-340.
  8. Hernández-Molina G, Cachafeiro-Vilar A, Villa AR, et al. Gout in renal allograft recipients according to the pretransplant hyperuricemic status. Transplantation. 2008;86:1543-1547.
  9. Aguayo RS, Baradad M, Soria X, et al. Unilateral milia‐type intradermal tophi associated with underlying urate subcutaneous deposition: an uncommon cutaneous presentation of gout. Clin Exp Dermatol. 2013;38:622-625.
Article PDF
Author and Disclosure Information

From the Duke University Medical Center, Durham, North Carolina. Drs. Patel, Eldik, and Nicholas are from the Department of Dermatology, and Drs. Selim and Al-Rohil are from the Department of Pathology.

The authors report no conflict of interest.

Correspondence: Matilda Nicholas, MD, PhD, Duke University School of Medicine, Department of Dermatology, 234 Crooked Creek Parkway, Durham, NC 27713 (matilda.nicholas@duke.edu).

Issue
Cutis - 111(4)
Publications
Topics
Page Number
E42-E44
Sections
Author and Disclosure Information

From the Duke University Medical Center, Durham, North Carolina. Drs. Patel, Eldik, and Nicholas are from the Department of Dermatology, and Drs. Selim and Al-Rohil are from the Department of Pathology.

The authors report no conflict of interest.

Correspondence: Matilda Nicholas, MD, PhD, Duke University School of Medicine, Department of Dermatology, 234 Crooked Creek Parkway, Durham, NC 27713 (matilda.nicholas@duke.edu).

Author and Disclosure Information

From the Duke University Medical Center, Durham, North Carolina. Drs. Patel, Eldik, and Nicholas are from the Department of Dermatology, and Drs. Selim and Al-Rohil are from the Department of Pathology.

The authors report no conflict of interest.

Correspondence: Matilda Nicholas, MD, PhD, Duke University School of Medicine, Department of Dermatology, 234 Crooked Creek Parkway, Durham, NC 27713 (matilda.nicholas@duke.edu).

Article PDF
Article PDF

To the Editor:

Miliarial gout is a rare intradermal manifestation of tophaceous gout. It was first described in 2007 when a patient presented with multiple small papules with a red base containing a white- to cream-colored substance,1 which has rarely been reported,1-6 according to a PubMed search of articles indexed for MEDLINE from 2007 to 2023 using the term miliarial gout. We describe a case of miliarial gout in a patient with a history of gout, uric acid levels within reference range, and immunocompromised status due to a prior orthotopic heart transplant.

Miliarial gout. Multiple subcutaneous, firm, popcornlike papules on the right posterior upper arm.
FIGURE 1. Miliarial gout. Multiple subcutaneous, firm, popcornlike papules on the right posterior upper arm.

A 59-year-old man presented with innumerable subcutaneous, firm, popcornlike clustered papules on the posterior surfaces of the upper arms and thighs of 5 years’ duration (Figure 1). The involved areas were sometimes painful on manipulation, but the patient was otherwise asymptomatic. His medical history was notable for tophaceous gout of more than 10 years’ duration, calcinosis cutis, adrenal insufficiency, essential hypertension, and an orthotopic heart transplant 2 years prior to the current presentation. At the current presentation he was taking tacrolimus, colchicine, febuxostat, and low-dose prednisone. The patient denied any other skin changes such as ulceration or bullae. In addition to the innumerable subcutaneous papules, he had much larger firm deep nodules bilaterally on the elbow (Figure 2). A complete blood cell count with differential and comprehensive metabolic panel results were within reference range. A 4-mm punch biopsy of the right posterior arm revealed dermal deposits consistent with gout on hematoxylin and eosin staining (Figure 3) but no calcium deposits on von Kossa staining, consistent with miliarial gout.

Firm nodules consistent with miliarial gout on the right elbow.
FIGURE 2. Firm nodules consistent with miliarial gout on the right elbow.

He was treated with 0.6 mg of colchicine daily, 80 mg of febuxostat twice daily, and 2.5 mg of prednisone daily. Unfortunately, the patient had difficulty affording his medications and therefore experienced frequent flares.

A, Low-power histopathology revealed nodular aggregates of acellular material with areas varying in color from pale to eosinophilic (H&E, original magnification ×2).
FIGURE 3. A, Low-power histopathology revealed nodular aggregates of acellular material with areas varying in color from pale to eosinophilic (H&E, original magnification ×2). B, On closer inspection, the acellular material showed a feathery appearance with prominent clefts and empty spaces (H&E, original magnification ×10). C, There was a multinucleated (foreign body–type) giant cell reaction around the amorphous material (H&E, original magnification ×40).

Gout is caused by inflammation that occurs from deposition of monosodium urate crystals in tissues, most commonly occurring in the skin and joints. Gout affects8.3 million individuals and is one of the most common rheumatic diseases of adulthood. The classic presentation of the acute form is monoarticular with associated swelling, erythema, and pain. The chronic form (also known as tophaceous gout) affects soft tissue and presents with smooth or multilobulated nodules.2 Miliarial gout is a rare variant of chronic tophaceous gout, and the diagnosis is based on atypical location, size, and distribution of tophi deposition.

In the updated American College of Rheumatology criteria for gout published in 2020, tophi are defined as draining or chalklike subcutaneous nodules that typically are located in joints, ears, olecranon bursae, finger pads, and tendons.3 The term miliarial gout, which is not universally defined, is used to describe the morphology and distribution of tophi deposition in areas outside of the typical locations defined by the American College of Rheumatology criteria. Miliarial refers to the small, multilobulated, and disseminated presentation of tophi. The involvement of atypical locations distinguishes miliarial gout from chronic tophaceous gout.

The cause of tophi deposition in atypical locations is unknown. It is thought that patients with a history of sustained hyperuricemia have a much greater burden of urate crystal deposition, which can lead to involvement of atypical locations. Our patient had innumerable, discrete, 1- to 5-mm, multilobulated tophi located on the posterior upper arms and thighs even though his uric acid levels were within reference range over the last 5 years.

Miliarial gout is a rare entity.1 In 2007, Shukla et al1 coined the term miliarial gout when reporting the first known presentation of a patient with multiple tiny papules containing a white or creamlike substance scattered on an erythematous base. Other cases of miliarial gout have commonly involved the metacarpophalangeal joints of the hands, knees, abdomen, extensor forearms, and thighs.5 Similarly, our patient had disease involvement of the posterior upper arms and thighs. Furthermore, miliarial gout has been associated with carpal tunnel syndrome; monosodium urate crystal deposition in this space can lead to a clinical diagnosis of this condition.6

With a history of orthotopic heart transplant, it is possible that our patient’s immunocompromised status could have increased his susceptibility for the miliarial form of chronic tophaceous gout. Gout reportedly is the most common inflammatory arthritis in transplant recipients, with the highest prevalence following renal and heart transplantation.7 Pretransplant hyperuricemia is correlated with higher probabilities of posttransplant gout.8 In patients with a heart transplant, hyperuricemia may be due to diuretic use. Additionally, the presence of a gout diagnosis before transplant nearly triples the likelihood of posttransplant gout, which often is more severe than de novo gout, as seen in our patient. Calcineurin inhibitors, including tacrolimus, also can predispose patients to hyperuricemia and more severe forms of gout in the posttransplant phase by limiting fractional urate excretion within the first 3 months of therapy.7 Treatment with oral steroids, as in our patient, also has been identified as a potential inciting factor for the development of cutaneous tophaceous gout.9

Treatment with allopurinol and colchicine has been effective in patients with miliarial gout. Obesity and long-term treatment with furosemide (which our patient was not taking) are considered risk factors for the deposition of dermal and hypodermal urates.9 Our patient had a body mass index of 35 (≥30 indicates obesity); therefore, he also should be counseled on lifestyle modifications for optimal disease control.

To the Editor:

Miliarial gout is a rare intradermal manifestation of tophaceous gout. It was first described in 2007 when a patient presented with multiple small papules with a red base containing a white- to cream-colored substance,1 which has rarely been reported,1-6 according to a PubMed search of articles indexed for MEDLINE from 2007 to 2023 using the term miliarial gout. We describe a case of miliarial gout in a patient with a history of gout, uric acid levels within reference range, and immunocompromised status due to a prior orthotopic heart transplant.

Miliarial gout. Multiple subcutaneous, firm, popcornlike papules on the right posterior upper arm.
FIGURE 1. Miliarial gout. Multiple subcutaneous, firm, popcornlike papules on the right posterior upper arm.

A 59-year-old man presented with innumerable subcutaneous, firm, popcornlike clustered papules on the posterior surfaces of the upper arms and thighs of 5 years’ duration (Figure 1). The involved areas were sometimes painful on manipulation, but the patient was otherwise asymptomatic. His medical history was notable for tophaceous gout of more than 10 years’ duration, calcinosis cutis, adrenal insufficiency, essential hypertension, and an orthotopic heart transplant 2 years prior to the current presentation. At the current presentation he was taking tacrolimus, colchicine, febuxostat, and low-dose prednisone. The patient denied any other skin changes such as ulceration or bullae. In addition to the innumerable subcutaneous papules, he had much larger firm deep nodules bilaterally on the elbow (Figure 2). A complete blood cell count with differential and comprehensive metabolic panel results were within reference range. A 4-mm punch biopsy of the right posterior arm revealed dermal deposits consistent with gout on hematoxylin and eosin staining (Figure 3) but no calcium deposits on von Kossa staining, consistent with miliarial gout.

Firm nodules consistent with miliarial gout on the right elbow.
FIGURE 2. Firm nodules consistent with miliarial gout on the right elbow.

He was treated with 0.6 mg of colchicine daily, 80 mg of febuxostat twice daily, and 2.5 mg of prednisone daily. Unfortunately, the patient had difficulty affording his medications and therefore experienced frequent flares.

A, Low-power histopathology revealed nodular aggregates of acellular material with areas varying in color from pale to eosinophilic (H&E, original magnification ×2).
FIGURE 3. A, Low-power histopathology revealed nodular aggregates of acellular material with areas varying in color from pale to eosinophilic (H&E, original magnification ×2). B, On closer inspection, the acellular material showed a feathery appearance with prominent clefts and empty spaces (H&E, original magnification ×10). C, There was a multinucleated (foreign body–type) giant cell reaction around the amorphous material (H&E, original magnification ×40).

Gout is caused by inflammation that occurs from deposition of monosodium urate crystals in tissues, most commonly occurring in the skin and joints. Gout affects8.3 million individuals and is one of the most common rheumatic diseases of adulthood. The classic presentation of the acute form is monoarticular with associated swelling, erythema, and pain. The chronic form (also known as tophaceous gout) affects soft tissue and presents with smooth or multilobulated nodules.2 Miliarial gout is a rare variant of chronic tophaceous gout, and the diagnosis is based on atypical location, size, and distribution of tophi deposition.

In the updated American College of Rheumatology criteria for gout published in 2020, tophi are defined as draining or chalklike subcutaneous nodules that typically are located in joints, ears, olecranon bursae, finger pads, and tendons.3 The term miliarial gout, which is not universally defined, is used to describe the morphology and distribution of tophi deposition in areas outside of the typical locations defined by the American College of Rheumatology criteria. Miliarial refers to the small, multilobulated, and disseminated presentation of tophi. The involvement of atypical locations distinguishes miliarial gout from chronic tophaceous gout.

The cause of tophi deposition in atypical locations is unknown. It is thought that patients with a history of sustained hyperuricemia have a much greater burden of urate crystal deposition, which can lead to involvement of atypical locations. Our patient had innumerable, discrete, 1- to 5-mm, multilobulated tophi located on the posterior upper arms and thighs even though his uric acid levels were within reference range over the last 5 years.

Miliarial gout is a rare entity.1 In 2007, Shukla et al1 coined the term miliarial gout when reporting the first known presentation of a patient with multiple tiny papules containing a white or creamlike substance scattered on an erythematous base. Other cases of miliarial gout have commonly involved the metacarpophalangeal joints of the hands, knees, abdomen, extensor forearms, and thighs.5 Similarly, our patient had disease involvement of the posterior upper arms and thighs. Furthermore, miliarial gout has been associated with carpal tunnel syndrome; monosodium urate crystal deposition in this space can lead to a clinical diagnosis of this condition.6

With a history of orthotopic heart transplant, it is possible that our patient’s immunocompromised status could have increased his susceptibility for the miliarial form of chronic tophaceous gout. Gout reportedly is the most common inflammatory arthritis in transplant recipients, with the highest prevalence following renal and heart transplantation.7 Pretransplant hyperuricemia is correlated with higher probabilities of posttransplant gout.8 In patients with a heart transplant, hyperuricemia may be due to diuretic use. Additionally, the presence of a gout diagnosis before transplant nearly triples the likelihood of posttransplant gout, which often is more severe than de novo gout, as seen in our patient. Calcineurin inhibitors, including tacrolimus, also can predispose patients to hyperuricemia and more severe forms of gout in the posttransplant phase by limiting fractional urate excretion within the first 3 months of therapy.7 Treatment with oral steroids, as in our patient, also has been identified as a potential inciting factor for the development of cutaneous tophaceous gout.9

Treatment with allopurinol and colchicine has been effective in patients with miliarial gout. Obesity and long-term treatment with furosemide (which our patient was not taking) are considered risk factors for the deposition of dermal and hypodermal urates.9 Our patient had a body mass index of 35 (≥30 indicates obesity); therefore, he also should be counseled on lifestyle modifications for optimal disease control.

References
  1. Shukla R, Vender RB, Alhabeeb A, et al. Miliarial gout (a new entity). J Cutan Med Surg. 2007;11:31-34.
  2. Zhu Y, Pandya BJ, Choi HK. Prevalence of gout and hyperuricemia in the US general population: the National Health and Nutrition Examination Survey 2007-2008. Arthritis Rheum. 2011;63:3136-3141.
  3. Neogi T, Jansen, TL, Dalbeth N, et al. 2015 gout classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheumatol. 2015;67:2557-2568.
  4. Hung TL, Wang WM, Chiang CP. Miliarial gout: a rare presentation of extensive cutaneous tophi. QJM. 2016;109:811-812.
  5. Mireku KA, Burgy JR, Davis LS. Miliarial gout: a rare clinical presentation. J Am Acad Dermatol. 2014;71:E17-E18.
  6. Sadovici-Bobeica V, Mazur-Nicorici L, Nicorici A, et al. Chronic miliarial gout associated with carpal tunnel syndrome: a very rare clinical presentation. Eur J Case Rep Intern Med. 2018;5:000926.
  7. Schwab P, Lipton S, Kerr GS. Rheumatologic sequelae and challenges in organ transplantation. Best Pract Res Clin Rheumatol. 2010;24:329-340.
  8. Hernández-Molina G, Cachafeiro-Vilar A, Villa AR, et al. Gout in renal allograft recipients according to the pretransplant hyperuricemic status. Transplantation. 2008;86:1543-1547.
  9. Aguayo RS, Baradad M, Soria X, et al. Unilateral milia‐type intradermal tophi associated with underlying urate subcutaneous deposition: an uncommon cutaneous presentation of gout. Clin Exp Dermatol. 2013;38:622-625.
References
  1. Shukla R, Vender RB, Alhabeeb A, et al. Miliarial gout (a new entity). J Cutan Med Surg. 2007;11:31-34.
  2. Zhu Y, Pandya BJ, Choi HK. Prevalence of gout and hyperuricemia in the US general population: the National Health and Nutrition Examination Survey 2007-2008. Arthritis Rheum. 2011;63:3136-3141.
  3. Neogi T, Jansen, TL, Dalbeth N, et al. 2015 gout classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheumatol. 2015;67:2557-2568.
  4. Hung TL, Wang WM, Chiang CP. Miliarial gout: a rare presentation of extensive cutaneous tophi. QJM. 2016;109:811-812.
  5. Mireku KA, Burgy JR, Davis LS. Miliarial gout: a rare clinical presentation. J Am Acad Dermatol. 2014;71:E17-E18.
  6. Sadovici-Bobeica V, Mazur-Nicorici L, Nicorici A, et al. Chronic miliarial gout associated with carpal tunnel syndrome: a very rare clinical presentation. Eur J Case Rep Intern Med. 2018;5:000926.
  7. Schwab P, Lipton S, Kerr GS. Rheumatologic sequelae and challenges in organ transplantation. Best Pract Res Clin Rheumatol. 2010;24:329-340.
  8. Hernández-Molina G, Cachafeiro-Vilar A, Villa AR, et al. Gout in renal allograft recipients according to the pretransplant hyperuricemic status. Transplantation. 2008;86:1543-1547.
  9. Aguayo RS, Baradad M, Soria X, et al. Unilateral milia‐type intradermal tophi associated with underlying urate subcutaneous deposition: an uncommon cutaneous presentation of gout. Clin Exp Dermatol. 2013;38:622-625.
Issue
Cutis - 111(4)
Issue
Cutis - 111(4)
Page Number
E42-E44
Page Number
E42-E44
Publications
Publications
Topics
Article Type
Display Headline
Miliarial Gout in an Immunocompromised Patient
Display Headline
Miliarial Gout in an Immunocompromised Patient
Sections
Inside the Article

Practice Points

  • Miliarial gout is a rare intradermal manifestation of tophaceous gout and often presents as multiple small papules containing a white- to cream-colored substance.
  • Immunocompromised status may be a risk factor for miliarial gout, especially in patients with a history of gout or hyperuricemia.
  • Effective treatments for miliarial gout include allopurinol and colchicine.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Botulinum Toxin and Glycopyrrolate Combination Therapy for Hailey-Hailey Disease

Article Type
Changed
Thu, 04/27/2023 - 09:22
Display Headline
Botulinum Toxin and Glycopyrrolate Combination Therapy for Hailey-Hailey Disease

To the Editor:

Hailey-Hailey disease (HHD)(also known as familial benign chronic pemphigus) is an inherited autosomal-dominant condition in the family of chronic bullous diseases. It is characterized by flaccid blisters, erosions, and macerated vegetative plaques with a predilection for intertriginous sites. Lesions often are weeping, painful, pruritic, and malodorous, leading to decreased quality of life for patients. Complications of this chronic disease include an increased risk for secondary infection and malignant transformation to squamous cell carcinoma.1

Treatment of HHD remains difficult. Topical steroids, oral steroids, and ablative techniques such as dermabrasion and ablative lasers are the most widely reported therapies. OnabotulinumtoxinA has been described as a successful treatment for patients with HHD, including for disease recalcitrant to other therapies.2 We describe 2 patients with HHD who responded to treatment with intralesional onabotulinumtoxinA injections with and without adjuvant oral glycopyrrolate.

A 54-year-old woman presented with painful flaccid blisters under the breasts (Figure 1A) and in the axillae and groin of 3 weeks’ duration. Biopsy results from this initial visit were consistent with a diagnosis of HHD. The patient reported that the onset of blisters coincided with episodes of severe hyperhidrosis. Therapy with topical and oral steroids, antifungals, antibiotics, and topical aluminum chloride failed to achieve adequate disease control. After a discussion of the risks and benefits, the patient agreed to treatment with injections of onabotulinumtoxinA. At months 0, 3, and 6, the patient received 50 U of onabotulinumtoxinA under the breasts and in the axillae and the groin, for a total of 250 U each session. Each injection consisted of 2.5 U of onabotulinumtoxinA spaced 1-cm apart. Clinical improvement was noted within 2 weeks of initiating neuromodulator therapy. Follow-up at 9 months demonstrated improvement (Figure 1B); however, complete clearance was not achieved, and the patient required ongoing treatment with onabotulinumtoxinA every 3 months.

Hailey-Hailey disease under the breast at presentation and 9 months after initiating treatment with onabotulinumtoxinA, respectively.
FIGURE 1. A and B, Hailey-Hailey disease under the breast at presentation and 9 months after initiating treatment with onabotulinumtoxinA, respectively

A 43-year-old woman presented with erythematous eroded plaques of the antecubital fossae, axillae, and chest (Figure 2A) of 10 years’ duration. A biopsy from an outside provider demonstrated findings consistent with a diagnosis of HHD. Prior therapies included topical and oral steroids. After a discussion of the risks and benefits, the patient was treated with onabotulinumtoxinA injections in combination with oral glycopyrrolate 5 mg daily. She received 30 U of onabotulinumtoxinA to each axilla, 10 U to each antecubital fossa, and 20 U to the central chest. At 1 month follow-up, the patient reported great improvement in lesion burden and active disease (Figure 2B). Nine months after treatment, her HHD was in complete remission with glycopyrrolate alone and she did not require further therapy with onabotulinumtoxinA.

Hailey-Hailey disease of the chest at presentation and 1 month after initiating treatment with onabotulinumtoxinA and glycopyrrolate, respectively.
FIGURE 2. A and B, Hailey-Hailey disease of the chest at presentation and 1 month after initiating treatment with onabotulinumtoxinA and glycopyrrolate, respectively.

Hailey-Hailey disease has been attributed to mutations of the ATPase secretory pathway Ca2+ transporting 1 gene, ATP2C1, that lead to aberrations in calcium signaling and subsequent impaired adhesion between keratinocytes.2 These compromised cell-cell connections are worsened by the presence of humidity, causing further acantholysis. Chemical denervation of the sweat glands with botulinum toxin has been postulated to improve HHD by reducing moisture in vulnerable areas. Our 2 cases add to the existing literature documenting tangible clinical results that correlate with this hypothesis.3-5

Our second case is unique in that the patient achieved rapid improvement using a combination of onabotulinumtoxinA and glycopyrrolate therapy. Both onabotulinumtoxinA and glycopyrrolate inhibit acetylcholine signaling that is required for sweat production; however, each drug exerts its effect on different zones of the cholinergic pathway, which may partially account for the synergistic effect of onabotulinumtoxinA and glycopyrrolate to improve HHD, as sweating is dually inhibited by the 2 drugs. Additionally, the combined local and systemic administration of these anticholinergic medications may further potentiate the sweat blockade, particularly in areas most prone to disease.

Botulinum toxin for the treatment of HHD is an effective monotherapy. The addition of an oral anticholinergic to local neuromodulator injections may speed symptom resolution and sustain disease remission. Further studies to evaluate this combination are warranted.

References
  1. Palmer DD, Perry HO. Benign familial chronic pemphigus. Arch Dermatol. 1962;86:493-502. doi:10.1001/archderm.1962.01590100107020
  2. Farahnik B, Blattner CM, Mortazie MB, et al. Interventional treatments for Hailey-Hailey disease. J Am Acad Dermatol. 2017;76:551-558.e553. doi:10.1016/j.jaad.2016.08.039
  3. Bessa GR, Glaziovine TC, Manzoni AP, et al. Hailey-Hailey disease treatment with botulinum toxin type A. An Bras Dermatol. 2010;85:717-722. doi:10.1590/s0365-05962010000500021
  4. Lapiere JC, Hirsh A, Gordon KB, et al. Botulinum toxin type A for the treatment of axillary Hailey-Hailey disease. Dermatol Surg. 2000;26:371-374. doi:10.1046/j.1524-4725.2000.99278.x
  5. Koeyers WJ, Van Der Geer S, Krekels G. Botulinum toxin type A as an adjuvant treatment modality for extensive Hailey-Hailey disease. J Dermatolog Treat. 2008;19:251-254. doi:10.1080/09546630801955135
Article PDF
Author and Disclosure Information

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

The authors report no conflict of interest.

Correspondence: Danielle P. Dubin, MD, Department of Dermatology, Icahn School of Medicine at Mount Sinai, 234 E 85th St, 5th Floor, New York, NY 10028 (DPDubin121@gmail.com).

Issue
Cutis - 111(4)
Publications
Topics
Page Number
E28-E29
Sections
Author and Disclosure Information

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

The authors report no conflict of interest.

Correspondence: Danielle P. Dubin, MD, Department of Dermatology, Icahn School of Medicine at Mount Sinai, 234 E 85th St, 5th Floor, New York, NY 10028 (DPDubin121@gmail.com).

Author and Disclosure Information

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

The authors report no conflict of interest.

Correspondence: Danielle P. Dubin, MD, Department of Dermatology, Icahn School of Medicine at Mount Sinai, 234 E 85th St, 5th Floor, New York, NY 10028 (DPDubin121@gmail.com).

Article PDF
Article PDF

To the Editor:

Hailey-Hailey disease (HHD)(also known as familial benign chronic pemphigus) is an inherited autosomal-dominant condition in the family of chronic bullous diseases. It is characterized by flaccid blisters, erosions, and macerated vegetative plaques with a predilection for intertriginous sites. Lesions often are weeping, painful, pruritic, and malodorous, leading to decreased quality of life for patients. Complications of this chronic disease include an increased risk for secondary infection and malignant transformation to squamous cell carcinoma.1

Treatment of HHD remains difficult. Topical steroids, oral steroids, and ablative techniques such as dermabrasion and ablative lasers are the most widely reported therapies. OnabotulinumtoxinA has been described as a successful treatment for patients with HHD, including for disease recalcitrant to other therapies.2 We describe 2 patients with HHD who responded to treatment with intralesional onabotulinumtoxinA injections with and without adjuvant oral glycopyrrolate.

A 54-year-old woman presented with painful flaccid blisters under the breasts (Figure 1A) and in the axillae and groin of 3 weeks’ duration. Biopsy results from this initial visit were consistent with a diagnosis of HHD. The patient reported that the onset of blisters coincided with episodes of severe hyperhidrosis. Therapy with topical and oral steroids, antifungals, antibiotics, and topical aluminum chloride failed to achieve adequate disease control. After a discussion of the risks and benefits, the patient agreed to treatment with injections of onabotulinumtoxinA. At months 0, 3, and 6, the patient received 50 U of onabotulinumtoxinA under the breasts and in the axillae and the groin, for a total of 250 U each session. Each injection consisted of 2.5 U of onabotulinumtoxinA spaced 1-cm apart. Clinical improvement was noted within 2 weeks of initiating neuromodulator therapy. Follow-up at 9 months demonstrated improvement (Figure 1B); however, complete clearance was not achieved, and the patient required ongoing treatment with onabotulinumtoxinA every 3 months.

Hailey-Hailey disease under the breast at presentation and 9 months after initiating treatment with onabotulinumtoxinA, respectively.
FIGURE 1. A and B, Hailey-Hailey disease under the breast at presentation and 9 months after initiating treatment with onabotulinumtoxinA, respectively

A 43-year-old woman presented with erythematous eroded plaques of the antecubital fossae, axillae, and chest (Figure 2A) of 10 years’ duration. A biopsy from an outside provider demonstrated findings consistent with a diagnosis of HHD. Prior therapies included topical and oral steroids. After a discussion of the risks and benefits, the patient was treated with onabotulinumtoxinA injections in combination with oral glycopyrrolate 5 mg daily. She received 30 U of onabotulinumtoxinA to each axilla, 10 U to each antecubital fossa, and 20 U to the central chest. At 1 month follow-up, the patient reported great improvement in lesion burden and active disease (Figure 2B). Nine months after treatment, her HHD was in complete remission with glycopyrrolate alone and she did not require further therapy with onabotulinumtoxinA.

Hailey-Hailey disease of the chest at presentation and 1 month after initiating treatment with onabotulinumtoxinA and glycopyrrolate, respectively.
FIGURE 2. A and B, Hailey-Hailey disease of the chest at presentation and 1 month after initiating treatment with onabotulinumtoxinA and glycopyrrolate, respectively.

Hailey-Hailey disease has been attributed to mutations of the ATPase secretory pathway Ca2+ transporting 1 gene, ATP2C1, that lead to aberrations in calcium signaling and subsequent impaired adhesion between keratinocytes.2 These compromised cell-cell connections are worsened by the presence of humidity, causing further acantholysis. Chemical denervation of the sweat glands with botulinum toxin has been postulated to improve HHD by reducing moisture in vulnerable areas. Our 2 cases add to the existing literature documenting tangible clinical results that correlate with this hypothesis.3-5

Our second case is unique in that the patient achieved rapid improvement using a combination of onabotulinumtoxinA and glycopyrrolate therapy. Both onabotulinumtoxinA and glycopyrrolate inhibit acetylcholine signaling that is required for sweat production; however, each drug exerts its effect on different zones of the cholinergic pathway, which may partially account for the synergistic effect of onabotulinumtoxinA and glycopyrrolate to improve HHD, as sweating is dually inhibited by the 2 drugs. Additionally, the combined local and systemic administration of these anticholinergic medications may further potentiate the sweat blockade, particularly in areas most prone to disease.

Botulinum toxin for the treatment of HHD is an effective monotherapy. The addition of an oral anticholinergic to local neuromodulator injections may speed symptom resolution and sustain disease remission. Further studies to evaluate this combination are warranted.

To the Editor:

Hailey-Hailey disease (HHD)(also known as familial benign chronic pemphigus) is an inherited autosomal-dominant condition in the family of chronic bullous diseases. It is characterized by flaccid blisters, erosions, and macerated vegetative plaques with a predilection for intertriginous sites. Lesions often are weeping, painful, pruritic, and malodorous, leading to decreased quality of life for patients. Complications of this chronic disease include an increased risk for secondary infection and malignant transformation to squamous cell carcinoma.1

Treatment of HHD remains difficult. Topical steroids, oral steroids, and ablative techniques such as dermabrasion and ablative lasers are the most widely reported therapies. OnabotulinumtoxinA has been described as a successful treatment for patients with HHD, including for disease recalcitrant to other therapies.2 We describe 2 patients with HHD who responded to treatment with intralesional onabotulinumtoxinA injections with and without adjuvant oral glycopyrrolate.

A 54-year-old woman presented with painful flaccid blisters under the breasts (Figure 1A) and in the axillae and groin of 3 weeks’ duration. Biopsy results from this initial visit were consistent with a diagnosis of HHD. The patient reported that the onset of blisters coincided with episodes of severe hyperhidrosis. Therapy with topical and oral steroids, antifungals, antibiotics, and topical aluminum chloride failed to achieve adequate disease control. After a discussion of the risks and benefits, the patient agreed to treatment with injections of onabotulinumtoxinA. At months 0, 3, and 6, the patient received 50 U of onabotulinumtoxinA under the breasts and in the axillae and the groin, for a total of 250 U each session. Each injection consisted of 2.5 U of onabotulinumtoxinA spaced 1-cm apart. Clinical improvement was noted within 2 weeks of initiating neuromodulator therapy. Follow-up at 9 months demonstrated improvement (Figure 1B); however, complete clearance was not achieved, and the patient required ongoing treatment with onabotulinumtoxinA every 3 months.

Hailey-Hailey disease under the breast at presentation and 9 months after initiating treatment with onabotulinumtoxinA, respectively.
FIGURE 1. A and B, Hailey-Hailey disease under the breast at presentation and 9 months after initiating treatment with onabotulinumtoxinA, respectively

A 43-year-old woman presented with erythematous eroded plaques of the antecubital fossae, axillae, and chest (Figure 2A) of 10 years’ duration. A biopsy from an outside provider demonstrated findings consistent with a diagnosis of HHD. Prior therapies included topical and oral steroids. After a discussion of the risks and benefits, the patient was treated with onabotulinumtoxinA injections in combination with oral glycopyrrolate 5 mg daily. She received 30 U of onabotulinumtoxinA to each axilla, 10 U to each antecubital fossa, and 20 U to the central chest. At 1 month follow-up, the patient reported great improvement in lesion burden and active disease (Figure 2B). Nine months after treatment, her HHD was in complete remission with glycopyrrolate alone and she did not require further therapy with onabotulinumtoxinA.

Hailey-Hailey disease of the chest at presentation and 1 month after initiating treatment with onabotulinumtoxinA and glycopyrrolate, respectively.
FIGURE 2. A and B, Hailey-Hailey disease of the chest at presentation and 1 month after initiating treatment with onabotulinumtoxinA and glycopyrrolate, respectively.

Hailey-Hailey disease has been attributed to mutations of the ATPase secretory pathway Ca2+ transporting 1 gene, ATP2C1, that lead to aberrations in calcium signaling and subsequent impaired adhesion between keratinocytes.2 These compromised cell-cell connections are worsened by the presence of humidity, causing further acantholysis. Chemical denervation of the sweat glands with botulinum toxin has been postulated to improve HHD by reducing moisture in vulnerable areas. Our 2 cases add to the existing literature documenting tangible clinical results that correlate with this hypothesis.3-5

Our second case is unique in that the patient achieved rapid improvement using a combination of onabotulinumtoxinA and glycopyrrolate therapy. Both onabotulinumtoxinA and glycopyrrolate inhibit acetylcholine signaling that is required for sweat production; however, each drug exerts its effect on different zones of the cholinergic pathway, which may partially account for the synergistic effect of onabotulinumtoxinA and glycopyrrolate to improve HHD, as sweating is dually inhibited by the 2 drugs. Additionally, the combined local and systemic administration of these anticholinergic medications may further potentiate the sweat blockade, particularly in areas most prone to disease.

Botulinum toxin for the treatment of HHD is an effective monotherapy. The addition of an oral anticholinergic to local neuromodulator injections may speed symptom resolution and sustain disease remission. Further studies to evaluate this combination are warranted.

References
  1. Palmer DD, Perry HO. Benign familial chronic pemphigus. Arch Dermatol. 1962;86:493-502. doi:10.1001/archderm.1962.01590100107020
  2. Farahnik B, Blattner CM, Mortazie MB, et al. Interventional treatments for Hailey-Hailey disease. J Am Acad Dermatol. 2017;76:551-558.e553. doi:10.1016/j.jaad.2016.08.039
  3. Bessa GR, Glaziovine TC, Manzoni AP, et al. Hailey-Hailey disease treatment with botulinum toxin type A. An Bras Dermatol. 2010;85:717-722. doi:10.1590/s0365-05962010000500021
  4. Lapiere JC, Hirsh A, Gordon KB, et al. Botulinum toxin type A for the treatment of axillary Hailey-Hailey disease. Dermatol Surg. 2000;26:371-374. doi:10.1046/j.1524-4725.2000.99278.x
  5. Koeyers WJ, Van Der Geer S, Krekels G. Botulinum toxin type A as an adjuvant treatment modality for extensive Hailey-Hailey disease. J Dermatolog Treat. 2008;19:251-254. doi:10.1080/09546630801955135
References
  1. Palmer DD, Perry HO. Benign familial chronic pemphigus. Arch Dermatol. 1962;86:493-502. doi:10.1001/archderm.1962.01590100107020
  2. Farahnik B, Blattner CM, Mortazie MB, et al. Interventional treatments for Hailey-Hailey disease. J Am Acad Dermatol. 2017;76:551-558.e553. doi:10.1016/j.jaad.2016.08.039
  3. Bessa GR, Glaziovine TC, Manzoni AP, et al. Hailey-Hailey disease treatment with botulinum toxin type A. An Bras Dermatol. 2010;85:717-722. doi:10.1590/s0365-05962010000500021
  4. Lapiere JC, Hirsh A, Gordon KB, et al. Botulinum toxin type A for the treatment of axillary Hailey-Hailey disease. Dermatol Surg. 2000;26:371-374. doi:10.1046/j.1524-4725.2000.99278.x
  5. Koeyers WJ, Van Der Geer S, Krekels G. Botulinum toxin type A as an adjuvant treatment modality for extensive Hailey-Hailey disease. J Dermatolog Treat. 2008;19:251-254. doi:10.1080/09546630801955135
Issue
Cutis - 111(4)
Issue
Cutis - 111(4)
Page Number
E28-E29
Page Number
E28-E29
Publications
Publications
Topics
Article Type
Display Headline
Botulinum Toxin and Glycopyrrolate Combination Therapy for Hailey-Hailey Disease
Display Headline
Botulinum Toxin and Glycopyrrolate Combination Therapy for Hailey-Hailey Disease
Sections
Inside the Article

Practice Points

  • Hailey-Hailey disease is associated with decreased quality of life for patients, and current treatment options are limited.
  • A combination of local neuromodulator injections and systemic oral anticholinergic therapy may provide sustained disease remission compared to neuromodulator therapy alone.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Acute Onset of Vitiligolike Depigmentation After Nivolumab Therapy for Systemic Melanoma

Article Type
Changed
Fri, 05/05/2023 - 09:17
Display Headline
Acute Onset of Vitiligolike Depigmentation After Nivolumab Therapy for Systemic Melanoma

To the Editor:

Vitiligolike depigmentation has been known to develop around the sites of origin of melanoma or more rarely in patients treated with antimelanoma therapy.1 Vitiligo is characterized by white patchy depigmentation of the skin caused by the loss of functional melanocytes from the epidermis. The exact mechanisms of disease are unknown and multifactorial; however, autoimmunity plays a central role. Interferon gamma (IFN-γ), C-X-C chemokine ligand 10, and IL-22 have been identified as key mediators in an inflammatory cascade leading to the stimulation of the innate immune response against melanocyte antigens.2,3 Research suggests melanoma-associated vitiligolike leukoderma also results from an immune reaction directed against antigenic determinants shared by both normal and malignant melanocytes.3 Vitiligolike lesions have been associated with the use of immunomodulatory agents such as nivolumab, a fully humanized monoclonal IgG4 antibody, which blocks the programmed cell death protein 1 (PD-1) receptor that normally is expressed on T cells during the effector phase of T-cell activation.4,5 In the tumor microenvironment, the PD-1 receptor is stimulated, leading to downregulation of the T-cell effector function and destruction of T cells.5 Due to T-cell apoptosis and consequent suppression of the immune response, tumorigenesis continues. By inhibiting the PD-1 receptor, nivolumab increases the number of active T cells and antitumor response. However, the distressing side effect of vitiligolike depigmentation has been reported in 15% to 25% of treated patients.6

In a meta-analysis by Teulings et al,7 patients with new-onset vitiligo and malignant melanoma demonstrated a 2-fold decrease in cancer progression and a 4-fold decreased risk for death vs patients without vitiligo development. Thus, in patients with melanoma, vitiligolike depigmentation should be considered a good prognostic indicator as well as a visible sign of spontaneous or therapy-induced antihumoral immune response against melanocyte differentiation antigens, as it is associated with a notable survival benefit in patients receiving immunotherapy for metastatic melanoma.3 We describe a case of diffuse vitiligolike depigmentation that developed suddenly during nivolumab treatment, causing much distress to the patient.

A 75-year-old woman presented to the clinic with a chief concern of sudden diffuse skin discoloration primarily affecting the face, hands, and extremities of 3 weeks’ duration. She had a medical history of metastatic melanoma—the site of the primary melanoma was never identified—and she was undergoing immune-modulating therapy with nivolumab. She was on her fifth month of treatment and was experiencing a robust therapeutic response with a reported 100% clearance of the metastatic melanoma as observed on a positron emission tomography scan. The patchy depigmentation of skin was causing her much distress. Physical examination revealed diffuse patches of hypopigmentation on the trunk, face, and extremities (Figure). Shave biopsies of the right lateral arm demonstrated changes consistent with vitiligo, with an adjacent biopsy illustrating normal skin characteristics. Triamcinolone ointment 0.1% was initiated, with instruction to apply it to affected areas twice daily for 2 weeks. However, there was no improvement, and she discontinued use.

Diffuse hypopigmented patches noted on the patient’s trunk, hands, and legs after nivolumab therapy.
A–C, Diffuse hypopigmented patches noted on the patient’s trunk, hands, and legs after nivolumab therapy.

At 3-month follow-up, the depigmentation persisted, prompting a trial of hydroquinone cream 4% to be used sparingly in cosmetically sensitive areas such as the face and dorsal aspects of the hands. Additionally, diligent photoprotection was advised. Upon re-evaluation 9 months later, the patient remained in cancer remission, continued nivolumab therapy, and reported improvement in the hypopigmentation with a more even skin color with topical hydroquinone use. She no longer noticed starkly contrasting hypopigmented patches.

Vitiligo is a benign skin condition characterized by white depigmented macules and patches. The key feature of the disorder is loss of functional melanocytes from the cutaneous epidermis and sometimes from the hair follicles, with various theories on the cause. It has been suggested that the disease is multifactorial, involving both genetics and environmental factors.2 Regardless of the exact mechanism, the result is always the same: loss of melanin pigment in cells due to loss of melanocytes.

Autoimmunity plays a central role in the causation of vitiligo and was first suspected as a possible cause due to the association of vitiligo with several other autoimmune disorders, such as thyroiditis.8 An epidemiological survey from the United Kingdom and North America (N=2624) found that 19.4% of vitiligo patients aged 20 years or older also reported a clinical history of autoimmune thyroid disease compared with 2.4% of the overall White population of the same age.9 Interferon gamma, C-X-C chemokine ligand 10, and IL-22 receptors stimulate the innate immune response, resulting in an overactive danger signaling cascade, which leads to proinflammatory signals against melanocyte antigens.2,3 The adaptive immune system also participates in the progression of vitiligo by activating dermal dendritic cells to attack melanocytes along with melanocyte-specific cytotoxic T cells.

Immunomodulatory agents utilized in the treatment of metastatic melanoma have been linked to vitiligolike depigmentation. In those receiving PD-1 immunotherapy for metastatic melanoma, vitiligolike lesions have been reported in 15% to 25% of patients.6 Typically, the PD-1 molecule has a regulatory function on effector T cells. Interaction of the PD-1 receptor with its ligands occurs primarily in peripheral tissue causing apoptosis and downregulation of effector T cells with the goal of decreasing collateral damage to surrounding tissues by active T cells.5 In the tumor microenvironment, however, suppression of the host’s immune response is enhanced by aberrant stimulation of the PD-1 receptor, causing downregulation of the T-cell effector function, T-cell destruction, and apoptosis, which results in continued tumor growth. Nivolumab, a fully humanized monoclonal IgG4 antibody, selectively inhibits the PD-1 receptor, disrupting the regulator pathway that would typically end in T-cell destruction.5 Accordingly, the population of active T cells is increased along with the antitumor response.4,10 Nivolumab exhibits success as an immunotherapeutic agent, with an overall survival rate in patients with metastatic melanoma undergoing nivolumab therapy of 41% to 42% at 3 years and 35% at 5 years.11 However, therapeutic manipulation of the host’s immune response does not come without a cost. Vitiligolike lesions have been reported in up to a quarter of patients receiving PD-1 immunotherapy for metastatic melanoma.6

 

 

The relationship between vitiligolike depigmentation and melanoma can be explained by the immune activation against antigens associated with melanoma that also are expressed by normal melanocytes. In clinical observations of patients with melanoma and patients with vitiligo, antibodies to human melanocyte antigens were present in 80% (24/30) of patients vs 7% (2/28) in the control group.12 The autoimmune response results from a cross-reaction of melanoma cells that share the same antigens as normal melanocytes, such as melanoma antigen recognized by T cells 1 (MART-1), gp100, and tyrosinase.13,14

Development of vitiligolike depigmentation in patients with metastatic melanoma treated with nivolumab has been reported to occur between 2 and 15 months after the start of PD-1 therapy. This side effect of treatment correlates with favorable clinical outcomes.15,16 Enhancing immune recognition of melanocytes in patients with melanoma confers a survival advantage, as studies by Koh et al17 and Norlund et al18 involving patients who developed vitiligolike hypopigmentation associated with malignant melanoma indicated a better prognosis than for those without hypopigmentation. The 5-year survival rate of patients with both malignant melanoma and vitiligo was reported as 60% to 67% when it was estimated that only 30% to 50% of patients should have survived that duration of time.17,18 Similarly, a systematic review of patients with melanoma stages III and IV reported that those with associated hypopigmentation had a 2- to 4-fold decreased risk of disease progression and death compared to patients without depigmentation.7

Use of traditional treatment therapies for vitiligo is based on the ability of the therapy to suppress the immune system. However, in patients with metastatic melanoma undergoing immune-modulating cancer therapies, traditional treatment options may counter the antitumor effects of the targeted immunotherapies and should be used with caution. Our patient displayed improvement in the appearance of her starkly contrasting hypopigmented patches with the use of hydroquinone cream 4%, which induced necrotic death of melanocytes by inhibiting the conversion of L-3,4-dihydroxyphenylalanine to melanin by tyrosinase.19 The effect achieved by using topical hydroquinone 4% was a lighter skin appearance in areas of application.

There is no cure for vitiligo, and although it is a benign condition, it can negatively impact a patient's quality of life. In some countries, vitiligo is confused with leprosy, resulting in a social stigma attached to the diagnosis. Many patients are frightened or embarrassed by the diagnosis of vitiligo and its effects, and they often experience discrimination.2 Patients with vitiligo also experience more psychological difficulties such as depression.20 The unpredictability of vitiligo is associated with negative emotions including fear of spreading the lesions, shame, insecurity, and sadness.21 Supportive care measures, including psychological support and counseling, are recommended. Additionally, upon initiation of anti–PD-1 therapies, expectations should be discussed with patients concerning the possibilities of depigmentation and associated treatment results. Although the occurrence of vitiligo may cause the patient concern, it should be communicated that its presence is a positive indicator of a vigorous antimelanoma immunity and an increased survival rate.7

Vitiligolike depigmentation is a known rare adverse effect of nivolumab treatment. Although aesthetically unfavorable for the patient, the development of vitiligolike lesions while undergoing immunotherapy for melanoma may be a sign of a promising clinical outcome due to an effective immune response to melanoma antigens. Our patient remains in remission without any evidence of melanoma after 9 months of therapy, which offers support for a promising outcome for melanoma patients who experience vitiligolike depigmentation.

References
  1. de Golian E, Kwong BY, Swetter SM, et al. Cutaneous complications of targeted melanoma therapy. Curr Treat Options Oncol. 2016;17:57.
  2. Ezzedine K, Eleftheriadou V, Whitton M, et al. Vitiligo. Lancet. 2015;386:74-84.
  3. Ortonne, JP, Passeron, T. Vitiligo and other disorders of hypopigmentation. In: Bolognia J, Schaffer JV, Cerroni L, eds. Dermatology. 4th ed. Elsevier; 2018:1087-1114.
  4. Opdivo. Package insert. Bristol-Myers Squibb Company; 2023.
  5. Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res. 2013;19:5300-5309.
  6. Hwang SJE, Carlos G, Wakade D, et al. Cutaneous adverse events (AEs) of anti-programmed cell death (PD)-1 therapy in patients with metastatic melanoma: a single-institution cohort. J Am Acad Dermatol. 2016;74:455-461.e1.
  7. Teulings HE, Limpens J, Jansen SN, et al. Vitiligo-like depigmentation in patients with stage III-IV melanoma receiving immunotherapy and its association with survival: a systematic review and meta-analysis. J Clin Oncol. 2015;33:773-781.
  8. Gey A, Diallo A, Seneschal J, et al. Autoimmune thyroid disease in vitiligo: multivariate analysis indicates intricate pathomechanisms. Br J Dermatol. 2013;168:756-761.
  9. Alkhateeb A, Fain PR, Thody A, et al. Epidemiology of vitiligo and associated autoimmune diseases in Caucasian probands and their families. Pigment Cell Res. 2003;16:208-214.
  10. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320-330.
  11. Hodi FS, Kluger H, Sznol M, et al. Durable, long-term survival in previously treated patients with advanced melanoma who received nivolumab monotherapy in a phase I trial. Cancer Res. 2016;76(14 suppl):CT001.
  12. Cui J, Bystryn JC. Melanoma and vitiligo are associated with antibody responses to similar antigens on pigment cells. Arch Dermatol. 1995;131:314-318. 
  13. Lynch SA, Bouchard BN, Vijayasaradhi S, et al. Antigens of melanocytes and melanoma. Cancer Metastasis Rev. 1991;10:141-150.
  14. Sanlorenzo M, Vujic I, Daud A, et al. Pembrolizumab cutaneous adverse events and their association with disease progression. JAMA Dermatol. 2015;15:1206-1212.
  15. Hua C, Boussemart L, Mateus C, et al. Association of vitiligo with tumor response in patients with metastatic melanoma treated with pembrolizumab. JAMA Dermatol. 2016;152:45-51.
  16. Nakamura Y, Tanaka R, Asami Y, et al. Correlation between vitiligo occurrence and clinical benefit in advanced melanoma patients treated with nivolumab: a multi-institutional retrospective study. J Dermatol. 2017;44:117-122.
  17. Koh HK, Sober AJ, Nakagawa H, et al. Malignant melanoma and vitiligo-like leukoderma: an electron microscope study. J Am Acad Dermatol. 1983;9:696-708.
  18. Nordlund JJ, Kirkwood JM, Forget BM, et al. Vitiligo in patients with metastatic melanoma: a good prognostic sign. J Am Acad Dermatol. 1983;9:689-696.
  19. Palumbo A, d’Ischia M, Misuraca G, et al. Mechanism of inhibition of melanogenesis by hydroquinone. Biochim Biophys Acta. 1991;1073:85-90.
  20. Lai YC, Yew YW, Kennedy C, et al. Vitiligo and depression: a systematic review and meta-analysis of observational studies. Br J Dermatol. 2017;177:708-718.
  21. Nogueira LSC, Zancanaro PCQ, Azambuja RD. Vitiligo and emotions. An Bras Dermatol. 2009;84:41-45.
Article PDF
Author and Disclosure Information

Ms. Kipers is from Texas A&M University School of Medicine, Bryan. Dr. Mullen is from Northeast Dermatology Associates, Exeter, New Hampshire. Dr. Blumetti is from HonorHealth/Affiliated Dermatology, Scottsdale, Arizona.

The authors report no conflict of interest.

Correspondence: Heidi E.K. Mullen, DO, 23 Hampton Rd, Exeter, NH 03833 (hekipers@gmail.com).

Issue
Cutis - 111(4)
Publications
Topics
Page Number
E15-E18
Sections
Author and Disclosure Information

Ms. Kipers is from Texas A&M University School of Medicine, Bryan. Dr. Mullen is from Northeast Dermatology Associates, Exeter, New Hampshire. Dr. Blumetti is from HonorHealth/Affiliated Dermatology, Scottsdale, Arizona.

The authors report no conflict of interest.

Correspondence: Heidi E.K. Mullen, DO, 23 Hampton Rd, Exeter, NH 03833 (hekipers@gmail.com).

Author and Disclosure Information

Ms. Kipers is from Texas A&M University School of Medicine, Bryan. Dr. Mullen is from Northeast Dermatology Associates, Exeter, New Hampshire. Dr. Blumetti is from HonorHealth/Affiliated Dermatology, Scottsdale, Arizona.

The authors report no conflict of interest.

Correspondence: Heidi E.K. Mullen, DO, 23 Hampton Rd, Exeter, NH 03833 (hekipers@gmail.com).

Article PDF
Article PDF

To the Editor:

Vitiligolike depigmentation has been known to develop around the sites of origin of melanoma or more rarely in patients treated with antimelanoma therapy.1 Vitiligo is characterized by white patchy depigmentation of the skin caused by the loss of functional melanocytes from the epidermis. The exact mechanisms of disease are unknown and multifactorial; however, autoimmunity plays a central role. Interferon gamma (IFN-γ), C-X-C chemokine ligand 10, and IL-22 have been identified as key mediators in an inflammatory cascade leading to the stimulation of the innate immune response against melanocyte antigens.2,3 Research suggests melanoma-associated vitiligolike leukoderma also results from an immune reaction directed against antigenic determinants shared by both normal and malignant melanocytes.3 Vitiligolike lesions have been associated with the use of immunomodulatory agents such as nivolumab, a fully humanized monoclonal IgG4 antibody, which blocks the programmed cell death protein 1 (PD-1) receptor that normally is expressed on T cells during the effector phase of T-cell activation.4,5 In the tumor microenvironment, the PD-1 receptor is stimulated, leading to downregulation of the T-cell effector function and destruction of T cells.5 Due to T-cell apoptosis and consequent suppression of the immune response, tumorigenesis continues. By inhibiting the PD-1 receptor, nivolumab increases the number of active T cells and antitumor response. However, the distressing side effect of vitiligolike depigmentation has been reported in 15% to 25% of treated patients.6

In a meta-analysis by Teulings et al,7 patients with new-onset vitiligo and malignant melanoma demonstrated a 2-fold decrease in cancer progression and a 4-fold decreased risk for death vs patients without vitiligo development. Thus, in patients with melanoma, vitiligolike depigmentation should be considered a good prognostic indicator as well as a visible sign of spontaneous or therapy-induced antihumoral immune response against melanocyte differentiation antigens, as it is associated with a notable survival benefit in patients receiving immunotherapy for metastatic melanoma.3 We describe a case of diffuse vitiligolike depigmentation that developed suddenly during nivolumab treatment, causing much distress to the patient.

A 75-year-old woman presented to the clinic with a chief concern of sudden diffuse skin discoloration primarily affecting the face, hands, and extremities of 3 weeks’ duration. She had a medical history of metastatic melanoma—the site of the primary melanoma was never identified—and she was undergoing immune-modulating therapy with nivolumab. She was on her fifth month of treatment and was experiencing a robust therapeutic response with a reported 100% clearance of the metastatic melanoma as observed on a positron emission tomography scan. The patchy depigmentation of skin was causing her much distress. Physical examination revealed diffuse patches of hypopigmentation on the trunk, face, and extremities (Figure). Shave biopsies of the right lateral arm demonstrated changes consistent with vitiligo, with an adjacent biopsy illustrating normal skin characteristics. Triamcinolone ointment 0.1% was initiated, with instruction to apply it to affected areas twice daily for 2 weeks. However, there was no improvement, and she discontinued use.

Diffuse hypopigmented patches noted on the patient’s trunk, hands, and legs after nivolumab therapy.
A–C, Diffuse hypopigmented patches noted on the patient’s trunk, hands, and legs after nivolumab therapy.

At 3-month follow-up, the depigmentation persisted, prompting a trial of hydroquinone cream 4% to be used sparingly in cosmetically sensitive areas such as the face and dorsal aspects of the hands. Additionally, diligent photoprotection was advised. Upon re-evaluation 9 months later, the patient remained in cancer remission, continued nivolumab therapy, and reported improvement in the hypopigmentation with a more even skin color with topical hydroquinone use. She no longer noticed starkly contrasting hypopigmented patches.

Vitiligo is a benign skin condition characterized by white depigmented macules and patches. The key feature of the disorder is loss of functional melanocytes from the cutaneous epidermis and sometimes from the hair follicles, with various theories on the cause. It has been suggested that the disease is multifactorial, involving both genetics and environmental factors.2 Regardless of the exact mechanism, the result is always the same: loss of melanin pigment in cells due to loss of melanocytes.

Autoimmunity plays a central role in the causation of vitiligo and was first suspected as a possible cause due to the association of vitiligo with several other autoimmune disorders, such as thyroiditis.8 An epidemiological survey from the United Kingdom and North America (N=2624) found that 19.4% of vitiligo patients aged 20 years or older also reported a clinical history of autoimmune thyroid disease compared with 2.4% of the overall White population of the same age.9 Interferon gamma, C-X-C chemokine ligand 10, and IL-22 receptors stimulate the innate immune response, resulting in an overactive danger signaling cascade, which leads to proinflammatory signals against melanocyte antigens.2,3 The adaptive immune system also participates in the progression of vitiligo by activating dermal dendritic cells to attack melanocytes along with melanocyte-specific cytotoxic T cells.

Immunomodulatory agents utilized in the treatment of metastatic melanoma have been linked to vitiligolike depigmentation. In those receiving PD-1 immunotherapy for metastatic melanoma, vitiligolike lesions have been reported in 15% to 25% of patients.6 Typically, the PD-1 molecule has a regulatory function on effector T cells. Interaction of the PD-1 receptor with its ligands occurs primarily in peripheral tissue causing apoptosis and downregulation of effector T cells with the goal of decreasing collateral damage to surrounding tissues by active T cells.5 In the tumor microenvironment, however, suppression of the host’s immune response is enhanced by aberrant stimulation of the PD-1 receptor, causing downregulation of the T-cell effector function, T-cell destruction, and apoptosis, which results in continued tumor growth. Nivolumab, a fully humanized monoclonal IgG4 antibody, selectively inhibits the PD-1 receptor, disrupting the regulator pathway that would typically end in T-cell destruction.5 Accordingly, the population of active T cells is increased along with the antitumor response.4,10 Nivolumab exhibits success as an immunotherapeutic agent, with an overall survival rate in patients with metastatic melanoma undergoing nivolumab therapy of 41% to 42% at 3 years and 35% at 5 years.11 However, therapeutic manipulation of the host’s immune response does not come without a cost. Vitiligolike lesions have been reported in up to a quarter of patients receiving PD-1 immunotherapy for metastatic melanoma.6

 

 

The relationship between vitiligolike depigmentation and melanoma can be explained by the immune activation against antigens associated with melanoma that also are expressed by normal melanocytes. In clinical observations of patients with melanoma and patients with vitiligo, antibodies to human melanocyte antigens were present in 80% (24/30) of patients vs 7% (2/28) in the control group.12 The autoimmune response results from a cross-reaction of melanoma cells that share the same antigens as normal melanocytes, such as melanoma antigen recognized by T cells 1 (MART-1), gp100, and tyrosinase.13,14

Development of vitiligolike depigmentation in patients with metastatic melanoma treated with nivolumab has been reported to occur between 2 and 15 months after the start of PD-1 therapy. This side effect of treatment correlates with favorable clinical outcomes.15,16 Enhancing immune recognition of melanocytes in patients with melanoma confers a survival advantage, as studies by Koh et al17 and Norlund et al18 involving patients who developed vitiligolike hypopigmentation associated with malignant melanoma indicated a better prognosis than for those without hypopigmentation. The 5-year survival rate of patients with both malignant melanoma and vitiligo was reported as 60% to 67% when it was estimated that only 30% to 50% of patients should have survived that duration of time.17,18 Similarly, a systematic review of patients with melanoma stages III and IV reported that those with associated hypopigmentation had a 2- to 4-fold decreased risk of disease progression and death compared to patients without depigmentation.7

Use of traditional treatment therapies for vitiligo is based on the ability of the therapy to suppress the immune system. However, in patients with metastatic melanoma undergoing immune-modulating cancer therapies, traditional treatment options may counter the antitumor effects of the targeted immunotherapies and should be used with caution. Our patient displayed improvement in the appearance of her starkly contrasting hypopigmented patches with the use of hydroquinone cream 4%, which induced necrotic death of melanocytes by inhibiting the conversion of L-3,4-dihydroxyphenylalanine to melanin by tyrosinase.19 The effect achieved by using topical hydroquinone 4% was a lighter skin appearance in areas of application.

There is no cure for vitiligo, and although it is a benign condition, it can negatively impact a patient's quality of life. In some countries, vitiligo is confused with leprosy, resulting in a social stigma attached to the diagnosis. Many patients are frightened or embarrassed by the diagnosis of vitiligo and its effects, and they often experience discrimination.2 Patients with vitiligo also experience more psychological difficulties such as depression.20 The unpredictability of vitiligo is associated with negative emotions including fear of spreading the lesions, shame, insecurity, and sadness.21 Supportive care measures, including psychological support and counseling, are recommended. Additionally, upon initiation of anti–PD-1 therapies, expectations should be discussed with patients concerning the possibilities of depigmentation and associated treatment results. Although the occurrence of vitiligo may cause the patient concern, it should be communicated that its presence is a positive indicator of a vigorous antimelanoma immunity and an increased survival rate.7

Vitiligolike depigmentation is a known rare adverse effect of nivolumab treatment. Although aesthetically unfavorable for the patient, the development of vitiligolike lesions while undergoing immunotherapy for melanoma may be a sign of a promising clinical outcome due to an effective immune response to melanoma antigens. Our patient remains in remission without any evidence of melanoma after 9 months of therapy, which offers support for a promising outcome for melanoma patients who experience vitiligolike depigmentation.

To the Editor:

Vitiligolike depigmentation has been known to develop around the sites of origin of melanoma or more rarely in patients treated with antimelanoma therapy.1 Vitiligo is characterized by white patchy depigmentation of the skin caused by the loss of functional melanocytes from the epidermis. The exact mechanisms of disease are unknown and multifactorial; however, autoimmunity plays a central role. Interferon gamma (IFN-γ), C-X-C chemokine ligand 10, and IL-22 have been identified as key mediators in an inflammatory cascade leading to the stimulation of the innate immune response against melanocyte antigens.2,3 Research suggests melanoma-associated vitiligolike leukoderma also results from an immune reaction directed against antigenic determinants shared by both normal and malignant melanocytes.3 Vitiligolike lesions have been associated with the use of immunomodulatory agents such as nivolumab, a fully humanized monoclonal IgG4 antibody, which blocks the programmed cell death protein 1 (PD-1) receptor that normally is expressed on T cells during the effector phase of T-cell activation.4,5 In the tumor microenvironment, the PD-1 receptor is stimulated, leading to downregulation of the T-cell effector function and destruction of T cells.5 Due to T-cell apoptosis and consequent suppression of the immune response, tumorigenesis continues. By inhibiting the PD-1 receptor, nivolumab increases the number of active T cells and antitumor response. However, the distressing side effect of vitiligolike depigmentation has been reported in 15% to 25% of treated patients.6

In a meta-analysis by Teulings et al,7 patients with new-onset vitiligo and malignant melanoma demonstrated a 2-fold decrease in cancer progression and a 4-fold decreased risk for death vs patients without vitiligo development. Thus, in patients with melanoma, vitiligolike depigmentation should be considered a good prognostic indicator as well as a visible sign of spontaneous or therapy-induced antihumoral immune response against melanocyte differentiation antigens, as it is associated with a notable survival benefit in patients receiving immunotherapy for metastatic melanoma.3 We describe a case of diffuse vitiligolike depigmentation that developed suddenly during nivolumab treatment, causing much distress to the patient.

A 75-year-old woman presented to the clinic with a chief concern of sudden diffuse skin discoloration primarily affecting the face, hands, and extremities of 3 weeks’ duration. She had a medical history of metastatic melanoma—the site of the primary melanoma was never identified—and she was undergoing immune-modulating therapy with nivolumab. She was on her fifth month of treatment and was experiencing a robust therapeutic response with a reported 100% clearance of the metastatic melanoma as observed on a positron emission tomography scan. The patchy depigmentation of skin was causing her much distress. Physical examination revealed diffuse patches of hypopigmentation on the trunk, face, and extremities (Figure). Shave biopsies of the right lateral arm demonstrated changes consistent with vitiligo, with an adjacent biopsy illustrating normal skin characteristics. Triamcinolone ointment 0.1% was initiated, with instruction to apply it to affected areas twice daily for 2 weeks. However, there was no improvement, and she discontinued use.

Diffuse hypopigmented patches noted on the patient’s trunk, hands, and legs after nivolumab therapy.
A–C, Diffuse hypopigmented patches noted on the patient’s trunk, hands, and legs after nivolumab therapy.

At 3-month follow-up, the depigmentation persisted, prompting a trial of hydroquinone cream 4% to be used sparingly in cosmetically sensitive areas such as the face and dorsal aspects of the hands. Additionally, diligent photoprotection was advised. Upon re-evaluation 9 months later, the patient remained in cancer remission, continued nivolumab therapy, and reported improvement in the hypopigmentation with a more even skin color with topical hydroquinone use. She no longer noticed starkly contrasting hypopigmented patches.

Vitiligo is a benign skin condition characterized by white depigmented macules and patches. The key feature of the disorder is loss of functional melanocytes from the cutaneous epidermis and sometimes from the hair follicles, with various theories on the cause. It has been suggested that the disease is multifactorial, involving both genetics and environmental factors.2 Regardless of the exact mechanism, the result is always the same: loss of melanin pigment in cells due to loss of melanocytes.

Autoimmunity plays a central role in the causation of vitiligo and was first suspected as a possible cause due to the association of vitiligo with several other autoimmune disorders, such as thyroiditis.8 An epidemiological survey from the United Kingdom and North America (N=2624) found that 19.4% of vitiligo patients aged 20 years or older also reported a clinical history of autoimmune thyroid disease compared with 2.4% of the overall White population of the same age.9 Interferon gamma, C-X-C chemokine ligand 10, and IL-22 receptors stimulate the innate immune response, resulting in an overactive danger signaling cascade, which leads to proinflammatory signals against melanocyte antigens.2,3 The adaptive immune system also participates in the progression of vitiligo by activating dermal dendritic cells to attack melanocytes along with melanocyte-specific cytotoxic T cells.

Immunomodulatory agents utilized in the treatment of metastatic melanoma have been linked to vitiligolike depigmentation. In those receiving PD-1 immunotherapy for metastatic melanoma, vitiligolike lesions have been reported in 15% to 25% of patients.6 Typically, the PD-1 molecule has a regulatory function on effector T cells. Interaction of the PD-1 receptor with its ligands occurs primarily in peripheral tissue causing apoptosis and downregulation of effector T cells with the goal of decreasing collateral damage to surrounding tissues by active T cells.5 In the tumor microenvironment, however, suppression of the host’s immune response is enhanced by aberrant stimulation of the PD-1 receptor, causing downregulation of the T-cell effector function, T-cell destruction, and apoptosis, which results in continued tumor growth. Nivolumab, a fully humanized monoclonal IgG4 antibody, selectively inhibits the PD-1 receptor, disrupting the regulator pathway that would typically end in T-cell destruction.5 Accordingly, the population of active T cells is increased along with the antitumor response.4,10 Nivolumab exhibits success as an immunotherapeutic agent, with an overall survival rate in patients with metastatic melanoma undergoing nivolumab therapy of 41% to 42% at 3 years and 35% at 5 years.11 However, therapeutic manipulation of the host’s immune response does not come without a cost. Vitiligolike lesions have been reported in up to a quarter of patients receiving PD-1 immunotherapy for metastatic melanoma.6

 

 

The relationship between vitiligolike depigmentation and melanoma can be explained by the immune activation against antigens associated with melanoma that also are expressed by normal melanocytes. In clinical observations of patients with melanoma and patients with vitiligo, antibodies to human melanocyte antigens were present in 80% (24/30) of patients vs 7% (2/28) in the control group.12 The autoimmune response results from a cross-reaction of melanoma cells that share the same antigens as normal melanocytes, such as melanoma antigen recognized by T cells 1 (MART-1), gp100, and tyrosinase.13,14

Development of vitiligolike depigmentation in patients with metastatic melanoma treated with nivolumab has been reported to occur between 2 and 15 months after the start of PD-1 therapy. This side effect of treatment correlates with favorable clinical outcomes.15,16 Enhancing immune recognition of melanocytes in patients with melanoma confers a survival advantage, as studies by Koh et al17 and Norlund et al18 involving patients who developed vitiligolike hypopigmentation associated with malignant melanoma indicated a better prognosis than for those without hypopigmentation. The 5-year survival rate of patients with both malignant melanoma and vitiligo was reported as 60% to 67% when it was estimated that only 30% to 50% of patients should have survived that duration of time.17,18 Similarly, a systematic review of patients with melanoma stages III and IV reported that those with associated hypopigmentation had a 2- to 4-fold decreased risk of disease progression and death compared to patients without depigmentation.7

Use of traditional treatment therapies for vitiligo is based on the ability of the therapy to suppress the immune system. However, in patients with metastatic melanoma undergoing immune-modulating cancer therapies, traditional treatment options may counter the antitumor effects of the targeted immunotherapies and should be used with caution. Our patient displayed improvement in the appearance of her starkly contrasting hypopigmented patches with the use of hydroquinone cream 4%, which induced necrotic death of melanocytes by inhibiting the conversion of L-3,4-dihydroxyphenylalanine to melanin by tyrosinase.19 The effect achieved by using topical hydroquinone 4% was a lighter skin appearance in areas of application.

There is no cure for vitiligo, and although it is a benign condition, it can negatively impact a patient's quality of life. In some countries, vitiligo is confused with leprosy, resulting in a social stigma attached to the diagnosis. Many patients are frightened or embarrassed by the diagnosis of vitiligo and its effects, and they often experience discrimination.2 Patients with vitiligo also experience more psychological difficulties such as depression.20 The unpredictability of vitiligo is associated with negative emotions including fear of spreading the lesions, shame, insecurity, and sadness.21 Supportive care measures, including psychological support and counseling, are recommended. Additionally, upon initiation of anti–PD-1 therapies, expectations should be discussed with patients concerning the possibilities of depigmentation and associated treatment results. Although the occurrence of vitiligo may cause the patient concern, it should be communicated that its presence is a positive indicator of a vigorous antimelanoma immunity and an increased survival rate.7

Vitiligolike depigmentation is a known rare adverse effect of nivolumab treatment. Although aesthetically unfavorable for the patient, the development of vitiligolike lesions while undergoing immunotherapy for melanoma may be a sign of a promising clinical outcome due to an effective immune response to melanoma antigens. Our patient remains in remission without any evidence of melanoma after 9 months of therapy, which offers support for a promising outcome for melanoma patients who experience vitiligolike depigmentation.

References
  1. de Golian E, Kwong BY, Swetter SM, et al. Cutaneous complications of targeted melanoma therapy. Curr Treat Options Oncol. 2016;17:57.
  2. Ezzedine K, Eleftheriadou V, Whitton M, et al. Vitiligo. Lancet. 2015;386:74-84.
  3. Ortonne, JP, Passeron, T. Vitiligo and other disorders of hypopigmentation. In: Bolognia J, Schaffer JV, Cerroni L, eds. Dermatology. 4th ed. Elsevier; 2018:1087-1114.
  4. Opdivo. Package insert. Bristol-Myers Squibb Company; 2023.
  5. Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res. 2013;19:5300-5309.
  6. Hwang SJE, Carlos G, Wakade D, et al. Cutaneous adverse events (AEs) of anti-programmed cell death (PD)-1 therapy in patients with metastatic melanoma: a single-institution cohort. J Am Acad Dermatol. 2016;74:455-461.e1.
  7. Teulings HE, Limpens J, Jansen SN, et al. Vitiligo-like depigmentation in patients with stage III-IV melanoma receiving immunotherapy and its association with survival: a systematic review and meta-analysis. J Clin Oncol. 2015;33:773-781.
  8. Gey A, Diallo A, Seneschal J, et al. Autoimmune thyroid disease in vitiligo: multivariate analysis indicates intricate pathomechanisms. Br J Dermatol. 2013;168:756-761.
  9. Alkhateeb A, Fain PR, Thody A, et al. Epidemiology of vitiligo and associated autoimmune diseases in Caucasian probands and their families. Pigment Cell Res. 2003;16:208-214.
  10. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320-330.
  11. Hodi FS, Kluger H, Sznol M, et al. Durable, long-term survival in previously treated patients with advanced melanoma who received nivolumab monotherapy in a phase I trial. Cancer Res. 2016;76(14 suppl):CT001.
  12. Cui J, Bystryn JC. Melanoma and vitiligo are associated with antibody responses to similar antigens on pigment cells. Arch Dermatol. 1995;131:314-318. 
  13. Lynch SA, Bouchard BN, Vijayasaradhi S, et al. Antigens of melanocytes and melanoma. Cancer Metastasis Rev. 1991;10:141-150.
  14. Sanlorenzo M, Vujic I, Daud A, et al. Pembrolizumab cutaneous adverse events and their association with disease progression. JAMA Dermatol. 2015;15:1206-1212.
  15. Hua C, Boussemart L, Mateus C, et al. Association of vitiligo with tumor response in patients with metastatic melanoma treated with pembrolizumab. JAMA Dermatol. 2016;152:45-51.
  16. Nakamura Y, Tanaka R, Asami Y, et al. Correlation between vitiligo occurrence and clinical benefit in advanced melanoma patients treated with nivolumab: a multi-institutional retrospective study. J Dermatol. 2017;44:117-122.
  17. Koh HK, Sober AJ, Nakagawa H, et al. Malignant melanoma and vitiligo-like leukoderma: an electron microscope study. J Am Acad Dermatol. 1983;9:696-708.
  18. Nordlund JJ, Kirkwood JM, Forget BM, et al. Vitiligo in patients with metastatic melanoma: a good prognostic sign. J Am Acad Dermatol. 1983;9:689-696.
  19. Palumbo A, d’Ischia M, Misuraca G, et al. Mechanism of inhibition of melanogenesis by hydroquinone. Biochim Biophys Acta. 1991;1073:85-90.
  20. Lai YC, Yew YW, Kennedy C, et al. Vitiligo and depression: a systematic review and meta-analysis of observational studies. Br J Dermatol. 2017;177:708-718.
  21. Nogueira LSC, Zancanaro PCQ, Azambuja RD. Vitiligo and emotions. An Bras Dermatol. 2009;84:41-45.
References
  1. de Golian E, Kwong BY, Swetter SM, et al. Cutaneous complications of targeted melanoma therapy. Curr Treat Options Oncol. 2016;17:57.
  2. Ezzedine K, Eleftheriadou V, Whitton M, et al. Vitiligo. Lancet. 2015;386:74-84.
  3. Ortonne, JP, Passeron, T. Vitiligo and other disorders of hypopigmentation. In: Bolognia J, Schaffer JV, Cerroni L, eds. Dermatology. 4th ed. Elsevier; 2018:1087-1114.
  4. Opdivo. Package insert. Bristol-Myers Squibb Company; 2023.
  5. Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res. 2013;19:5300-5309.
  6. Hwang SJE, Carlos G, Wakade D, et al. Cutaneous adverse events (AEs) of anti-programmed cell death (PD)-1 therapy in patients with metastatic melanoma: a single-institution cohort. J Am Acad Dermatol. 2016;74:455-461.e1.
  7. Teulings HE, Limpens J, Jansen SN, et al. Vitiligo-like depigmentation in patients with stage III-IV melanoma receiving immunotherapy and its association with survival: a systematic review and meta-analysis. J Clin Oncol. 2015;33:773-781.
  8. Gey A, Diallo A, Seneschal J, et al. Autoimmune thyroid disease in vitiligo: multivariate analysis indicates intricate pathomechanisms. Br J Dermatol. 2013;168:756-761.
  9. Alkhateeb A, Fain PR, Thody A, et al. Epidemiology of vitiligo and associated autoimmune diseases in Caucasian probands and their families. Pigment Cell Res. 2003;16:208-214.
  10. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320-330.
  11. Hodi FS, Kluger H, Sznol M, et al. Durable, long-term survival in previously treated patients with advanced melanoma who received nivolumab monotherapy in a phase I trial. Cancer Res. 2016;76(14 suppl):CT001.
  12. Cui J, Bystryn JC. Melanoma and vitiligo are associated with antibody responses to similar antigens on pigment cells. Arch Dermatol. 1995;131:314-318. 
  13. Lynch SA, Bouchard BN, Vijayasaradhi S, et al. Antigens of melanocytes and melanoma. Cancer Metastasis Rev. 1991;10:141-150.
  14. Sanlorenzo M, Vujic I, Daud A, et al. Pembrolizumab cutaneous adverse events and their association with disease progression. JAMA Dermatol. 2015;15:1206-1212.
  15. Hua C, Boussemart L, Mateus C, et al. Association of vitiligo with tumor response in patients with metastatic melanoma treated with pembrolizumab. JAMA Dermatol. 2016;152:45-51.
  16. Nakamura Y, Tanaka R, Asami Y, et al. Correlation between vitiligo occurrence and clinical benefit in advanced melanoma patients treated with nivolumab: a multi-institutional retrospective study. J Dermatol. 2017;44:117-122.
  17. Koh HK, Sober AJ, Nakagawa H, et al. Malignant melanoma and vitiligo-like leukoderma: an electron microscope study. J Am Acad Dermatol. 1983;9:696-708.
  18. Nordlund JJ, Kirkwood JM, Forget BM, et al. Vitiligo in patients with metastatic melanoma: a good prognostic sign. J Am Acad Dermatol. 1983;9:689-696.
  19. Palumbo A, d’Ischia M, Misuraca G, et al. Mechanism of inhibition of melanogenesis by hydroquinone. Biochim Biophys Acta. 1991;1073:85-90.
  20. Lai YC, Yew YW, Kennedy C, et al. Vitiligo and depression: a systematic review and meta-analysis of observational studies. Br J Dermatol. 2017;177:708-718.
  21. Nogueira LSC, Zancanaro PCQ, Azambuja RD. Vitiligo and emotions. An Bras Dermatol. 2009;84:41-45.
Issue
Cutis - 111(4)
Issue
Cutis - 111(4)
Page Number
E15-E18
Page Number
E15-E18
Publications
Publications
Topics
Article Type
Display Headline
Acute Onset of Vitiligolike Depigmentation After Nivolumab Therapy for Systemic Melanoma
Display Headline
Acute Onset of Vitiligolike Depigmentation After Nivolumab Therapy for Systemic Melanoma
Sections
Inside the Article

Practice Points

  • New-onset vitiligo coinciding with malignant melanoma should be considered a good prognostic indicator.
  • Daily use of hydroquinone cream 4% in conjunction with diligent photoprotection was shown to even overall skin tone in a patient experiencing leukoderma from nivolumab therapy.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Collision Course of a Basal Cell Carcinoma and Apocrine Hidrocystoma on the Scalp

Article Type
Changed
Thu, 04/20/2023 - 09:50
Display Headline
Collision Course of a Basal Cell Carcinoma and Apocrine Hidrocystoma on the Scalp

To the Editor:

A collision tumor is the coexistence of 2 discrete tumors in the same neoplasm, possibly comprising a malignant tumor and a benign tumor, and thereby complicating appropriate diagnosis and treatment. We present a case of a basal cell carcinoma (BCC) of the scalp that was later found to be in collision with an apocrine hidrocystoma that might have arisen from a nevus sebaceus. Although rare, BCC can coexist with apocrine hidrocystoma. Jayaprakasam and Rene1 reported a case of a collision tumor containing BCC and hidrocystoma on the eyelid.1 We present a case of a BCC on the scalp that was later found to be in collision with an apocrine hidrocystoma that possibly arose from a nevus sebaceus.

A nodular and pigmented basal cell carcinoma in a 92-year-old Black woman (H&E, original magnification ×40).
FIGURE 1. A nodular and pigmented basal cell carcinoma in a 92-year-old Black woman (H&E, original magnification ×40).

A 92-year-old Black woman with a biopsy-confirmed primary BCC of the left parietal scalp presented for Mohs micrographic surgery. The pathology report from an outside facility was reviewed. The initial diagnosis had been made with 2 punch biopsies from separate areas of the large nodule—one consistent with nodular and pigmented BCC (Figure 1), and the other revealed nodular ulcerated BCC. Physical examination prior to Mohs surgery revealed a mobile, flesh-colored, 6.2×6.0-cm nodule with minimal overlying hair on the left parietal scalp (Figure 2). During stage-I processing by the histopathology laboratory, large cystic structures were encountered; en face frozen sections showed a cystic tumor. Excised tissue was submitted for permanent processing to aid in diagnosis; the initial diagnostic biopsy slides were requested from the outside facility for review.

A 6.2×6.0-cm flesh-colored nodule on the scalp was examined prior to Mohs micrographic surgery.
FIGURE 2. A 6.2×6.0-cm flesh-colored nodule on the scalp was examined prior to Mohs micrographic surgery.

The initial diagnostic biopsy slides were reviewed and found to be consistent with nodular and pigmented BCC, as previously reported. Findings from hematoxylin and eosin staining of tissue obtained from Mohs sections were consistent with a combined neoplasm comprising BCC (Figure 3A) and apocrine hidrocystoma (Figure 3B). In addition, one section was characterized by acanthosis, papillomatosis, and sebaceous glands—similar to findings that are seen in a nevus sebaceus (Figure 3C).

A, Histopathology of Mohs section revealed nests of palisading blue cells with fibrous stroma in the dermis, consistent with a basal cell carcinoma (H&E, original magnification ×200)
FIGURE 3. A, Histopathology of Mohs section revealed nests of palisading blue cells with fibrous stroma in the dermis, consistent with a basal cell carcinoma (H&E, original magnification ×200). B, A cystic space lined by a double layer of epithelial cells with secretion through decapitation (arrow) and numerous papillary projections into the central cavity was consistent with an apocrine hidrocystoma (H&E, original magnification ×200). C, Acanthosis, papillomatosis, and sebaceous glands were identified in another Mohs section—similar to findings in a nevus sebaceus (H&E, original magnification ×40).

The BCC was cleared after stage I; the final wound size was 7×6.6 cm. Although benign apocrine hidrocystoma was still evident at the margin, further excision was not performed at the request of the patient and her family. Partial primary wound closure was performed with pulley sutures. A xenograft was placed over the unclosed central portion. The wound was permitted to heal by second intention.

The clinical differential diagnosis of a scalp nodule includes a pilar cyst, BCC, squamous cell carcinoma, melanoma, cutaneous metastasis, adnexal tumor, atypical fibroxanthoma, and collision tumor. A collision tumor—the association of 2 or more benign or malignant neoplasms—represents a well-known pitfall in making a correct clinical and pathologic diagnosis.2 Many theories have been proposed to explain the pathophysiology of collision tumors. Some authors have speculated that they arise from involvement of related cell types.1 Other theories include induction by cytokines and growth factors secreted from one tumor that provides an ideal environment for proliferation of other cell types, a field cancerization effect of sun-damaged skin, or a coincidence.2

In our case, it is possible that the 2 tumors arose from a nevus sebaceus. One retrospective study of 706 cases of nevus sebaceus (707 specimens) found that 22.5% of cases developed secondary proliferation; of those cases, 18.9% were benign.3 Additionally, in 4.2% of cases of nevus sebaceus, proliferation of 2 or more tumors developed. The most common malignant neoplasm to develop from nevus sebaceus was BCC, followed by squamous cell carcinoma and sebaceous carcinoma. The most common benign neoplasm to develop from nevus sebaceus was trichoblastoma, followed by syringocystadenoma papilliferum.3

Our case highlights the possibility of a sampling error when performing a biopsy of any large neoplasm. Additionally, Mohs surgeons should maintain high clinical suspicion for collision tumors when encountering a large tumor with pathology inconsistent with the original biopsy. Apocrine hidrocystoma should be considered in the differential diagnosis of a large cystic mass of the scalp. Also, it is important to recognize that malignant lesions, such as BCC, can coexist with another benign tumor. Basal cell carcinoma is rare in Black patients, supporting our belief that our patient’s tumors arose from a nevus sebaceus.

It also is important for Mohs surgeons to consider any potential discrepancy between the initial pathology report and Mohs intraoperative pathology that can impact diagnosis, the aggressiveness of the tumors identified, and how such aggressiveness may affect management options.4,5 Some dermatology practices request biopsy slides from patients who are referred for Mohs micrographic surgery for internal review by a dermatopathologist before surgery is performed; however, this protocol requires additional time and adds costs for the overall health care system.4 One study found that internal review of outside biopsy slides resulted in a change in diagnosis in 2.2% of patients (N=3345)—affecting management in 61% of cases in which the diagnosis was changed.4 Another study (N=163) found that the reported aggressiveness of 50.5% of nonmelanoma cases in an initial biopsy report was changed during Mohs micrographic surgery.5 Mohs surgeons should be aware that discrepancies can occur, and if a discrepancy is discovered, the procedure may be paused until the initial biopsy slide is reviewed and further information is collected.

References
  1. Jayaprakasam A, Rene C. A benign or malignant eyelid lump—can you tell? an unusual collision tumour highlighting the difficulty differentiating a hidrocystoma from a basal cell carcinoma. BMJ Case Reports. 2012;2012:bcr1220115307. doi:10.1136/bcr.12.2011.5307
  2. Miteva M, Herschthal D, Ricotti C, et al. A rare case of a cutaneous squamomelanocytic tumor: revisiting the histogenesis of combined neoplasms. Am J Dermatopathol. 2009;31:599-603. doi:10.1097/DAD.0b013e3181a88116
  3. Idriss MH, Elston DM. Secondary neoplasms associated with nevus sebaceus of Jadassohn: a study of 707 cases. J Am Acad Dermatol. 2014;70:332-337. doi:10.1016/j.jaad.2013.10.004
  4. Butler ST, Youker SR, Mandrell J, et al. The importance of reviewing pathology specimens before Mohs surgery. Dermatol Surg. 2009;35:407-412. doi:10.1111/j.1524-4725.2008.01056.x
  5. Stiegel E, Lam C, Schowalter M, et al. Correlation between original biopsy pathology and Mohs intraoperative pathology. Dermatol Surg. 2018;44:193-197. doi:10.1097/DSS.0000000000001276
Article PDF
Author and Disclosure Information

From the Department of Dermatology, University of Texas Medical Branch, Galveston.

The authors report no conflict of interest.

This case was presented at the American Society for Dermatologic Surgery Annual Meeting; October 24-27, 2019; Chicago, Illinois.

Correspondence: Kristyna L. Iyer, MD (kiyer@usdermpartners.com).

Issue
Cutis - 111(4)
Publications
Topics
Page Number
E9-E11
Sections
Author and Disclosure Information

From the Department of Dermatology, University of Texas Medical Branch, Galveston.

The authors report no conflict of interest.

This case was presented at the American Society for Dermatologic Surgery Annual Meeting; October 24-27, 2019; Chicago, Illinois.

Correspondence: Kristyna L. Iyer, MD (kiyer@usdermpartners.com).

Author and Disclosure Information

From the Department of Dermatology, University of Texas Medical Branch, Galveston.

The authors report no conflict of interest.

This case was presented at the American Society for Dermatologic Surgery Annual Meeting; October 24-27, 2019; Chicago, Illinois.

Correspondence: Kristyna L. Iyer, MD (kiyer@usdermpartners.com).

Article PDF
Article PDF

To the Editor:

A collision tumor is the coexistence of 2 discrete tumors in the same neoplasm, possibly comprising a malignant tumor and a benign tumor, and thereby complicating appropriate diagnosis and treatment. We present a case of a basal cell carcinoma (BCC) of the scalp that was later found to be in collision with an apocrine hidrocystoma that might have arisen from a nevus sebaceus. Although rare, BCC can coexist with apocrine hidrocystoma. Jayaprakasam and Rene1 reported a case of a collision tumor containing BCC and hidrocystoma on the eyelid.1 We present a case of a BCC on the scalp that was later found to be in collision with an apocrine hidrocystoma that possibly arose from a nevus sebaceus.

A nodular and pigmented basal cell carcinoma in a 92-year-old Black woman (H&E, original magnification ×40).
FIGURE 1. A nodular and pigmented basal cell carcinoma in a 92-year-old Black woman (H&E, original magnification ×40).

A 92-year-old Black woman with a biopsy-confirmed primary BCC of the left parietal scalp presented for Mohs micrographic surgery. The pathology report from an outside facility was reviewed. The initial diagnosis had been made with 2 punch biopsies from separate areas of the large nodule—one consistent with nodular and pigmented BCC (Figure 1), and the other revealed nodular ulcerated BCC. Physical examination prior to Mohs surgery revealed a mobile, flesh-colored, 6.2×6.0-cm nodule with minimal overlying hair on the left parietal scalp (Figure 2). During stage-I processing by the histopathology laboratory, large cystic structures were encountered; en face frozen sections showed a cystic tumor. Excised tissue was submitted for permanent processing to aid in diagnosis; the initial diagnostic biopsy slides were requested from the outside facility for review.

A 6.2×6.0-cm flesh-colored nodule on the scalp was examined prior to Mohs micrographic surgery.
FIGURE 2. A 6.2×6.0-cm flesh-colored nodule on the scalp was examined prior to Mohs micrographic surgery.

The initial diagnostic biopsy slides were reviewed and found to be consistent with nodular and pigmented BCC, as previously reported. Findings from hematoxylin and eosin staining of tissue obtained from Mohs sections were consistent with a combined neoplasm comprising BCC (Figure 3A) and apocrine hidrocystoma (Figure 3B). In addition, one section was characterized by acanthosis, papillomatosis, and sebaceous glands—similar to findings that are seen in a nevus sebaceus (Figure 3C).

A, Histopathology of Mohs section revealed nests of palisading blue cells with fibrous stroma in the dermis, consistent with a basal cell carcinoma (H&E, original magnification ×200)
FIGURE 3. A, Histopathology of Mohs section revealed nests of palisading blue cells with fibrous stroma in the dermis, consistent with a basal cell carcinoma (H&E, original magnification ×200). B, A cystic space lined by a double layer of epithelial cells with secretion through decapitation (arrow) and numerous papillary projections into the central cavity was consistent with an apocrine hidrocystoma (H&E, original magnification ×200). C, Acanthosis, papillomatosis, and sebaceous glands were identified in another Mohs section—similar to findings in a nevus sebaceus (H&E, original magnification ×40).

The BCC was cleared after stage I; the final wound size was 7×6.6 cm. Although benign apocrine hidrocystoma was still evident at the margin, further excision was not performed at the request of the patient and her family. Partial primary wound closure was performed with pulley sutures. A xenograft was placed over the unclosed central portion. The wound was permitted to heal by second intention.

The clinical differential diagnosis of a scalp nodule includes a pilar cyst, BCC, squamous cell carcinoma, melanoma, cutaneous metastasis, adnexal tumor, atypical fibroxanthoma, and collision tumor. A collision tumor—the association of 2 or more benign or malignant neoplasms—represents a well-known pitfall in making a correct clinical and pathologic diagnosis.2 Many theories have been proposed to explain the pathophysiology of collision tumors. Some authors have speculated that they arise from involvement of related cell types.1 Other theories include induction by cytokines and growth factors secreted from one tumor that provides an ideal environment for proliferation of other cell types, a field cancerization effect of sun-damaged skin, or a coincidence.2

In our case, it is possible that the 2 tumors arose from a nevus sebaceus. One retrospective study of 706 cases of nevus sebaceus (707 specimens) found that 22.5% of cases developed secondary proliferation; of those cases, 18.9% were benign.3 Additionally, in 4.2% of cases of nevus sebaceus, proliferation of 2 or more tumors developed. The most common malignant neoplasm to develop from nevus sebaceus was BCC, followed by squamous cell carcinoma and sebaceous carcinoma. The most common benign neoplasm to develop from nevus sebaceus was trichoblastoma, followed by syringocystadenoma papilliferum.3

Our case highlights the possibility of a sampling error when performing a biopsy of any large neoplasm. Additionally, Mohs surgeons should maintain high clinical suspicion for collision tumors when encountering a large tumor with pathology inconsistent with the original biopsy. Apocrine hidrocystoma should be considered in the differential diagnosis of a large cystic mass of the scalp. Also, it is important to recognize that malignant lesions, such as BCC, can coexist with another benign tumor. Basal cell carcinoma is rare in Black patients, supporting our belief that our patient’s tumors arose from a nevus sebaceus.

It also is important for Mohs surgeons to consider any potential discrepancy between the initial pathology report and Mohs intraoperative pathology that can impact diagnosis, the aggressiveness of the tumors identified, and how such aggressiveness may affect management options.4,5 Some dermatology practices request biopsy slides from patients who are referred for Mohs micrographic surgery for internal review by a dermatopathologist before surgery is performed; however, this protocol requires additional time and adds costs for the overall health care system.4 One study found that internal review of outside biopsy slides resulted in a change in diagnosis in 2.2% of patients (N=3345)—affecting management in 61% of cases in which the diagnosis was changed.4 Another study (N=163) found that the reported aggressiveness of 50.5% of nonmelanoma cases in an initial biopsy report was changed during Mohs micrographic surgery.5 Mohs surgeons should be aware that discrepancies can occur, and if a discrepancy is discovered, the procedure may be paused until the initial biopsy slide is reviewed and further information is collected.

To the Editor:

A collision tumor is the coexistence of 2 discrete tumors in the same neoplasm, possibly comprising a malignant tumor and a benign tumor, and thereby complicating appropriate diagnosis and treatment. We present a case of a basal cell carcinoma (BCC) of the scalp that was later found to be in collision with an apocrine hidrocystoma that might have arisen from a nevus sebaceus. Although rare, BCC can coexist with apocrine hidrocystoma. Jayaprakasam and Rene1 reported a case of a collision tumor containing BCC and hidrocystoma on the eyelid.1 We present a case of a BCC on the scalp that was later found to be in collision with an apocrine hidrocystoma that possibly arose from a nevus sebaceus.

A nodular and pigmented basal cell carcinoma in a 92-year-old Black woman (H&E, original magnification ×40).
FIGURE 1. A nodular and pigmented basal cell carcinoma in a 92-year-old Black woman (H&E, original magnification ×40).

A 92-year-old Black woman with a biopsy-confirmed primary BCC of the left parietal scalp presented for Mohs micrographic surgery. The pathology report from an outside facility was reviewed. The initial diagnosis had been made with 2 punch biopsies from separate areas of the large nodule—one consistent with nodular and pigmented BCC (Figure 1), and the other revealed nodular ulcerated BCC. Physical examination prior to Mohs surgery revealed a mobile, flesh-colored, 6.2×6.0-cm nodule with minimal overlying hair on the left parietal scalp (Figure 2). During stage-I processing by the histopathology laboratory, large cystic structures were encountered; en face frozen sections showed a cystic tumor. Excised tissue was submitted for permanent processing to aid in diagnosis; the initial diagnostic biopsy slides were requested from the outside facility for review.

A 6.2×6.0-cm flesh-colored nodule on the scalp was examined prior to Mohs micrographic surgery.
FIGURE 2. A 6.2×6.0-cm flesh-colored nodule on the scalp was examined prior to Mohs micrographic surgery.

The initial diagnostic biopsy slides were reviewed and found to be consistent with nodular and pigmented BCC, as previously reported. Findings from hematoxylin and eosin staining of tissue obtained from Mohs sections were consistent with a combined neoplasm comprising BCC (Figure 3A) and apocrine hidrocystoma (Figure 3B). In addition, one section was characterized by acanthosis, papillomatosis, and sebaceous glands—similar to findings that are seen in a nevus sebaceus (Figure 3C).

A, Histopathology of Mohs section revealed nests of palisading blue cells with fibrous stroma in the dermis, consistent with a basal cell carcinoma (H&E, original magnification ×200)
FIGURE 3. A, Histopathology of Mohs section revealed nests of palisading blue cells with fibrous stroma in the dermis, consistent with a basal cell carcinoma (H&E, original magnification ×200). B, A cystic space lined by a double layer of epithelial cells with secretion through decapitation (arrow) and numerous papillary projections into the central cavity was consistent with an apocrine hidrocystoma (H&E, original magnification ×200). C, Acanthosis, papillomatosis, and sebaceous glands were identified in another Mohs section—similar to findings in a nevus sebaceus (H&E, original magnification ×40).

The BCC was cleared after stage I; the final wound size was 7×6.6 cm. Although benign apocrine hidrocystoma was still evident at the margin, further excision was not performed at the request of the patient and her family. Partial primary wound closure was performed with pulley sutures. A xenograft was placed over the unclosed central portion. The wound was permitted to heal by second intention.

The clinical differential diagnosis of a scalp nodule includes a pilar cyst, BCC, squamous cell carcinoma, melanoma, cutaneous metastasis, adnexal tumor, atypical fibroxanthoma, and collision tumor. A collision tumor—the association of 2 or more benign or malignant neoplasms—represents a well-known pitfall in making a correct clinical and pathologic diagnosis.2 Many theories have been proposed to explain the pathophysiology of collision tumors. Some authors have speculated that they arise from involvement of related cell types.1 Other theories include induction by cytokines and growth factors secreted from one tumor that provides an ideal environment for proliferation of other cell types, a field cancerization effect of sun-damaged skin, or a coincidence.2

In our case, it is possible that the 2 tumors arose from a nevus sebaceus. One retrospective study of 706 cases of nevus sebaceus (707 specimens) found that 22.5% of cases developed secondary proliferation; of those cases, 18.9% were benign.3 Additionally, in 4.2% of cases of nevus sebaceus, proliferation of 2 or more tumors developed. The most common malignant neoplasm to develop from nevus sebaceus was BCC, followed by squamous cell carcinoma and sebaceous carcinoma. The most common benign neoplasm to develop from nevus sebaceus was trichoblastoma, followed by syringocystadenoma papilliferum.3

Our case highlights the possibility of a sampling error when performing a biopsy of any large neoplasm. Additionally, Mohs surgeons should maintain high clinical suspicion for collision tumors when encountering a large tumor with pathology inconsistent with the original biopsy. Apocrine hidrocystoma should be considered in the differential diagnosis of a large cystic mass of the scalp. Also, it is important to recognize that malignant lesions, such as BCC, can coexist with another benign tumor. Basal cell carcinoma is rare in Black patients, supporting our belief that our patient’s tumors arose from a nevus sebaceus.

It also is important for Mohs surgeons to consider any potential discrepancy between the initial pathology report and Mohs intraoperative pathology that can impact diagnosis, the aggressiveness of the tumors identified, and how such aggressiveness may affect management options.4,5 Some dermatology practices request biopsy slides from patients who are referred for Mohs micrographic surgery for internal review by a dermatopathologist before surgery is performed; however, this protocol requires additional time and adds costs for the overall health care system.4 One study found that internal review of outside biopsy slides resulted in a change in diagnosis in 2.2% of patients (N=3345)—affecting management in 61% of cases in which the diagnosis was changed.4 Another study (N=163) found that the reported aggressiveness of 50.5% of nonmelanoma cases in an initial biopsy report was changed during Mohs micrographic surgery.5 Mohs surgeons should be aware that discrepancies can occur, and if a discrepancy is discovered, the procedure may be paused until the initial biopsy slide is reviewed and further information is collected.

References
  1. Jayaprakasam A, Rene C. A benign or malignant eyelid lump—can you tell? an unusual collision tumour highlighting the difficulty differentiating a hidrocystoma from a basal cell carcinoma. BMJ Case Reports. 2012;2012:bcr1220115307. doi:10.1136/bcr.12.2011.5307
  2. Miteva M, Herschthal D, Ricotti C, et al. A rare case of a cutaneous squamomelanocytic tumor: revisiting the histogenesis of combined neoplasms. Am J Dermatopathol. 2009;31:599-603. doi:10.1097/DAD.0b013e3181a88116
  3. Idriss MH, Elston DM. Secondary neoplasms associated with nevus sebaceus of Jadassohn: a study of 707 cases. J Am Acad Dermatol. 2014;70:332-337. doi:10.1016/j.jaad.2013.10.004
  4. Butler ST, Youker SR, Mandrell J, et al. The importance of reviewing pathology specimens before Mohs surgery. Dermatol Surg. 2009;35:407-412. doi:10.1111/j.1524-4725.2008.01056.x
  5. Stiegel E, Lam C, Schowalter M, et al. Correlation between original biopsy pathology and Mohs intraoperative pathology. Dermatol Surg. 2018;44:193-197. doi:10.1097/DSS.0000000000001276
References
  1. Jayaprakasam A, Rene C. A benign or malignant eyelid lump—can you tell? an unusual collision tumour highlighting the difficulty differentiating a hidrocystoma from a basal cell carcinoma. BMJ Case Reports. 2012;2012:bcr1220115307. doi:10.1136/bcr.12.2011.5307
  2. Miteva M, Herschthal D, Ricotti C, et al. A rare case of a cutaneous squamomelanocytic tumor: revisiting the histogenesis of combined neoplasms. Am J Dermatopathol. 2009;31:599-603. doi:10.1097/DAD.0b013e3181a88116
  3. Idriss MH, Elston DM. Secondary neoplasms associated with nevus sebaceus of Jadassohn: a study of 707 cases. J Am Acad Dermatol. 2014;70:332-337. doi:10.1016/j.jaad.2013.10.004
  4. Butler ST, Youker SR, Mandrell J, et al. The importance of reviewing pathology specimens before Mohs surgery. Dermatol Surg. 2009;35:407-412. doi:10.1111/j.1524-4725.2008.01056.x
  5. Stiegel E, Lam C, Schowalter M, et al. Correlation between original biopsy pathology and Mohs intraoperative pathology. Dermatol Surg. 2018;44:193-197. doi:10.1097/DSS.0000000000001276
Issue
Cutis - 111(4)
Issue
Cutis - 111(4)
Page Number
E9-E11
Page Number
E9-E11
Publications
Publications
Topics
Article Type
Display Headline
Collision Course of a Basal Cell Carcinoma and Apocrine Hidrocystoma on the Scalp
Display Headline
Collision Course of a Basal Cell Carcinoma and Apocrine Hidrocystoma on the Scalp
Sections
Inside the Article

PRACTICE POINTS

  • When collision tumors are encountered during Mohs micrographic surgery, review of the initial diagnostic material is recommended.
  • Permanent processing of Mohs excisions may be helpful in determining the diagnosis of the occult second tumor diagnosis.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Subcutaneous Panniculitic T-cell Lymphoma Presenting With Anasarca in a Patient With Known Chronic Lymphocytic Leukemia

Article Type
Changed
Fri, 03/31/2023 - 09:52
Display Headline
Subcutaneous Panniculitic T-cell Lymphoma Presenting With Anasarca in a Patient With Known Chronic Lymphocytic Leukemia

To the Editor:

Subcutaneous panniculitic T-cell lymphoma (SPTCL) is a rare cutaneous T-cell lymphoma that was first described in 19911 and comprises less than 1% of all non-Hodgkin lymphomas (NHLs). It most commonly occurs in young adults, with a median patient age of 36 years and a slight female predominance.2 Patients typically present with skin nodules or deep-seated plaques involving the legs, arms, and/or trunk. Presentation on the face is less common.2,3 Paraneoplastic edema has been reported in several cases of SPTCL with facial and periorbital swelling.4-9

Diagnosis of SPTCL is achieved via analysis of a deep tissue skin biopsy and close clinicopathologic correlation. Histopathology demonstrates lobular panniculitis with an atypical lymphoid infiltrate in the subcutaneous tissue with predominantly CD8+ T cells without overlying epidermotropism or interface dermatitis.3 The degree of cellular atypia, fat necrosis, karyorrhexis, cytophagia, and lack of angioinvasion can help to distinguish SPTCL from other panniculitides.2,3

The prognosis of SPTCL is good, with a 5-year survival rate of 82%, and many patients are able to achieve remission.2 However, SPTCL can progress to a fatal hemophagocytic syndrome, which has been reported in 17% of cases, making early diagnosis and treatment of this malignancy imperative.1,2 Treatment varies depending on the progression and extent of disease and can include the use of steroids, multidrug chemotherapy regimens, radiotherapy, and stem cell transplant in refractory cases.2-4,10,11

Subcutaneous panniculitic T-cell lymphoma with edema has been reported in a 2-year-old child.12 We present a case of SPTCL in an adult patient with known stage IV chronic lymphocytic leukemia (CLL) who also had full-body edema.

A 60-year-old woman with a 7-year history of stage IV CLL presented with anasarca of 3 months’ duration. At the time of presentation to dermatology, physical examination revealed erythematous tender nodules on the arms and legs. She had no other medical conditions and was undergoing treatment with ibrutinib for the CLL. The patient reported profound fatigue but no fever, chills, night sweats, cough, or dyspnea. The swelling had begun initially in the legs and progressively worsened to involve the arms, face, and body. She was hospitalized and treated with intravenous steroids and antihistamines, which led to minor improvement in the swelling. The patient’s preliminary diagnosis of erythema nodosum was thought to be related to the CLL or ibrutinib; therefore, treatment subsequently was discontinued and she was discharged from the hospital.

The swelling continued to worsen over the following 3 months, and the patient gained approximately 25 pounds. She presented to our office again with severe periorbital, facial, and lip edema as well as diffuse edema of the torso, arms, and legs (Figure 1). Erythematous tender subcutaneous nodules were noted on the right proximal thigh, left lateral calf, and forearms. She was again hospitalized, and extensive evaluation was performed to exclude other causes of anasarca, including a complete blood cell count; comprehensive metabolic profile; hepatitis panels; HIV test; C3 and C4, complement CH50, C1 esterase inhibitor, IgE, and angiotensin-converting enzyme levels; urine protein to creatinine ratio; computed tomography of the chest, abdomen, and pelvis; and allergy evaluation. The analyses failed to reveal the cause of the anasarca.

A, A 60-year-old woman with periorbital, facial, and lip edema. B, The lower extremities also showed edema, erythema, and a left lateral subcutaneous nodule (arrow).
FIGURE 1. A, A 60-year-old woman with periorbital, facial, and lip edema. B, The lower extremities also showed edema, erythema, and a left lateral subcutaneous nodule (arrow).

During hospitalization, the patient underwent a lymph node biopsy, bone marrow biopsy, and a 6-mm punch biopsy of the right thigh nodule. The lymph node and bone marrow biopsy results were consistent with the known diagnosis of CLL, and the patient was started on intravenous chemotherapy with bendamustine. The skin biopsy demonstrated a predominant T-cell infiltrate consistent with a lobular panniculitis with variable amounts of adipocytes rimmed by lymphocytes, nuclear debris, and karyorrhexis (Figure 2). CD3+, CD8+, and CD4 T cells were positive for T-cell receptor (TCR) βF1 and negative for TCR-γ with strong expression of cytotoxic markers including granzyme B, perforin, and T-cell intracytoplasmic antigen 1. Rare CD56+ cells also were noted. The biopsy did not demonstrate any notable interface dermatitis, epidermotropism, or angioinvasion. T-cell receptor gene rearrangement studies did not show clonality for γ- or β-chain probes. Subcutaneous panniculitic T-cell lymphoma was diagnosed, making this case unique with the presentation of anasarca. This case also is noteworthy due to the rare diagnosis of the secondary malignancy of SPTCL in a patient with known CLL. The patient opted to pursue hospice and comfort measures due to the effects of persistent pancytopenia and the progression of CLL. She died 2 months later.

A, A punch biopsy demonstrated a predominant T-cell infiltrate within the subcutaneous adipose tissue (H&E, original magnification ×4). B, Variable amounts of adipocytes rimmed by lymphocytes, nuclear debris, and karyorrhexis were shown on high power
FIGURE 2. A, A punch biopsy demonstrated a predominant T-cell infiltrate within the subcutaneous adipose tissue (H&E, original magnification ×4). B, Variable amounts of adipocytes rimmed by lymphocytes, nuclear debris, and karyorrhexis were shown on high power (H&E, original magnification ×200). C, An immunostain for T-cell receptor βF1 highlighted lymphocytes surrounding adipocytes (original magnification ×40).

 

 

Clinical courses of SPTCL vary based on the TCR phenotype and immunophenotypic characteristics of the tumor cells. The TCR-αβ phenotype, as described in this case, typically is CD4, CD8+, and CD56 and leads to a more indolent disease course. Lymphomas with the TCR-γδ phenotype typically are CD4, CD8, and CD56+; they often are associated with hemophagocytic syndrome and thus a worse prognosis. In 2009, the World Health Organization–European Organization for Research and Treatment of Cancer classification of primary cutaneous lymphomas restricted the category of SPTCL to the TCR-αβ phenotype due to the stark differences between the 2 types. The TCR-γδ phenotype was given its own diagnostic category—primary cutaneous γδ T-cell lymphoma.3

Patients with SPTCL commonly present with nodular skin lesions or deep-seated plaques on the legs, arms, and/or trunk; presentation on the face is rare.2,3 Fever, chills, night sweats, and/or weight loss were present in approximately 50% of recorded cases. Underlying autoimmune disease was present in 12 of 63 (19%) patients in a 2008 study.2 Facial and periorbital swelling with SPTCL has been reported.4-9 The presentation of anasarca, as seen in our adult patient, has been reported in a 2-year-old child.12 Anasarca as a presenting symptom of NHL is a rare phenomenon proposed to be induced by malignant cells secreting a cytokine that causes a vascular leak syndrome.13 Specifically, tumor necrosis factor α was found to be elevated in at least 2 patients with NHL presenting with anasarca in a prior study. Tumor necrosis factor α is known to cause increased capillary permeability, vascular leakage, and development of edema.13 In retrospect, obtaining cytokine levels in our patient would have been useful to support or refute tumor necrosis factor α as a possible cause of anasarca in the setting of NHL. This case continues to highlight that a diagnosis of SPTCL and analysis of a skin biopsy should be considered in cases of sudden unremitting facial and/or body swelling that cannot be explained by other more common causes.

Subcutaneous panniculitic T-cell lymphoma can be diagnosed and distinguished from other panniculitides via analysis of a deep tissue skin biopsy. Multiple biopsies may be required to ensure an adequate sample is obtained.4 Histopathology displays an atypical lymphoid infiltrate with a predominant presence of T cells. Neoplastic cells show CD3+, CD8+, and CD4 T cells, which strongly express cytotoxic proteins such as granzyme B, T-cell intracellular antigen 1, and perforin.3 The degree of cellular atypia, fat necrosis, karyorrhexis, and cytophagia, as well as the lack of angioinvasion, interface dermatitis, and epidermotropism help to distinguish SPTCL from other panniculitides.2,3 According to a previous study, clonal TCR gene rearrangement was identified in 50% to 80% of cases, but the absence of this clonal rearrangement does not exclude the diagnosis.14

This case also highlights the occurrence of secondary malignancies in patients with CLL, an NHL that is classified as a low-grade lymphoproliferative malignancy with clonal expansion of B cells.15 Secondary CTCLs in patients with CLL are rare, but they have been previously described. In 2017, Chang et al16 identified 12 patients with CLL who subsequently developed CTCL between 1992 and 2008. Of the 12 patients, 7 developed mycosis fungoides, 3 had CTCL not otherwise specified, 1 had mature T-cell lymphoma not otherwise specified, and 1 had primary cutaneous CD30+ T-cell lymphoma.16 The proliferation of 2 separate lymphocytic lineages is rare, but this study demonstrated an increased risk for CTCL to develop in patients with CLL. One possible explanation is that malignant cells come from a common stem cell progenitor or from genetic events. They occur secondary to carcinogens, viruses, or cytokines from T-cell or B-cell clones; they evolve due to treatment of the preexisting lymphoproliferative disease; or they occur simply by coincidence. The behavior of the CTCL may be more aggressive in patients with CLL due to immunosuppression, which may have contributed to the extreme presentation in our patient.16 Subcutaneous panniculitic T-cell lymphoma also has been reported in a patient with CLL that was thought to be associated with prior rituximab treatment.17

Treatment of SPTCL depends on the severity and course of the disease. In patients with more indolent disease, systemic steroids have been the most frequently used initial treatment.2,3,10 However, the disease often will progress after steroid tapering and require further intervention. Localized lesions may be treated with radiation alone or in combination with other systemic therapies.3,10 In refractory, aggressive, or relapsing cases, polychemotherapeutic regimens have proven to produce long-term remission in 30% of patients, with an overall response rate of 50%.10 These regimens most commonly have included cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or CHOP-like treatment (EPOCH regimen [etoposide, prednisone, oncovin, cyclophosphamide, and doxorubicin hydrochloride]).3,10 A stem cell transplant can be considered in patients with recurrent and refractory disease, and it also has been shown to induce remission.4,17 In patients with a good response to therapy, the disease often can be controlled for long periods of time, with an estimated 5-year survival rate of 80%.15

This case highlights the diagnostic challenges and variable presentations of SPTCL. Dermatologists, oncologists, and dermatopathologists should be aware of this condition and consider it in the differential diagnosis of a patient with a hematologic malignancy and unremitting facial and/or body swelling without any other cause. The possibility of a secondary hematologic cancer in a patient with CLL also must be taken into consideration. Early diagnosis and treatment can minimize morbidity and induce remission in most patients.

References
  1. Gonzalez CL, Medeiros LJ, Braziel RM, et al. T-cell lymphoma involving subcutaneous tissue. a clinicopathologic entity commonly associated with hemophagocytic syndrome. Am J Surg Pathol. 1991;15:17-27.
  2. Willemze R, Jansen P, Cerroni L, et al. Subcutaneous panniculitis-like T-cell lymphoma: definition, classification, and prognostic factors: an EORTC Cutaneous Lymphoma Group Study of 83 cases. Blood. 2008;111:38-45.
  3. Parveen Z, Thompson K. Subcutaneous panniculitis-like T-cell lymphoma: redefinition of diagnostic criteria in the recent World Health Organization–European Organization for Research and Treatment of Cancer classification for cutaneous lymphomas. Arch Pathol Lab Med. 2009;133:303-308.
  4. Velez N, Ishizawar R, Dellaripa P, et al. Full facial edema: a novel presentation of subcutaneous panniculitis-like T-cell lymphoma. J Clin Oncol. 2012;30:e233-236.
  5. Asati D, Ingle V, Joshi D, et al. Subcutaneous panniculitis-like T-cell lymphoma with macrophage activation syndrome treated by cyclosporine and prednisolone. Indian Dermatol Online J. 2016;7:529-532.
  6. Fricker M, Dubach P, Helbing A, et al. Not all facial swellings are angioedemas! J Investig Allergol Clin Immunol. 2015;25:146-147.
  7. Kosari F, Akbarzadeh H. Local facial edema: a novel presentation of subcutaneous panniculitis-like T-cell lymphoma in a 30-year-old Iranian woman. Acta Med Iran. 2014;52:950-953.
  8. Bhojaraja M, Kistampally P, Udupa K, et al. Subcutaneous panniculitis-like T-cell lymphoma: a rare tumour. J Clin Diagn Res. 2016;10:OD29-OD30.
  9. Hashimoto R, Uchiyama M, Maeno T. Case report of subcutaneous panniculitis-like T-cell lymphoma complicated by eyelid swelling. BMC Ophthalmol. 2016;16:117.
  10. Chinello MN, Naviglio S, Remotti D, et al. Subcutaneous panniculitis-like T-cell lymphoma presenting with diffuse cutaneous edema in a 2-year-old child. J Pediatr Hematol Oncol. 2015;37:329-330.
  11. Chang TW, Weaver AL, Shanafelt TD, et al. Risk of cutaneous T-cell lymphoma in patients with chronic lymphocytic leukemia and other subtypes of non-Hodgkin lymphoma. Int J Dermatol. 2017;56:1125-1129.
  12. Chinello MN, Naviglio S, Remotti D, et al. Subcutaneous panniculitis-like T-cell lymphoma presenting with diffuse cutaneous edema in a 2-year-old child. J Pediatr Hematol Oncol. 2015;37:329-330.
  13. Jillella A, Day D, Severson K, et al. Non-Hodgkin’s lymphoma presenting as anasarca: probably mediated by tumor necrosis factor alpha (TNF-α). Leuk Lymphoma. 2000;38:419-422.
  14. Lee D-W, Yang J-H, Lee S-M, et al. Subcutaneous panniculitis-like T-cell lymphoma: a clinical and pathologic study of 14 Korean patients. Ann Dermatol. 2011;23:329-337.
  15. Jaffe ES. The 2008 WHO classification of lymphomas: implications for clinical practice and translational research [published online January 1, 2009]. Hematology Am Soc Hematol Educ Program. https://doi.org/10.1182/asheducation-2009.1.523
  16. Chang TW, Weaver AL, Shanafelt TD, et al. Risk of cutaneous T-cell lymphoma in patients with chronic lymphocytic leukemia and other subtypes of non-Hodgkin lymphoma. Int J Dermatol. 2017;56:1125-1129.
  17. Hall M, Sluzevich J, Snow J. Generalized subcutaneous panniculitis-like T-cell lymphoma following rituximab for hemolytic anemia in a patient with chronic lymphocytic leukemia. J Am Acad Dermatol. 2010;62(suppl 1):AB96.
Article PDF
Author and Disclosure Information

Dr. Reinhold is from Beaumont Health-Royal Oak Internal Medicine, Michigan. Dr. Neidenbach is from Westside Dermatology, Spartanburg, South Carolina.

The authors report no conflict of interest.

Correspondence: Lauren Reinhold, DO, 3601 W 13 Mile Rd, Royal Oak, MI 48073 (lauren.reinhold@beaumont.org).

Issue
Cutis - 111(3)
Publications
Topics
Page Number
E28-E31
Sections
Author and Disclosure Information

Dr. Reinhold is from Beaumont Health-Royal Oak Internal Medicine, Michigan. Dr. Neidenbach is from Westside Dermatology, Spartanburg, South Carolina.

The authors report no conflict of interest.

Correspondence: Lauren Reinhold, DO, 3601 W 13 Mile Rd, Royal Oak, MI 48073 (lauren.reinhold@beaumont.org).

Author and Disclosure Information

Dr. Reinhold is from Beaumont Health-Royal Oak Internal Medicine, Michigan. Dr. Neidenbach is from Westside Dermatology, Spartanburg, South Carolina.

The authors report no conflict of interest.

Correspondence: Lauren Reinhold, DO, 3601 W 13 Mile Rd, Royal Oak, MI 48073 (lauren.reinhold@beaumont.org).

Article PDF
Article PDF

To the Editor:

Subcutaneous panniculitic T-cell lymphoma (SPTCL) is a rare cutaneous T-cell lymphoma that was first described in 19911 and comprises less than 1% of all non-Hodgkin lymphomas (NHLs). It most commonly occurs in young adults, with a median patient age of 36 years and a slight female predominance.2 Patients typically present with skin nodules or deep-seated plaques involving the legs, arms, and/or trunk. Presentation on the face is less common.2,3 Paraneoplastic edema has been reported in several cases of SPTCL with facial and periorbital swelling.4-9

Diagnosis of SPTCL is achieved via analysis of a deep tissue skin biopsy and close clinicopathologic correlation. Histopathology demonstrates lobular panniculitis with an atypical lymphoid infiltrate in the subcutaneous tissue with predominantly CD8+ T cells without overlying epidermotropism or interface dermatitis.3 The degree of cellular atypia, fat necrosis, karyorrhexis, cytophagia, and lack of angioinvasion can help to distinguish SPTCL from other panniculitides.2,3

The prognosis of SPTCL is good, with a 5-year survival rate of 82%, and many patients are able to achieve remission.2 However, SPTCL can progress to a fatal hemophagocytic syndrome, which has been reported in 17% of cases, making early diagnosis and treatment of this malignancy imperative.1,2 Treatment varies depending on the progression and extent of disease and can include the use of steroids, multidrug chemotherapy regimens, radiotherapy, and stem cell transplant in refractory cases.2-4,10,11

Subcutaneous panniculitic T-cell lymphoma with edema has been reported in a 2-year-old child.12 We present a case of SPTCL in an adult patient with known stage IV chronic lymphocytic leukemia (CLL) who also had full-body edema.

A 60-year-old woman with a 7-year history of stage IV CLL presented with anasarca of 3 months’ duration. At the time of presentation to dermatology, physical examination revealed erythematous tender nodules on the arms and legs. She had no other medical conditions and was undergoing treatment with ibrutinib for the CLL. The patient reported profound fatigue but no fever, chills, night sweats, cough, or dyspnea. The swelling had begun initially in the legs and progressively worsened to involve the arms, face, and body. She was hospitalized and treated with intravenous steroids and antihistamines, which led to minor improvement in the swelling. The patient’s preliminary diagnosis of erythema nodosum was thought to be related to the CLL or ibrutinib; therefore, treatment subsequently was discontinued and she was discharged from the hospital.

The swelling continued to worsen over the following 3 months, and the patient gained approximately 25 pounds. She presented to our office again with severe periorbital, facial, and lip edema as well as diffuse edema of the torso, arms, and legs (Figure 1). Erythematous tender subcutaneous nodules were noted on the right proximal thigh, left lateral calf, and forearms. She was again hospitalized, and extensive evaluation was performed to exclude other causes of anasarca, including a complete blood cell count; comprehensive metabolic profile; hepatitis panels; HIV test; C3 and C4, complement CH50, C1 esterase inhibitor, IgE, and angiotensin-converting enzyme levels; urine protein to creatinine ratio; computed tomography of the chest, abdomen, and pelvis; and allergy evaluation. The analyses failed to reveal the cause of the anasarca.

A, A 60-year-old woman with periorbital, facial, and lip edema. B, The lower extremities also showed edema, erythema, and a left lateral subcutaneous nodule (arrow).
FIGURE 1. A, A 60-year-old woman with periorbital, facial, and lip edema. B, The lower extremities also showed edema, erythema, and a left lateral subcutaneous nodule (arrow).

During hospitalization, the patient underwent a lymph node biopsy, bone marrow biopsy, and a 6-mm punch biopsy of the right thigh nodule. The lymph node and bone marrow biopsy results were consistent with the known diagnosis of CLL, and the patient was started on intravenous chemotherapy with bendamustine. The skin biopsy demonstrated a predominant T-cell infiltrate consistent with a lobular panniculitis with variable amounts of adipocytes rimmed by lymphocytes, nuclear debris, and karyorrhexis (Figure 2). CD3+, CD8+, and CD4 T cells were positive for T-cell receptor (TCR) βF1 and negative for TCR-γ with strong expression of cytotoxic markers including granzyme B, perforin, and T-cell intracytoplasmic antigen 1. Rare CD56+ cells also were noted. The biopsy did not demonstrate any notable interface dermatitis, epidermotropism, or angioinvasion. T-cell receptor gene rearrangement studies did not show clonality for γ- or β-chain probes. Subcutaneous panniculitic T-cell lymphoma was diagnosed, making this case unique with the presentation of anasarca. This case also is noteworthy due to the rare diagnosis of the secondary malignancy of SPTCL in a patient with known CLL. The patient opted to pursue hospice and comfort measures due to the effects of persistent pancytopenia and the progression of CLL. She died 2 months later.

A, A punch biopsy demonstrated a predominant T-cell infiltrate within the subcutaneous adipose tissue (H&E, original magnification ×4). B, Variable amounts of adipocytes rimmed by lymphocytes, nuclear debris, and karyorrhexis were shown on high power
FIGURE 2. A, A punch biopsy demonstrated a predominant T-cell infiltrate within the subcutaneous adipose tissue (H&E, original magnification ×4). B, Variable amounts of adipocytes rimmed by lymphocytes, nuclear debris, and karyorrhexis were shown on high power (H&E, original magnification ×200). C, An immunostain for T-cell receptor βF1 highlighted lymphocytes surrounding adipocytes (original magnification ×40).

 

 

Clinical courses of SPTCL vary based on the TCR phenotype and immunophenotypic characteristics of the tumor cells. The TCR-αβ phenotype, as described in this case, typically is CD4, CD8+, and CD56 and leads to a more indolent disease course. Lymphomas with the TCR-γδ phenotype typically are CD4, CD8, and CD56+; they often are associated with hemophagocytic syndrome and thus a worse prognosis. In 2009, the World Health Organization–European Organization for Research and Treatment of Cancer classification of primary cutaneous lymphomas restricted the category of SPTCL to the TCR-αβ phenotype due to the stark differences between the 2 types. The TCR-γδ phenotype was given its own diagnostic category—primary cutaneous γδ T-cell lymphoma.3

Patients with SPTCL commonly present with nodular skin lesions or deep-seated plaques on the legs, arms, and/or trunk; presentation on the face is rare.2,3 Fever, chills, night sweats, and/or weight loss were present in approximately 50% of recorded cases. Underlying autoimmune disease was present in 12 of 63 (19%) patients in a 2008 study.2 Facial and periorbital swelling with SPTCL has been reported.4-9 The presentation of anasarca, as seen in our adult patient, has been reported in a 2-year-old child.12 Anasarca as a presenting symptom of NHL is a rare phenomenon proposed to be induced by malignant cells secreting a cytokine that causes a vascular leak syndrome.13 Specifically, tumor necrosis factor α was found to be elevated in at least 2 patients with NHL presenting with anasarca in a prior study. Tumor necrosis factor α is known to cause increased capillary permeability, vascular leakage, and development of edema.13 In retrospect, obtaining cytokine levels in our patient would have been useful to support or refute tumor necrosis factor α as a possible cause of anasarca in the setting of NHL. This case continues to highlight that a diagnosis of SPTCL and analysis of a skin biopsy should be considered in cases of sudden unremitting facial and/or body swelling that cannot be explained by other more common causes.

Subcutaneous panniculitic T-cell lymphoma can be diagnosed and distinguished from other panniculitides via analysis of a deep tissue skin biopsy. Multiple biopsies may be required to ensure an adequate sample is obtained.4 Histopathology displays an atypical lymphoid infiltrate with a predominant presence of T cells. Neoplastic cells show CD3+, CD8+, and CD4 T cells, which strongly express cytotoxic proteins such as granzyme B, T-cell intracellular antigen 1, and perforin.3 The degree of cellular atypia, fat necrosis, karyorrhexis, and cytophagia, as well as the lack of angioinvasion, interface dermatitis, and epidermotropism help to distinguish SPTCL from other panniculitides.2,3 According to a previous study, clonal TCR gene rearrangement was identified in 50% to 80% of cases, but the absence of this clonal rearrangement does not exclude the diagnosis.14

This case also highlights the occurrence of secondary malignancies in patients with CLL, an NHL that is classified as a low-grade lymphoproliferative malignancy with clonal expansion of B cells.15 Secondary CTCLs in patients with CLL are rare, but they have been previously described. In 2017, Chang et al16 identified 12 patients with CLL who subsequently developed CTCL between 1992 and 2008. Of the 12 patients, 7 developed mycosis fungoides, 3 had CTCL not otherwise specified, 1 had mature T-cell lymphoma not otherwise specified, and 1 had primary cutaneous CD30+ T-cell lymphoma.16 The proliferation of 2 separate lymphocytic lineages is rare, but this study demonstrated an increased risk for CTCL to develop in patients with CLL. One possible explanation is that malignant cells come from a common stem cell progenitor or from genetic events. They occur secondary to carcinogens, viruses, or cytokines from T-cell or B-cell clones; they evolve due to treatment of the preexisting lymphoproliferative disease; or they occur simply by coincidence. The behavior of the CTCL may be more aggressive in patients with CLL due to immunosuppression, which may have contributed to the extreme presentation in our patient.16 Subcutaneous panniculitic T-cell lymphoma also has been reported in a patient with CLL that was thought to be associated with prior rituximab treatment.17

Treatment of SPTCL depends on the severity and course of the disease. In patients with more indolent disease, systemic steroids have been the most frequently used initial treatment.2,3,10 However, the disease often will progress after steroid tapering and require further intervention. Localized lesions may be treated with radiation alone or in combination with other systemic therapies.3,10 In refractory, aggressive, or relapsing cases, polychemotherapeutic regimens have proven to produce long-term remission in 30% of patients, with an overall response rate of 50%.10 These regimens most commonly have included cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or CHOP-like treatment (EPOCH regimen [etoposide, prednisone, oncovin, cyclophosphamide, and doxorubicin hydrochloride]).3,10 A stem cell transplant can be considered in patients with recurrent and refractory disease, and it also has been shown to induce remission.4,17 In patients with a good response to therapy, the disease often can be controlled for long periods of time, with an estimated 5-year survival rate of 80%.15

This case highlights the diagnostic challenges and variable presentations of SPTCL. Dermatologists, oncologists, and dermatopathologists should be aware of this condition and consider it in the differential diagnosis of a patient with a hematologic malignancy and unremitting facial and/or body swelling without any other cause. The possibility of a secondary hematologic cancer in a patient with CLL also must be taken into consideration. Early diagnosis and treatment can minimize morbidity and induce remission in most patients.

To the Editor:

Subcutaneous panniculitic T-cell lymphoma (SPTCL) is a rare cutaneous T-cell lymphoma that was first described in 19911 and comprises less than 1% of all non-Hodgkin lymphomas (NHLs). It most commonly occurs in young adults, with a median patient age of 36 years and a slight female predominance.2 Patients typically present with skin nodules or deep-seated plaques involving the legs, arms, and/or trunk. Presentation on the face is less common.2,3 Paraneoplastic edema has been reported in several cases of SPTCL with facial and periorbital swelling.4-9

Diagnosis of SPTCL is achieved via analysis of a deep tissue skin biopsy and close clinicopathologic correlation. Histopathology demonstrates lobular panniculitis with an atypical lymphoid infiltrate in the subcutaneous tissue with predominantly CD8+ T cells without overlying epidermotropism or interface dermatitis.3 The degree of cellular atypia, fat necrosis, karyorrhexis, cytophagia, and lack of angioinvasion can help to distinguish SPTCL from other panniculitides.2,3

The prognosis of SPTCL is good, with a 5-year survival rate of 82%, and many patients are able to achieve remission.2 However, SPTCL can progress to a fatal hemophagocytic syndrome, which has been reported in 17% of cases, making early diagnosis and treatment of this malignancy imperative.1,2 Treatment varies depending on the progression and extent of disease and can include the use of steroids, multidrug chemotherapy regimens, radiotherapy, and stem cell transplant in refractory cases.2-4,10,11

Subcutaneous panniculitic T-cell lymphoma with edema has been reported in a 2-year-old child.12 We present a case of SPTCL in an adult patient with known stage IV chronic lymphocytic leukemia (CLL) who also had full-body edema.

A 60-year-old woman with a 7-year history of stage IV CLL presented with anasarca of 3 months’ duration. At the time of presentation to dermatology, physical examination revealed erythematous tender nodules on the arms and legs. She had no other medical conditions and was undergoing treatment with ibrutinib for the CLL. The patient reported profound fatigue but no fever, chills, night sweats, cough, or dyspnea. The swelling had begun initially in the legs and progressively worsened to involve the arms, face, and body. She was hospitalized and treated with intravenous steroids and antihistamines, which led to minor improvement in the swelling. The patient’s preliminary diagnosis of erythema nodosum was thought to be related to the CLL or ibrutinib; therefore, treatment subsequently was discontinued and she was discharged from the hospital.

The swelling continued to worsen over the following 3 months, and the patient gained approximately 25 pounds. She presented to our office again with severe periorbital, facial, and lip edema as well as diffuse edema of the torso, arms, and legs (Figure 1). Erythematous tender subcutaneous nodules were noted on the right proximal thigh, left lateral calf, and forearms. She was again hospitalized, and extensive evaluation was performed to exclude other causes of anasarca, including a complete blood cell count; comprehensive metabolic profile; hepatitis panels; HIV test; C3 and C4, complement CH50, C1 esterase inhibitor, IgE, and angiotensin-converting enzyme levels; urine protein to creatinine ratio; computed tomography of the chest, abdomen, and pelvis; and allergy evaluation. The analyses failed to reveal the cause of the anasarca.

A, A 60-year-old woman with periorbital, facial, and lip edema. B, The lower extremities also showed edema, erythema, and a left lateral subcutaneous nodule (arrow).
FIGURE 1. A, A 60-year-old woman with periorbital, facial, and lip edema. B, The lower extremities also showed edema, erythema, and a left lateral subcutaneous nodule (arrow).

During hospitalization, the patient underwent a lymph node biopsy, bone marrow biopsy, and a 6-mm punch biopsy of the right thigh nodule. The lymph node and bone marrow biopsy results were consistent with the known diagnosis of CLL, and the patient was started on intravenous chemotherapy with bendamustine. The skin biopsy demonstrated a predominant T-cell infiltrate consistent with a lobular panniculitis with variable amounts of adipocytes rimmed by lymphocytes, nuclear debris, and karyorrhexis (Figure 2). CD3+, CD8+, and CD4 T cells were positive for T-cell receptor (TCR) βF1 and negative for TCR-γ with strong expression of cytotoxic markers including granzyme B, perforin, and T-cell intracytoplasmic antigen 1. Rare CD56+ cells also were noted. The biopsy did not demonstrate any notable interface dermatitis, epidermotropism, or angioinvasion. T-cell receptor gene rearrangement studies did not show clonality for γ- or β-chain probes. Subcutaneous panniculitic T-cell lymphoma was diagnosed, making this case unique with the presentation of anasarca. This case also is noteworthy due to the rare diagnosis of the secondary malignancy of SPTCL in a patient with known CLL. The patient opted to pursue hospice and comfort measures due to the effects of persistent pancytopenia and the progression of CLL. She died 2 months later.

A, A punch biopsy demonstrated a predominant T-cell infiltrate within the subcutaneous adipose tissue (H&E, original magnification ×4). B, Variable amounts of adipocytes rimmed by lymphocytes, nuclear debris, and karyorrhexis were shown on high power
FIGURE 2. A, A punch biopsy demonstrated a predominant T-cell infiltrate within the subcutaneous adipose tissue (H&E, original magnification ×4). B, Variable amounts of adipocytes rimmed by lymphocytes, nuclear debris, and karyorrhexis were shown on high power (H&E, original magnification ×200). C, An immunostain for T-cell receptor βF1 highlighted lymphocytes surrounding adipocytes (original magnification ×40).

 

 

Clinical courses of SPTCL vary based on the TCR phenotype and immunophenotypic characteristics of the tumor cells. The TCR-αβ phenotype, as described in this case, typically is CD4, CD8+, and CD56 and leads to a more indolent disease course. Lymphomas with the TCR-γδ phenotype typically are CD4, CD8, and CD56+; they often are associated with hemophagocytic syndrome and thus a worse prognosis. In 2009, the World Health Organization–European Organization for Research and Treatment of Cancer classification of primary cutaneous lymphomas restricted the category of SPTCL to the TCR-αβ phenotype due to the stark differences between the 2 types. The TCR-γδ phenotype was given its own diagnostic category—primary cutaneous γδ T-cell lymphoma.3

Patients with SPTCL commonly present with nodular skin lesions or deep-seated plaques on the legs, arms, and/or trunk; presentation on the face is rare.2,3 Fever, chills, night sweats, and/or weight loss were present in approximately 50% of recorded cases. Underlying autoimmune disease was present in 12 of 63 (19%) patients in a 2008 study.2 Facial and periorbital swelling with SPTCL has been reported.4-9 The presentation of anasarca, as seen in our adult patient, has been reported in a 2-year-old child.12 Anasarca as a presenting symptom of NHL is a rare phenomenon proposed to be induced by malignant cells secreting a cytokine that causes a vascular leak syndrome.13 Specifically, tumor necrosis factor α was found to be elevated in at least 2 patients with NHL presenting with anasarca in a prior study. Tumor necrosis factor α is known to cause increased capillary permeability, vascular leakage, and development of edema.13 In retrospect, obtaining cytokine levels in our patient would have been useful to support or refute tumor necrosis factor α as a possible cause of anasarca in the setting of NHL. This case continues to highlight that a diagnosis of SPTCL and analysis of a skin biopsy should be considered in cases of sudden unremitting facial and/or body swelling that cannot be explained by other more common causes.

Subcutaneous panniculitic T-cell lymphoma can be diagnosed and distinguished from other panniculitides via analysis of a deep tissue skin biopsy. Multiple biopsies may be required to ensure an adequate sample is obtained.4 Histopathology displays an atypical lymphoid infiltrate with a predominant presence of T cells. Neoplastic cells show CD3+, CD8+, and CD4 T cells, which strongly express cytotoxic proteins such as granzyme B, T-cell intracellular antigen 1, and perforin.3 The degree of cellular atypia, fat necrosis, karyorrhexis, and cytophagia, as well as the lack of angioinvasion, interface dermatitis, and epidermotropism help to distinguish SPTCL from other panniculitides.2,3 According to a previous study, clonal TCR gene rearrangement was identified in 50% to 80% of cases, but the absence of this clonal rearrangement does not exclude the diagnosis.14

This case also highlights the occurrence of secondary malignancies in patients with CLL, an NHL that is classified as a low-grade lymphoproliferative malignancy with clonal expansion of B cells.15 Secondary CTCLs in patients with CLL are rare, but they have been previously described. In 2017, Chang et al16 identified 12 patients with CLL who subsequently developed CTCL between 1992 and 2008. Of the 12 patients, 7 developed mycosis fungoides, 3 had CTCL not otherwise specified, 1 had mature T-cell lymphoma not otherwise specified, and 1 had primary cutaneous CD30+ T-cell lymphoma.16 The proliferation of 2 separate lymphocytic lineages is rare, but this study demonstrated an increased risk for CTCL to develop in patients with CLL. One possible explanation is that malignant cells come from a common stem cell progenitor or from genetic events. They occur secondary to carcinogens, viruses, or cytokines from T-cell or B-cell clones; they evolve due to treatment of the preexisting lymphoproliferative disease; or they occur simply by coincidence. The behavior of the CTCL may be more aggressive in patients with CLL due to immunosuppression, which may have contributed to the extreme presentation in our patient.16 Subcutaneous panniculitic T-cell lymphoma also has been reported in a patient with CLL that was thought to be associated with prior rituximab treatment.17

Treatment of SPTCL depends on the severity and course of the disease. In patients with more indolent disease, systemic steroids have been the most frequently used initial treatment.2,3,10 However, the disease often will progress after steroid tapering and require further intervention. Localized lesions may be treated with radiation alone or in combination with other systemic therapies.3,10 In refractory, aggressive, or relapsing cases, polychemotherapeutic regimens have proven to produce long-term remission in 30% of patients, with an overall response rate of 50%.10 These regimens most commonly have included cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or CHOP-like treatment (EPOCH regimen [etoposide, prednisone, oncovin, cyclophosphamide, and doxorubicin hydrochloride]).3,10 A stem cell transplant can be considered in patients with recurrent and refractory disease, and it also has been shown to induce remission.4,17 In patients with a good response to therapy, the disease often can be controlled for long periods of time, with an estimated 5-year survival rate of 80%.15

This case highlights the diagnostic challenges and variable presentations of SPTCL. Dermatologists, oncologists, and dermatopathologists should be aware of this condition and consider it in the differential diagnosis of a patient with a hematologic malignancy and unremitting facial and/or body swelling without any other cause. The possibility of a secondary hematologic cancer in a patient with CLL also must be taken into consideration. Early diagnosis and treatment can minimize morbidity and induce remission in most patients.

References
  1. Gonzalez CL, Medeiros LJ, Braziel RM, et al. T-cell lymphoma involving subcutaneous tissue. a clinicopathologic entity commonly associated with hemophagocytic syndrome. Am J Surg Pathol. 1991;15:17-27.
  2. Willemze R, Jansen P, Cerroni L, et al. Subcutaneous panniculitis-like T-cell lymphoma: definition, classification, and prognostic factors: an EORTC Cutaneous Lymphoma Group Study of 83 cases. Blood. 2008;111:38-45.
  3. Parveen Z, Thompson K. Subcutaneous panniculitis-like T-cell lymphoma: redefinition of diagnostic criteria in the recent World Health Organization–European Organization for Research and Treatment of Cancer classification for cutaneous lymphomas. Arch Pathol Lab Med. 2009;133:303-308.
  4. Velez N, Ishizawar R, Dellaripa P, et al. Full facial edema: a novel presentation of subcutaneous panniculitis-like T-cell lymphoma. J Clin Oncol. 2012;30:e233-236.
  5. Asati D, Ingle V, Joshi D, et al. Subcutaneous panniculitis-like T-cell lymphoma with macrophage activation syndrome treated by cyclosporine and prednisolone. Indian Dermatol Online J. 2016;7:529-532.
  6. Fricker M, Dubach P, Helbing A, et al. Not all facial swellings are angioedemas! J Investig Allergol Clin Immunol. 2015;25:146-147.
  7. Kosari F, Akbarzadeh H. Local facial edema: a novel presentation of subcutaneous panniculitis-like T-cell lymphoma in a 30-year-old Iranian woman. Acta Med Iran. 2014;52:950-953.
  8. Bhojaraja M, Kistampally P, Udupa K, et al. Subcutaneous panniculitis-like T-cell lymphoma: a rare tumour. J Clin Diagn Res. 2016;10:OD29-OD30.
  9. Hashimoto R, Uchiyama M, Maeno T. Case report of subcutaneous panniculitis-like T-cell lymphoma complicated by eyelid swelling. BMC Ophthalmol. 2016;16:117.
  10. Chinello MN, Naviglio S, Remotti D, et al. Subcutaneous panniculitis-like T-cell lymphoma presenting with diffuse cutaneous edema in a 2-year-old child. J Pediatr Hematol Oncol. 2015;37:329-330.
  11. Chang TW, Weaver AL, Shanafelt TD, et al. Risk of cutaneous T-cell lymphoma in patients with chronic lymphocytic leukemia and other subtypes of non-Hodgkin lymphoma. Int J Dermatol. 2017;56:1125-1129.
  12. Chinello MN, Naviglio S, Remotti D, et al. Subcutaneous panniculitis-like T-cell lymphoma presenting with diffuse cutaneous edema in a 2-year-old child. J Pediatr Hematol Oncol. 2015;37:329-330.
  13. Jillella A, Day D, Severson K, et al. Non-Hodgkin’s lymphoma presenting as anasarca: probably mediated by tumor necrosis factor alpha (TNF-α). Leuk Lymphoma. 2000;38:419-422.
  14. Lee D-W, Yang J-H, Lee S-M, et al. Subcutaneous panniculitis-like T-cell lymphoma: a clinical and pathologic study of 14 Korean patients. Ann Dermatol. 2011;23:329-337.
  15. Jaffe ES. The 2008 WHO classification of lymphomas: implications for clinical practice and translational research [published online January 1, 2009]. Hematology Am Soc Hematol Educ Program. https://doi.org/10.1182/asheducation-2009.1.523
  16. Chang TW, Weaver AL, Shanafelt TD, et al. Risk of cutaneous T-cell lymphoma in patients with chronic lymphocytic leukemia and other subtypes of non-Hodgkin lymphoma. Int J Dermatol. 2017;56:1125-1129.
  17. Hall M, Sluzevich J, Snow J. Generalized subcutaneous panniculitis-like T-cell lymphoma following rituximab for hemolytic anemia in a patient with chronic lymphocytic leukemia. J Am Acad Dermatol. 2010;62(suppl 1):AB96.
References
  1. Gonzalez CL, Medeiros LJ, Braziel RM, et al. T-cell lymphoma involving subcutaneous tissue. a clinicopathologic entity commonly associated with hemophagocytic syndrome. Am J Surg Pathol. 1991;15:17-27.
  2. Willemze R, Jansen P, Cerroni L, et al. Subcutaneous panniculitis-like T-cell lymphoma: definition, classification, and prognostic factors: an EORTC Cutaneous Lymphoma Group Study of 83 cases. Blood. 2008;111:38-45.
  3. Parveen Z, Thompson K. Subcutaneous panniculitis-like T-cell lymphoma: redefinition of diagnostic criteria in the recent World Health Organization–European Organization for Research and Treatment of Cancer classification for cutaneous lymphomas. Arch Pathol Lab Med. 2009;133:303-308.
  4. Velez N, Ishizawar R, Dellaripa P, et al. Full facial edema: a novel presentation of subcutaneous panniculitis-like T-cell lymphoma. J Clin Oncol. 2012;30:e233-236.
  5. Asati D, Ingle V, Joshi D, et al. Subcutaneous panniculitis-like T-cell lymphoma with macrophage activation syndrome treated by cyclosporine and prednisolone. Indian Dermatol Online J. 2016;7:529-532.
  6. Fricker M, Dubach P, Helbing A, et al. Not all facial swellings are angioedemas! J Investig Allergol Clin Immunol. 2015;25:146-147.
  7. Kosari F, Akbarzadeh H. Local facial edema: a novel presentation of subcutaneous panniculitis-like T-cell lymphoma in a 30-year-old Iranian woman. Acta Med Iran. 2014;52:950-953.
  8. Bhojaraja M, Kistampally P, Udupa K, et al. Subcutaneous panniculitis-like T-cell lymphoma: a rare tumour. J Clin Diagn Res. 2016;10:OD29-OD30.
  9. Hashimoto R, Uchiyama M, Maeno T. Case report of subcutaneous panniculitis-like T-cell lymphoma complicated by eyelid swelling. BMC Ophthalmol. 2016;16:117.
  10. Chinello MN, Naviglio S, Remotti D, et al. Subcutaneous panniculitis-like T-cell lymphoma presenting with diffuse cutaneous edema in a 2-year-old child. J Pediatr Hematol Oncol. 2015;37:329-330.
  11. Chang TW, Weaver AL, Shanafelt TD, et al. Risk of cutaneous T-cell lymphoma in patients with chronic lymphocytic leukemia and other subtypes of non-Hodgkin lymphoma. Int J Dermatol. 2017;56:1125-1129.
  12. Chinello MN, Naviglio S, Remotti D, et al. Subcutaneous panniculitis-like T-cell lymphoma presenting with diffuse cutaneous edema in a 2-year-old child. J Pediatr Hematol Oncol. 2015;37:329-330.
  13. Jillella A, Day D, Severson K, et al. Non-Hodgkin’s lymphoma presenting as anasarca: probably mediated by tumor necrosis factor alpha (TNF-α). Leuk Lymphoma. 2000;38:419-422.
  14. Lee D-W, Yang J-H, Lee S-M, et al. Subcutaneous panniculitis-like T-cell lymphoma: a clinical and pathologic study of 14 Korean patients. Ann Dermatol. 2011;23:329-337.
  15. Jaffe ES. The 2008 WHO classification of lymphomas: implications for clinical practice and translational research [published online January 1, 2009]. Hematology Am Soc Hematol Educ Program. https://doi.org/10.1182/asheducation-2009.1.523
  16. Chang TW, Weaver AL, Shanafelt TD, et al. Risk of cutaneous T-cell lymphoma in patients with chronic lymphocytic leukemia and other subtypes of non-Hodgkin lymphoma. Int J Dermatol. 2017;56:1125-1129.
  17. Hall M, Sluzevich J, Snow J. Generalized subcutaneous panniculitis-like T-cell lymphoma following rituximab for hemolytic anemia in a patient with chronic lymphocytic leukemia. J Am Acad Dermatol. 2010;62(suppl 1):AB96.
Issue
Cutis - 111(3)
Issue
Cutis - 111(3)
Page Number
E28-E31
Page Number
E28-E31
Publications
Publications
Topics
Article Type
Display Headline
Subcutaneous Panniculitic T-cell Lymphoma Presenting With Anasarca in a Patient With Known Chronic Lymphocytic Leukemia
Display Headline
Subcutaneous Panniculitic T-cell Lymphoma Presenting With Anasarca in a Patient With Known Chronic Lymphocytic Leukemia
Sections
Inside the Article

Practice Points

  • Subcutaneous panniculitic T-cell lymphoma (SPTCL) is a rare type of cutaneous T-cell lymphoma that may be complicated by fatal hemophagocytic syndrome.
  • Patients typically present with deep-seated plaques or nodules that may be masked by localized edema.
  • A biopsy is necessary to diagnose SPTCL, as well as to assess the degree of cellular atypia, fat necrosis, karyorrhexis, cytophagia, and angioinvasion to distinguish it from other panniculitides.
  • In patients with a known hematologic malignancy, a secondary malignancy must be considered in the differential diagnosis of paraneoplastic edema.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Progressive Primary Cutaneous Nocardiosis in an Immunocompetent Patient

Article Type
Changed
Fri, 03/31/2023 - 09:51
Display Headline
Progressive Primary Cutaneous Nocardiosis in an Immunocompetent Patient

To the Editor:

The organisms of the genus Nocardia are gram-positive, ubiquitous, aerobic actinomycetes found worldwide in soil, decaying organic material, and water.1 The genus Nocardia includes more than 50 species; some species, such as Nocardia asteroides, Nocardia farcinica, Nocardia nova, and Nocardia brasiliensis, are the cause of nocardiosis in humans and animals.2,3 Nocardiosis is a rare and opportunistic infection that predominantly affects immunocompromised individuals; however, up to 30% of infections can occur in immunocompetent hosts.4 Nocardiosis can manifest in 3 disease forms: cutaneous, pulmonary, or disseminated. Cutaneous nocardiosis commonly develops in immunocompetent individuals who have experienced a predisposing traumatic injury to the skin,5 and it can exhibit a diverse variety of clinical manifestations, making diagnosis difficult. We describe a case of serious progressive primary cutaneous nocardiosis with an unusual presentation in an immunocompetent patient.

A 26-year-old immunocompetent man presented with pain, swelling, nodules, abscesses, ulcers, and sinus drainage of the left arm. The left elbow lesion initially developed at the site of a trauma 6 years prior that was painless but was contaminated with mossy soil. The condition slowly progressed over the next 2 years, and the patient experienced increased swelling and eventually developed multiple draining sinus tracts. Over the next 4 years, the lesions multiplied, spreading to the forearm and upper arm; associated severe pain and swelling at the elbow and wrist joint developed. The patient sought medical care at a local hospital and subsequently was diagnosed with suspected cutaneous tuberculosis. The patient was empirically treated with a 6-month course of isoniazid, rifampicin, pyrazinamide, and ethambutol; however, the lesions continued to progress and worsen. The patient had to stop antibiotic treatment because of substantially elevated alanine aminotransferase and aspartate aminotransferase levels.

He subsequently was evaluated at our hospital. He had no notable medical history and was afebrile. Physical examination revealed multiple erythematous nodules, abscesses, and ulcers on the left arm. There were several nodules with open sinus tracts and seropurulent crusts along with numerous atrophic, ovoid, stellate scars. Other nodules and ulcers with purulent drainage were located along the lymphatic nodes extending up the patient’s left forearm (Figure 1A). The yellowish-white pus discharge from several active sinuses contained no apparent granules. The lesions were densely distributed along the elbow, wrist, and shoulder, which resulted in associated skin swelling and restricted joint movement. The left axillary lymph nodes were enlarged.

Progressive primary cutaneous nocardiosis.
FIGURE 1. Progressive primary cutaneous nocardiosis. A, Skin lesions on the patient’s left forearm at the initial visit. B, After 6 months of antibiotic treatment, the cutaneous lesions and left limb swelling completely subsided.

Laboratory analyses revealed a hemoglobin level of 9.6 g/dL (reference range, 13–17.5 g/dL), platelet count of 621×109/L (reference range, 125–350×109/L), and leukocyte count of 14.3×109/L (reference range, 3.5–9.5 ×109/L). C-reactive protein level was 88.4 mg/L (reference range, 0–10 mg/L). Blood, renal, and liver tests, as well as tumor marker, peripheral blood lymphocyte subset, immunoglobulin, and complement results were within reference ranges. Results for Treponema pallidum and HIV antibody tests were negative. Hepatitis B virus markers were positive for hepatitis B surface antigen, hepatitis B e antigen, and hepatitis B core antibody, and the serum concentration of hepatitis B virus DNA was 3.12×107 IU/mL (reference range, <5×102 IU/mL). Computed tomography of the chest and cranium were unremarkable. Ultrasonography of the left arm revealed multiple vertical sinus tracts and several horizontal communicating branches that were accompanied by worm-eaten bone destruction (Figure 2).

Ultrasonography of the patient’s left arm revealed multiple vertical sinus tracts and several horizontal communicating branches.
FIGURE 2. Ultrasonography of the patient’s left arm revealed multiple vertical sinus tracts and several horizontal communicating branches.

Additional testing included histopathologic staining of a skin tissue specimen—hematoxylin and eosin, periodic acid–Schiff, and acid-fast staining—showed nonspecific, diffuse, inflammatory cell infiltration suggestive of chronic suppurative granuloma (Figure 3) but failed to reveal any special strains or organisms. Gram stain examination of the purulent fluid collected from the subcutaneous tissue showed no apparent positive bacillus or filamentous granules. The specimen was then inoculated on Sabouraud dextrose agar and Lowenstein-Jensen medium for fungus and mycobacteria culture, respectively. After 5 days, chalky, yellow, adherent colonies were observed on the Löwenstein-Jensen medium, and after 26 days, yellow crinkled colonies were observed on Sabouraud dextrose agar. The colonies were then inoculated on Columbia blood agar and incubated for 1 week to aid in the identification of organisms. Growth of yellow colonies that were adherent to the agar, moist, and smooth with a velvety surface, as well as a characteristic moldy odor resulted. Gram staining revealed gram-positive, thin, and beaded branching filaments (Figure 4). Based on colony characteristics, physiological properties, and biochemical tests, the isolate was identified as Nocardia. Results of further investigations employing polymerase chain reaction analysis of the skin specimen and bacterial colonies using a Nocardia genus 596-bp fragment of 16S ribosomal RNA primer (forward primer NG1: 5’-ACCGACCACAAGGGG-3’, reverse primer NG2: 5’-GGTTGTAACCTCTTCGA-3’)6 were completely consistent with the reference for identification of N brasiliensis. Evaluation of these results led to a diagnosis of cutaneous nocardiosis after traumatic inoculation.

Histopathology showed irregular hyperplasia of epidermal cells and infiltration of inflammatory cells
FIGURE 3. Histopathology showed irregular hyperplasia of epidermal cells and infiltration of inflammatory cells (H&E, original magnification ×40).

Because there was a high suspicion of actinophytosis or nocardiosis at admission, the patient received a combination antibiotic treatment with intravenous aqueous penicillin (4 million U every 4 hours) and oral trimethoprim-sulfamethoxazole (160/800 mg twice daily). Subsequently, treatment was changed to a combination of oral trimethoprim-sulfamethoxazole (160/800 mg twice daily) and moxifloxacin (400 mg once daily) based on pathogen identification and antibiotic sensitivity testing. After 1 month of treatment, the cutaneous lesions and left limb swelling dramatically improved and purulent drainage ceased, though some scarring occurred during the healing process. In addition, the mobility of the affected shoulder, elbow, and wrist joints slightly improved. Notable improvement in the mobility and swelling of the joints was observed at 6-month follow-up (Figure 1B). The patient continues to be monitored on an outpatient basis.

Gram stain from colonies grown on Columbia blood agar showed branching, filamentous, gram-positive bacilli
FIGURE 4. Gram stain from colonies grown on Columbia blood agar showed branching, filamentous, gram-positive bacilli (original magnification ×1000).

Cutaneous nocardiosis is a disfiguring granulomatous infection involving cutaneous and subcutaneous tissue that can progress to cause injury to viscera and bone.7 It has been called one of the great imitators because cutaneous nocardiosis can present in multiple forms,8,9 including mycetoma, sporotrichoid infection, superficial skin infection, and disseminated infection with cutaneous involvement. The differential diagnoses of cutaneous nocardiosis are broad and include tuberculosis; actinomycosis; deep fungal infections such as sporotrichosis, blastomycosis, phaeohyphomycosis, histoplasmosis, and coccidioidomycosis; other bacterial causes of cellulitis, abscess, or ecthyma; and malignancies.10 The principle method of diagnosis is the identification of Nocardia from the infection site.

 

 

Our patient ultimately was diagnosed with primary cutaneous nocardiosis resulting from a traumatic injury to the skin that was contaminated with soil. The clinical manifestation pattern was a compound type, including both mycetoma and sporotrichoid infections. Initially, Nocardia mycetoma occurred with subcutaneous infection by direct extension10,11 and appeared as dense, predominantly painless, swollen lesions. After 4 years, the skin lesions continued to spread linearly to the patient’s upper arm and forearm and manifested as the sporotrichoid infection type with painful swollen lesions at the site of inoculation and painful enlargement of the ipsilateral axillary lymph node.

Although nocardiosis is found worldwide, it is endemic to tropical and subtropical regions such as India, Africa, Southeast Asia, and Latin America.12 Nocardiosis most often is observed in individuals aged 20 to 40 years. It affects men more than women, and it commonly occurs in field laborers and cultivators whose occupations involve direct contact with the soil.13 Most lesions are found on the lower extremities, though localized nocardiosis infections can occur in other areas such as the neck, breasts, back, buttocks, and elbows.

Our patient initially was misdiagnosed, and treatment was delayed for several reasons. First, nocardiosis is not common in China, and most clinicians are unfamiliar with the disease. Second, the related lesions do not have specific features, and our patient had a complex clinical presentation that included mycetoma and sporotrichoid infection. Third, the characteristic grain of Nocardia species is small but that of N brasiliensis is even smaller (approximately 0.1–0.2 mm in diameter), which makes visualization difficult in both histopathologic and microbiologic examinations.14 The histopathologic examination results of our patient in the local hospital were nonspecific. Fourth, our patient did not initially go to the hospital but instead purchased some over-the-counter antibiotic ointments for external application because the lesions were painless. Moreover, microbiologic smear and culture examinations were not conducted in the local hospital before administering antituberculosis treatment to the patient. Instead, a polymerase chain reaction examination of skin lesion tissue for tubercle bacilli and atypical mycobacteria was negative. These findings imply that the traditional microbial smear and culture evaluations cannot be omitted. Furthermore, culture examinations should be conducted on multiple skin tissue and purulent fluid specimens to increase the likelihood of detection. These cultures should be monitored for at least 2 to 4 weeks because Nocardia is a slow-growing organism.10

The optimal antimicrobial treatment regimens for nocardiosis have not been firmly established.15 Trimethoprim-sulfamethoxazole is regarded as the first-line antimicrobial agent for treatment of nocardial infections. The optimal duration of antimicrobial therapy for nocardiosis also has not been determined, and the treatment regimen depends on the severity and extent of the infection as well as on the presence of infection-related complications. The main complication is bone involvement. Notable bony changes include periosteal thickening, osteoporosis, and osteolysis.

We considered the severity of skin lesions and bone marrow invasion in our patient and planned to treat him continually with oral trimethoprim-sulfamethoxazole according to the in vitro drug susceptibility test. The patient showed clinical improvement after 1 month of treatment, and he continued to improve after 6 months of treatment. To prevent recurrence, we found it necessary to treat the patient with a long-term antibiotic course over 6 to 12 months.16

Cutaneous nocardiosis remains a diagnostic challenge because of its nonspecific and diverse clinical and histopathological presentations. Diagnosis is further complicated by the inherent difficulty of cultivating and identifying the clinical isolate in the laboratory. A high degree of clinical suspicion followed by successful identification of the organism by a laboratory technologist will aid in the early diagnosis and treatment of the infection, ultimately reducing the risk for complications and morbidity.

References
  1. McNeil MM, Brown JM. The medically important aerobic actinomycetes: epidemiology and microbiology. Clin Microbiol Rev. 1994;7:357-417.
  2. Brown-Elliott BA, Brown JM, Conville PS, et al. Clinical and laboratory features of the Nocardia spp. based on current molecular taxonomy. Clin Microbiol Rev. 2006;19:259-282.
  3. Fatahi-Bafghi M. Nocardiosis from 1888 to 2017. Microb Pathog. 2018;114:369-384.
  4. Beaman BL, Burnside J, Edwards B, et al. Nocardial infections in the United States, 1972-1974. J Infect Dis. 1976;134:286-289.
  5. Lerner PI. Nocardiosis. Clin Infect Dis. 1996;22:891-903.
  6. Laurent FJ, Provost F, Boiron P. Rapid identification of clinically relevant Nocardia species to genus level by 16S rRNA gene PCR. J Clin Microbiol. 1999;37:99-102.
  7. Nguyen NM, Sink JR, Carter AJ, et al. Nocardiosis incognito: primary cutaneous nocardiosis with extension to myositis and pleural infection. JAAD Case Rep. 2018;4:33-35.
  8. Sharna NL, Mahajan VK, Agarwal S, et al. Nocardial mycetoma: diverse clinical presentations. Indian J Dermatol Venereol Leprol. 2008;74:635-640.
  9. Huang L, Chen X, Xu H, et al. Clinical features, identification, antimicrobial resistance patterns of Nocardia species in China: 2009-2017. Diagn Microbiol Infect Dis. 2019;94:165-172.
  10. Bonifaz A, Tirado-Sánchez A, Calderón L, et al. Mycetoma: experience of 482 cases in a single center in Mexico. PLoS Negl Trop Dis. 2014;8:E3102.
  11. Welsh O, Vero-Cabrera L, Salinas-Carmona MC. Mycetoma. Clin Dermatol. 2007;25:195-202.
  12. Nenoff P, van de Sande WWJ, Fahal AH, et al. Eumycetoma and actinomycetoma—an update on causative agents, epidemiology, pathogenesis, diagnostics and therapy. J Eur Acad Dermatol Venereol. 2015;29:1873-1883.
  13. Emmanuel P, Dumre SP, John S, et al. Mycetoma: a clinical dilemma in resource limited settings. Ann Clin Microbiol Antimicrob. 2018;17:35.
  14. Reis CMS, Reis-Filho EGM. Mycetomas: an epidemiological, etiological, clinical, laboratory and therapeutic review. An Bras Dermatol. 2018;93:8-18.
  15. Wilson JW. Nocardiosis: updates and clinical overview. Mayo Clin Proc. 2012;87:403-407.
  16. Welsh O, Vera-Cabrera L, Salinas-Carmona MC. Current treatment for Nocardia infections. Expert Opin Pharmacother. 2013;14:2387-2398.
Article PDF
Author and Disclosure Information

From Shanghai Dermatology Hospital, China. Drs. Yu, Song, Tan, and Yang are from the Department of Medical Mycology; Dr. Liu is from the Department of Pathology; Dr. Yanrui Gao is from the Department of Skin & Cosmetic Research; and Drs. Yunlu Gao and Ding are from the Department of Dermatology.

The authors report no conflict of interest.

This work was supported by the Science and Technology Commission of Shanghai Municipality (No. 18411969700). The funder drafted the manuscript and collected the clinical data.

Correspondence: Lianjuan Yang, MD, Department of Medical Mycology, Shanghai Dermatology Hospital, 1278 Baode Rd, Shanghai SH021, China (lianjuanyang@163.com).

Issue
Cutis - 111(3)
Publications
Topics
Page Number
E22-E25
Sections
Author and Disclosure Information

From Shanghai Dermatology Hospital, China. Drs. Yu, Song, Tan, and Yang are from the Department of Medical Mycology; Dr. Liu is from the Department of Pathology; Dr. Yanrui Gao is from the Department of Skin & Cosmetic Research; and Drs. Yunlu Gao and Ding are from the Department of Dermatology.

The authors report no conflict of interest.

This work was supported by the Science and Technology Commission of Shanghai Municipality (No. 18411969700). The funder drafted the manuscript and collected the clinical data.

Correspondence: Lianjuan Yang, MD, Department of Medical Mycology, Shanghai Dermatology Hospital, 1278 Baode Rd, Shanghai SH021, China (lianjuanyang@163.com).

Author and Disclosure Information

From Shanghai Dermatology Hospital, China. Drs. Yu, Song, Tan, and Yang are from the Department of Medical Mycology; Dr. Liu is from the Department of Pathology; Dr. Yanrui Gao is from the Department of Skin & Cosmetic Research; and Drs. Yunlu Gao and Ding are from the Department of Dermatology.

The authors report no conflict of interest.

This work was supported by the Science and Technology Commission of Shanghai Municipality (No. 18411969700). The funder drafted the manuscript and collected the clinical data.

Correspondence: Lianjuan Yang, MD, Department of Medical Mycology, Shanghai Dermatology Hospital, 1278 Baode Rd, Shanghai SH021, China (lianjuanyang@163.com).

Article PDF
Article PDF

To the Editor:

The organisms of the genus Nocardia are gram-positive, ubiquitous, aerobic actinomycetes found worldwide in soil, decaying organic material, and water.1 The genus Nocardia includes more than 50 species; some species, such as Nocardia asteroides, Nocardia farcinica, Nocardia nova, and Nocardia brasiliensis, are the cause of nocardiosis in humans and animals.2,3 Nocardiosis is a rare and opportunistic infection that predominantly affects immunocompromised individuals; however, up to 30% of infections can occur in immunocompetent hosts.4 Nocardiosis can manifest in 3 disease forms: cutaneous, pulmonary, or disseminated. Cutaneous nocardiosis commonly develops in immunocompetent individuals who have experienced a predisposing traumatic injury to the skin,5 and it can exhibit a diverse variety of clinical manifestations, making diagnosis difficult. We describe a case of serious progressive primary cutaneous nocardiosis with an unusual presentation in an immunocompetent patient.

A 26-year-old immunocompetent man presented with pain, swelling, nodules, abscesses, ulcers, and sinus drainage of the left arm. The left elbow lesion initially developed at the site of a trauma 6 years prior that was painless but was contaminated with mossy soil. The condition slowly progressed over the next 2 years, and the patient experienced increased swelling and eventually developed multiple draining sinus tracts. Over the next 4 years, the lesions multiplied, spreading to the forearm and upper arm; associated severe pain and swelling at the elbow and wrist joint developed. The patient sought medical care at a local hospital and subsequently was diagnosed with suspected cutaneous tuberculosis. The patient was empirically treated with a 6-month course of isoniazid, rifampicin, pyrazinamide, and ethambutol; however, the lesions continued to progress and worsen. The patient had to stop antibiotic treatment because of substantially elevated alanine aminotransferase and aspartate aminotransferase levels.

He subsequently was evaluated at our hospital. He had no notable medical history and was afebrile. Physical examination revealed multiple erythematous nodules, abscesses, and ulcers on the left arm. There were several nodules with open sinus tracts and seropurulent crusts along with numerous atrophic, ovoid, stellate scars. Other nodules and ulcers with purulent drainage were located along the lymphatic nodes extending up the patient’s left forearm (Figure 1A). The yellowish-white pus discharge from several active sinuses contained no apparent granules. The lesions were densely distributed along the elbow, wrist, and shoulder, which resulted in associated skin swelling and restricted joint movement. The left axillary lymph nodes were enlarged.

Progressive primary cutaneous nocardiosis.
FIGURE 1. Progressive primary cutaneous nocardiosis. A, Skin lesions on the patient’s left forearm at the initial visit. B, After 6 months of antibiotic treatment, the cutaneous lesions and left limb swelling completely subsided.

Laboratory analyses revealed a hemoglobin level of 9.6 g/dL (reference range, 13–17.5 g/dL), platelet count of 621×109/L (reference range, 125–350×109/L), and leukocyte count of 14.3×109/L (reference range, 3.5–9.5 ×109/L). C-reactive protein level was 88.4 mg/L (reference range, 0–10 mg/L). Blood, renal, and liver tests, as well as tumor marker, peripheral blood lymphocyte subset, immunoglobulin, and complement results were within reference ranges. Results for Treponema pallidum and HIV antibody tests were negative. Hepatitis B virus markers were positive for hepatitis B surface antigen, hepatitis B e antigen, and hepatitis B core antibody, and the serum concentration of hepatitis B virus DNA was 3.12×107 IU/mL (reference range, <5×102 IU/mL). Computed tomography of the chest and cranium were unremarkable. Ultrasonography of the left arm revealed multiple vertical sinus tracts and several horizontal communicating branches that were accompanied by worm-eaten bone destruction (Figure 2).

Ultrasonography of the patient’s left arm revealed multiple vertical sinus tracts and several horizontal communicating branches.
FIGURE 2. Ultrasonography of the patient’s left arm revealed multiple vertical sinus tracts and several horizontal communicating branches.

Additional testing included histopathologic staining of a skin tissue specimen—hematoxylin and eosin, periodic acid–Schiff, and acid-fast staining—showed nonspecific, diffuse, inflammatory cell infiltration suggestive of chronic suppurative granuloma (Figure 3) but failed to reveal any special strains or organisms. Gram stain examination of the purulent fluid collected from the subcutaneous tissue showed no apparent positive bacillus or filamentous granules. The specimen was then inoculated on Sabouraud dextrose agar and Lowenstein-Jensen medium for fungus and mycobacteria culture, respectively. After 5 days, chalky, yellow, adherent colonies were observed on the Löwenstein-Jensen medium, and after 26 days, yellow crinkled colonies were observed on Sabouraud dextrose agar. The colonies were then inoculated on Columbia blood agar and incubated for 1 week to aid in the identification of organisms. Growth of yellow colonies that were adherent to the agar, moist, and smooth with a velvety surface, as well as a characteristic moldy odor resulted. Gram staining revealed gram-positive, thin, and beaded branching filaments (Figure 4). Based on colony characteristics, physiological properties, and biochemical tests, the isolate was identified as Nocardia. Results of further investigations employing polymerase chain reaction analysis of the skin specimen and bacterial colonies using a Nocardia genus 596-bp fragment of 16S ribosomal RNA primer (forward primer NG1: 5’-ACCGACCACAAGGGG-3’, reverse primer NG2: 5’-GGTTGTAACCTCTTCGA-3’)6 were completely consistent with the reference for identification of N brasiliensis. Evaluation of these results led to a diagnosis of cutaneous nocardiosis after traumatic inoculation.

Histopathology showed irregular hyperplasia of epidermal cells and infiltration of inflammatory cells
FIGURE 3. Histopathology showed irregular hyperplasia of epidermal cells and infiltration of inflammatory cells (H&E, original magnification ×40).

Because there was a high suspicion of actinophytosis or nocardiosis at admission, the patient received a combination antibiotic treatment with intravenous aqueous penicillin (4 million U every 4 hours) and oral trimethoprim-sulfamethoxazole (160/800 mg twice daily). Subsequently, treatment was changed to a combination of oral trimethoprim-sulfamethoxazole (160/800 mg twice daily) and moxifloxacin (400 mg once daily) based on pathogen identification and antibiotic sensitivity testing. After 1 month of treatment, the cutaneous lesions and left limb swelling dramatically improved and purulent drainage ceased, though some scarring occurred during the healing process. In addition, the mobility of the affected shoulder, elbow, and wrist joints slightly improved. Notable improvement in the mobility and swelling of the joints was observed at 6-month follow-up (Figure 1B). The patient continues to be monitored on an outpatient basis.

Gram stain from colonies grown on Columbia blood agar showed branching, filamentous, gram-positive bacilli
FIGURE 4. Gram stain from colonies grown on Columbia blood agar showed branching, filamentous, gram-positive bacilli (original magnification ×1000).

Cutaneous nocardiosis is a disfiguring granulomatous infection involving cutaneous and subcutaneous tissue that can progress to cause injury to viscera and bone.7 It has been called one of the great imitators because cutaneous nocardiosis can present in multiple forms,8,9 including mycetoma, sporotrichoid infection, superficial skin infection, and disseminated infection with cutaneous involvement. The differential diagnoses of cutaneous nocardiosis are broad and include tuberculosis; actinomycosis; deep fungal infections such as sporotrichosis, blastomycosis, phaeohyphomycosis, histoplasmosis, and coccidioidomycosis; other bacterial causes of cellulitis, abscess, or ecthyma; and malignancies.10 The principle method of diagnosis is the identification of Nocardia from the infection site.

 

 

Our patient ultimately was diagnosed with primary cutaneous nocardiosis resulting from a traumatic injury to the skin that was contaminated with soil. The clinical manifestation pattern was a compound type, including both mycetoma and sporotrichoid infections. Initially, Nocardia mycetoma occurred with subcutaneous infection by direct extension10,11 and appeared as dense, predominantly painless, swollen lesions. After 4 years, the skin lesions continued to spread linearly to the patient’s upper arm and forearm and manifested as the sporotrichoid infection type with painful swollen lesions at the site of inoculation and painful enlargement of the ipsilateral axillary lymph node.

Although nocardiosis is found worldwide, it is endemic to tropical and subtropical regions such as India, Africa, Southeast Asia, and Latin America.12 Nocardiosis most often is observed in individuals aged 20 to 40 years. It affects men more than women, and it commonly occurs in field laborers and cultivators whose occupations involve direct contact with the soil.13 Most lesions are found on the lower extremities, though localized nocardiosis infections can occur in other areas such as the neck, breasts, back, buttocks, and elbows.

Our patient initially was misdiagnosed, and treatment was delayed for several reasons. First, nocardiosis is not common in China, and most clinicians are unfamiliar with the disease. Second, the related lesions do not have specific features, and our patient had a complex clinical presentation that included mycetoma and sporotrichoid infection. Third, the characteristic grain of Nocardia species is small but that of N brasiliensis is even smaller (approximately 0.1–0.2 mm in diameter), which makes visualization difficult in both histopathologic and microbiologic examinations.14 The histopathologic examination results of our patient in the local hospital were nonspecific. Fourth, our patient did not initially go to the hospital but instead purchased some over-the-counter antibiotic ointments for external application because the lesions were painless. Moreover, microbiologic smear and culture examinations were not conducted in the local hospital before administering antituberculosis treatment to the patient. Instead, a polymerase chain reaction examination of skin lesion tissue for tubercle bacilli and atypical mycobacteria was negative. These findings imply that the traditional microbial smear and culture evaluations cannot be omitted. Furthermore, culture examinations should be conducted on multiple skin tissue and purulent fluid specimens to increase the likelihood of detection. These cultures should be monitored for at least 2 to 4 weeks because Nocardia is a slow-growing organism.10

The optimal antimicrobial treatment regimens for nocardiosis have not been firmly established.15 Trimethoprim-sulfamethoxazole is regarded as the first-line antimicrobial agent for treatment of nocardial infections. The optimal duration of antimicrobial therapy for nocardiosis also has not been determined, and the treatment regimen depends on the severity and extent of the infection as well as on the presence of infection-related complications. The main complication is bone involvement. Notable bony changes include periosteal thickening, osteoporosis, and osteolysis.

We considered the severity of skin lesions and bone marrow invasion in our patient and planned to treat him continually with oral trimethoprim-sulfamethoxazole according to the in vitro drug susceptibility test. The patient showed clinical improvement after 1 month of treatment, and he continued to improve after 6 months of treatment. To prevent recurrence, we found it necessary to treat the patient with a long-term antibiotic course over 6 to 12 months.16

Cutaneous nocardiosis remains a diagnostic challenge because of its nonspecific and diverse clinical and histopathological presentations. Diagnosis is further complicated by the inherent difficulty of cultivating and identifying the clinical isolate in the laboratory. A high degree of clinical suspicion followed by successful identification of the organism by a laboratory technologist will aid in the early diagnosis and treatment of the infection, ultimately reducing the risk for complications and morbidity.

To the Editor:

The organisms of the genus Nocardia are gram-positive, ubiquitous, aerobic actinomycetes found worldwide in soil, decaying organic material, and water.1 The genus Nocardia includes more than 50 species; some species, such as Nocardia asteroides, Nocardia farcinica, Nocardia nova, and Nocardia brasiliensis, are the cause of nocardiosis in humans and animals.2,3 Nocardiosis is a rare and opportunistic infection that predominantly affects immunocompromised individuals; however, up to 30% of infections can occur in immunocompetent hosts.4 Nocardiosis can manifest in 3 disease forms: cutaneous, pulmonary, or disseminated. Cutaneous nocardiosis commonly develops in immunocompetent individuals who have experienced a predisposing traumatic injury to the skin,5 and it can exhibit a diverse variety of clinical manifestations, making diagnosis difficult. We describe a case of serious progressive primary cutaneous nocardiosis with an unusual presentation in an immunocompetent patient.

A 26-year-old immunocompetent man presented with pain, swelling, nodules, abscesses, ulcers, and sinus drainage of the left arm. The left elbow lesion initially developed at the site of a trauma 6 years prior that was painless but was contaminated with mossy soil. The condition slowly progressed over the next 2 years, and the patient experienced increased swelling and eventually developed multiple draining sinus tracts. Over the next 4 years, the lesions multiplied, spreading to the forearm and upper arm; associated severe pain and swelling at the elbow and wrist joint developed. The patient sought medical care at a local hospital and subsequently was diagnosed with suspected cutaneous tuberculosis. The patient was empirically treated with a 6-month course of isoniazid, rifampicin, pyrazinamide, and ethambutol; however, the lesions continued to progress and worsen. The patient had to stop antibiotic treatment because of substantially elevated alanine aminotransferase and aspartate aminotransferase levels.

He subsequently was evaluated at our hospital. He had no notable medical history and was afebrile. Physical examination revealed multiple erythematous nodules, abscesses, and ulcers on the left arm. There were several nodules with open sinus tracts and seropurulent crusts along with numerous atrophic, ovoid, stellate scars. Other nodules and ulcers with purulent drainage were located along the lymphatic nodes extending up the patient’s left forearm (Figure 1A). The yellowish-white pus discharge from several active sinuses contained no apparent granules. The lesions were densely distributed along the elbow, wrist, and shoulder, which resulted in associated skin swelling and restricted joint movement. The left axillary lymph nodes were enlarged.

Progressive primary cutaneous nocardiosis.
FIGURE 1. Progressive primary cutaneous nocardiosis. A, Skin lesions on the patient’s left forearm at the initial visit. B, After 6 months of antibiotic treatment, the cutaneous lesions and left limb swelling completely subsided.

Laboratory analyses revealed a hemoglobin level of 9.6 g/dL (reference range, 13–17.5 g/dL), platelet count of 621×109/L (reference range, 125–350×109/L), and leukocyte count of 14.3×109/L (reference range, 3.5–9.5 ×109/L). C-reactive protein level was 88.4 mg/L (reference range, 0–10 mg/L). Blood, renal, and liver tests, as well as tumor marker, peripheral blood lymphocyte subset, immunoglobulin, and complement results were within reference ranges. Results for Treponema pallidum and HIV antibody tests were negative. Hepatitis B virus markers were positive for hepatitis B surface antigen, hepatitis B e antigen, and hepatitis B core antibody, and the serum concentration of hepatitis B virus DNA was 3.12×107 IU/mL (reference range, <5×102 IU/mL). Computed tomography of the chest and cranium were unremarkable. Ultrasonography of the left arm revealed multiple vertical sinus tracts and several horizontal communicating branches that were accompanied by worm-eaten bone destruction (Figure 2).

Ultrasonography of the patient’s left arm revealed multiple vertical sinus tracts and several horizontal communicating branches.
FIGURE 2. Ultrasonography of the patient’s left arm revealed multiple vertical sinus tracts and several horizontal communicating branches.

Additional testing included histopathologic staining of a skin tissue specimen—hematoxylin and eosin, periodic acid–Schiff, and acid-fast staining—showed nonspecific, diffuse, inflammatory cell infiltration suggestive of chronic suppurative granuloma (Figure 3) but failed to reveal any special strains or organisms. Gram stain examination of the purulent fluid collected from the subcutaneous tissue showed no apparent positive bacillus or filamentous granules. The specimen was then inoculated on Sabouraud dextrose agar and Lowenstein-Jensen medium for fungus and mycobacteria culture, respectively. After 5 days, chalky, yellow, adherent colonies were observed on the Löwenstein-Jensen medium, and after 26 days, yellow crinkled colonies were observed on Sabouraud dextrose agar. The colonies were then inoculated on Columbia blood agar and incubated for 1 week to aid in the identification of organisms. Growth of yellow colonies that were adherent to the agar, moist, and smooth with a velvety surface, as well as a characteristic moldy odor resulted. Gram staining revealed gram-positive, thin, and beaded branching filaments (Figure 4). Based on colony characteristics, physiological properties, and biochemical tests, the isolate was identified as Nocardia. Results of further investigations employing polymerase chain reaction analysis of the skin specimen and bacterial colonies using a Nocardia genus 596-bp fragment of 16S ribosomal RNA primer (forward primer NG1: 5’-ACCGACCACAAGGGG-3’, reverse primer NG2: 5’-GGTTGTAACCTCTTCGA-3’)6 were completely consistent with the reference for identification of N brasiliensis. Evaluation of these results led to a diagnosis of cutaneous nocardiosis after traumatic inoculation.

Histopathology showed irregular hyperplasia of epidermal cells and infiltration of inflammatory cells
FIGURE 3. Histopathology showed irregular hyperplasia of epidermal cells and infiltration of inflammatory cells (H&E, original magnification ×40).

Because there was a high suspicion of actinophytosis or nocardiosis at admission, the patient received a combination antibiotic treatment with intravenous aqueous penicillin (4 million U every 4 hours) and oral trimethoprim-sulfamethoxazole (160/800 mg twice daily). Subsequently, treatment was changed to a combination of oral trimethoprim-sulfamethoxazole (160/800 mg twice daily) and moxifloxacin (400 mg once daily) based on pathogen identification and antibiotic sensitivity testing. After 1 month of treatment, the cutaneous lesions and left limb swelling dramatically improved and purulent drainage ceased, though some scarring occurred during the healing process. In addition, the mobility of the affected shoulder, elbow, and wrist joints slightly improved. Notable improvement in the mobility and swelling of the joints was observed at 6-month follow-up (Figure 1B). The patient continues to be monitored on an outpatient basis.

Gram stain from colonies grown on Columbia blood agar showed branching, filamentous, gram-positive bacilli
FIGURE 4. Gram stain from colonies grown on Columbia blood agar showed branching, filamentous, gram-positive bacilli (original magnification ×1000).

Cutaneous nocardiosis is a disfiguring granulomatous infection involving cutaneous and subcutaneous tissue that can progress to cause injury to viscera and bone.7 It has been called one of the great imitators because cutaneous nocardiosis can present in multiple forms,8,9 including mycetoma, sporotrichoid infection, superficial skin infection, and disseminated infection with cutaneous involvement. The differential diagnoses of cutaneous nocardiosis are broad and include tuberculosis; actinomycosis; deep fungal infections such as sporotrichosis, blastomycosis, phaeohyphomycosis, histoplasmosis, and coccidioidomycosis; other bacterial causes of cellulitis, abscess, or ecthyma; and malignancies.10 The principle method of diagnosis is the identification of Nocardia from the infection site.

 

 

Our patient ultimately was diagnosed with primary cutaneous nocardiosis resulting from a traumatic injury to the skin that was contaminated with soil. The clinical manifestation pattern was a compound type, including both mycetoma and sporotrichoid infections. Initially, Nocardia mycetoma occurred with subcutaneous infection by direct extension10,11 and appeared as dense, predominantly painless, swollen lesions. After 4 years, the skin lesions continued to spread linearly to the patient’s upper arm and forearm and manifested as the sporotrichoid infection type with painful swollen lesions at the site of inoculation and painful enlargement of the ipsilateral axillary lymph node.

Although nocardiosis is found worldwide, it is endemic to tropical and subtropical regions such as India, Africa, Southeast Asia, and Latin America.12 Nocardiosis most often is observed in individuals aged 20 to 40 years. It affects men more than women, and it commonly occurs in field laborers and cultivators whose occupations involve direct contact with the soil.13 Most lesions are found on the lower extremities, though localized nocardiosis infections can occur in other areas such as the neck, breasts, back, buttocks, and elbows.

Our patient initially was misdiagnosed, and treatment was delayed for several reasons. First, nocardiosis is not common in China, and most clinicians are unfamiliar with the disease. Second, the related lesions do not have specific features, and our patient had a complex clinical presentation that included mycetoma and sporotrichoid infection. Third, the characteristic grain of Nocardia species is small but that of N brasiliensis is even smaller (approximately 0.1–0.2 mm in diameter), which makes visualization difficult in both histopathologic and microbiologic examinations.14 The histopathologic examination results of our patient in the local hospital were nonspecific. Fourth, our patient did not initially go to the hospital but instead purchased some over-the-counter antibiotic ointments for external application because the lesions were painless. Moreover, microbiologic smear and culture examinations were not conducted in the local hospital before administering antituberculosis treatment to the patient. Instead, a polymerase chain reaction examination of skin lesion tissue for tubercle bacilli and atypical mycobacteria was negative. These findings imply that the traditional microbial smear and culture evaluations cannot be omitted. Furthermore, culture examinations should be conducted on multiple skin tissue and purulent fluid specimens to increase the likelihood of detection. These cultures should be monitored for at least 2 to 4 weeks because Nocardia is a slow-growing organism.10

The optimal antimicrobial treatment regimens for nocardiosis have not been firmly established.15 Trimethoprim-sulfamethoxazole is regarded as the first-line antimicrobial agent for treatment of nocardial infections. The optimal duration of antimicrobial therapy for nocardiosis also has not been determined, and the treatment regimen depends on the severity and extent of the infection as well as on the presence of infection-related complications. The main complication is bone involvement. Notable bony changes include periosteal thickening, osteoporosis, and osteolysis.

We considered the severity of skin lesions and bone marrow invasion in our patient and planned to treat him continually with oral trimethoprim-sulfamethoxazole according to the in vitro drug susceptibility test. The patient showed clinical improvement after 1 month of treatment, and he continued to improve after 6 months of treatment. To prevent recurrence, we found it necessary to treat the patient with a long-term antibiotic course over 6 to 12 months.16

Cutaneous nocardiosis remains a diagnostic challenge because of its nonspecific and diverse clinical and histopathological presentations. Diagnosis is further complicated by the inherent difficulty of cultivating and identifying the clinical isolate in the laboratory. A high degree of clinical suspicion followed by successful identification of the organism by a laboratory technologist will aid in the early diagnosis and treatment of the infection, ultimately reducing the risk for complications and morbidity.

References
  1. McNeil MM, Brown JM. The medically important aerobic actinomycetes: epidemiology and microbiology. Clin Microbiol Rev. 1994;7:357-417.
  2. Brown-Elliott BA, Brown JM, Conville PS, et al. Clinical and laboratory features of the Nocardia spp. based on current molecular taxonomy. Clin Microbiol Rev. 2006;19:259-282.
  3. Fatahi-Bafghi M. Nocardiosis from 1888 to 2017. Microb Pathog. 2018;114:369-384.
  4. Beaman BL, Burnside J, Edwards B, et al. Nocardial infections in the United States, 1972-1974. J Infect Dis. 1976;134:286-289.
  5. Lerner PI. Nocardiosis. Clin Infect Dis. 1996;22:891-903.
  6. Laurent FJ, Provost F, Boiron P. Rapid identification of clinically relevant Nocardia species to genus level by 16S rRNA gene PCR. J Clin Microbiol. 1999;37:99-102.
  7. Nguyen NM, Sink JR, Carter AJ, et al. Nocardiosis incognito: primary cutaneous nocardiosis with extension to myositis and pleural infection. JAAD Case Rep. 2018;4:33-35.
  8. Sharna NL, Mahajan VK, Agarwal S, et al. Nocardial mycetoma: diverse clinical presentations. Indian J Dermatol Venereol Leprol. 2008;74:635-640.
  9. Huang L, Chen X, Xu H, et al. Clinical features, identification, antimicrobial resistance patterns of Nocardia species in China: 2009-2017. Diagn Microbiol Infect Dis. 2019;94:165-172.
  10. Bonifaz A, Tirado-Sánchez A, Calderón L, et al. Mycetoma: experience of 482 cases in a single center in Mexico. PLoS Negl Trop Dis. 2014;8:E3102.
  11. Welsh O, Vero-Cabrera L, Salinas-Carmona MC. Mycetoma. Clin Dermatol. 2007;25:195-202.
  12. Nenoff P, van de Sande WWJ, Fahal AH, et al. Eumycetoma and actinomycetoma—an update on causative agents, epidemiology, pathogenesis, diagnostics and therapy. J Eur Acad Dermatol Venereol. 2015;29:1873-1883.
  13. Emmanuel P, Dumre SP, John S, et al. Mycetoma: a clinical dilemma in resource limited settings. Ann Clin Microbiol Antimicrob. 2018;17:35.
  14. Reis CMS, Reis-Filho EGM. Mycetomas: an epidemiological, etiological, clinical, laboratory and therapeutic review. An Bras Dermatol. 2018;93:8-18.
  15. Wilson JW. Nocardiosis: updates and clinical overview. Mayo Clin Proc. 2012;87:403-407.
  16. Welsh O, Vera-Cabrera L, Salinas-Carmona MC. Current treatment for Nocardia infections. Expert Opin Pharmacother. 2013;14:2387-2398.
References
  1. McNeil MM, Brown JM. The medically important aerobic actinomycetes: epidemiology and microbiology. Clin Microbiol Rev. 1994;7:357-417.
  2. Brown-Elliott BA, Brown JM, Conville PS, et al. Clinical and laboratory features of the Nocardia spp. based on current molecular taxonomy. Clin Microbiol Rev. 2006;19:259-282.
  3. Fatahi-Bafghi M. Nocardiosis from 1888 to 2017. Microb Pathog. 2018;114:369-384.
  4. Beaman BL, Burnside J, Edwards B, et al. Nocardial infections in the United States, 1972-1974. J Infect Dis. 1976;134:286-289.
  5. Lerner PI. Nocardiosis. Clin Infect Dis. 1996;22:891-903.
  6. Laurent FJ, Provost F, Boiron P. Rapid identification of clinically relevant Nocardia species to genus level by 16S rRNA gene PCR. J Clin Microbiol. 1999;37:99-102.
  7. Nguyen NM, Sink JR, Carter AJ, et al. Nocardiosis incognito: primary cutaneous nocardiosis with extension to myositis and pleural infection. JAAD Case Rep. 2018;4:33-35.
  8. Sharna NL, Mahajan VK, Agarwal S, et al. Nocardial mycetoma: diverse clinical presentations. Indian J Dermatol Venereol Leprol. 2008;74:635-640.
  9. Huang L, Chen X, Xu H, et al. Clinical features, identification, antimicrobial resistance patterns of Nocardia species in China: 2009-2017. Diagn Microbiol Infect Dis. 2019;94:165-172.
  10. Bonifaz A, Tirado-Sánchez A, Calderón L, et al. Mycetoma: experience of 482 cases in a single center in Mexico. PLoS Negl Trop Dis. 2014;8:E3102.
  11. Welsh O, Vero-Cabrera L, Salinas-Carmona MC. Mycetoma. Clin Dermatol. 2007;25:195-202.
  12. Nenoff P, van de Sande WWJ, Fahal AH, et al. Eumycetoma and actinomycetoma—an update on causative agents, epidemiology, pathogenesis, diagnostics and therapy. J Eur Acad Dermatol Venereol. 2015;29:1873-1883.
  13. Emmanuel P, Dumre SP, John S, et al. Mycetoma: a clinical dilemma in resource limited settings. Ann Clin Microbiol Antimicrob. 2018;17:35.
  14. Reis CMS, Reis-Filho EGM. Mycetomas: an epidemiological, etiological, clinical, laboratory and therapeutic review. An Bras Dermatol. 2018;93:8-18.
  15. Wilson JW. Nocardiosis: updates and clinical overview. Mayo Clin Proc. 2012;87:403-407.
  16. Welsh O, Vera-Cabrera L, Salinas-Carmona MC. Current treatment for Nocardia infections. Expert Opin Pharmacother. 2013;14:2387-2398.
Issue
Cutis - 111(3)
Issue
Cutis - 111(3)
Page Number
E22-E25
Page Number
E22-E25
Publications
Publications
Topics
Article Type
Display Headline
Progressive Primary Cutaneous Nocardiosis in an Immunocompetent Patient
Display Headline
Progressive Primary Cutaneous Nocardiosis in an Immunocompetent Patient
Sections
Inside the Article

Practice Points

  • Although unusual, cutaneous nocardiosis can present with both mycetoma and sporotrichoid infection, which should be treated based on pathogen identification and antibiotic sensitivity testing.
  • A high degree of clinical suspicion by clinicians followed by successful identification of the organism by a laboratory technologist will aid in the early diagnosis and treatment of the infection, ultimately reducing the risk for complications and morbidity.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Hair Repigmentation as a Melanoma Warning Sign

Article Type
Changed
Wed, 03/22/2023 - 17:18
Display Headline
Hair Repigmentation as a Melanoma Warning Sign

To the Editor:

An 85-year-old man with a history of hypertension and chronic kidney disease presented with a localized darkening patch of hair on the left parietal scalp that had progressed over the last 7 years (Figure 1A). He had no prior history of skin cancer. Physical examination revealed the remainder of the hair was gray. There was an irregularly pigmented plaque on the skin underlying the darkened hair measuring 5.0 cm in diameter that was confirmed to be melanoma (Figure 1B). He underwent a staged excision to remove the lesion. The surgical defect was closed via a 5.0×6.0-cm full-thickness skin graft. 

A, Localized darkening of hair on the left parietal scalp. B, An irregularly pigmented plaque measuring 5.0 cm in diameter was noted underlying the darkened hair.
FIGURE 1. A, Localized darkening of hair on the left parietal scalp. B, An irregularly pigmented plaque measuring 5.0 cm in diameter was noted underlying the darkened hair.

The initial biopsy showed melanoma in situ. However, the final pathology report following the excision revealed an invasive melanoma with a Breslow depth of 1.0 mm (Clark level IV; American Joint Committee on Cancer T1b).1 Histopathology showed pigment deposition with surrounding deep follicular extension of melanoma (Figure 2).

Staged excision of the pigmented area of the left parietal scalp revealed an emerging hair shaft with dark, variably chunky pigment deposition seen in association with surrounding melanoma
FIGURE 2. Staged excision of the pigmented area of the left parietal scalp revealed an emerging hair shaft with dark, variably chunky pigment deposition seen in association with surrounding melanoma (H&E, original magnification ×100).

The patient declined a sentinel lymph node biopsy and agreed to a genetic profile assessment.2 The results of the test identified the patient had a low probability of a positive sentinel lymph node and the lowest risk of melanoma recurrence within 5 years. The patient was clear of disease at 12-month follow-up.

Based on a PubMed search of articles indexed for MEDLINE using the terms hair repigmentation and melanoma, there have been 11 other reported cases of hair repigmentation associated with melanoma (Table).3-13 It initially was suspected that this rare phenomenon primarily existed in the female population, as the first 5 cases were reported solely in females,3-7 possibly due to the prevalence of androgenetic alopecia in males.11 However, 6 cases of repigmentation associated with melanoma were later reported in males8-13; our patient represents an additional reported case in a male. It is unknown if there is a higher prevalence of this phenomenon among males or females.

Summary of Reported Cases of Hair Repigmentation in Association With Melanoma

Most previously reported cases of repigmentation were associated with melanoma in situ, lentigo maligna type. Repigmentation also has been reported in malignant melanoma, as documented in our patient, as well as desmoplastic and amelanotic melanoma.5,6 In every case, the color of the repigmentation was darker than the rest of the patient’s hair; however, the repigmentation color can be different from the patient’s original hair color from their youth.4,5,11

The exact mechanism responsible for hair repigmentation in the setting of melanoma is unclear. It has been speculated from prior cases that repigmentation may be caused by paracrine stimulation from melanoma cells activating adjacent benign hair follicle melanocytes to produce melanin.7,14,15 This process likely is due to cytokines or growth factors, such as c-kit ligand.14,15 Several neural and immune networks and mediators activate the receptor tyrosine kinase KIT, which is thought to play a role in activating melanogenesis within the hair bulb.14 These signals also could originate from changes in the microenvironment instead of the melanoma cells themselves.6 Another possible mechanism is that repigmentation was caused by melanin-producing malignant melanocytes.4

Because this phenomenon typically occurs in older patients, the cause of repigmentation also could be related to chronic sun damage, which may result in upregulation of stem cell factor and α-melanocyte–stimulating hormone, as well as other molecules associated with melanogenesis, such as c-KIT receptor and tyrosinase.15,16 Upregulation of these molecules can lead to an increased number of melanocytes within the hair bulb. In addition, UVA and narrowband UVB have been recognized as major players in melanocyte stimulation. Phototherapy with UVA or narrowband UVB has been used for repigmentation in vitiligo patients.17

 

 

In cases without invasion of hair follicles by malignant cells, repigmentation more likely results from external signals stimulating benign bulbar melanocytes to produce melanin rather than melanoma cell growth extending into the hair bulb.6 In these cases, there is an increase in the number of hair bulbar melanocytes with a lack of malignant morphology in the hair bulb.8 If the signals are directly from melanoma cells in the hair bulb, it is unknown how the malignant cells upregulated melanogenesis in adjacent benign melanocytes or which specific signals required for normal pigmentation were involved in these repigmentation cases.6

Use of medications was ruled out as an underlying cause of the repigmentation in our patient. Drug-related repigmentation of the hair typically is observed in a diffuse generalized pattern. In our case, the repigmentation was localized to the area of the underlying dark patch, and the patient was not on any medications that could cause hair hyperpigmentation. Hyperpigmentation has been associated with acitretin, lenalidomide, corticosteroids, erlotinib, latanoprost, verapamil, tamoxifen, levodopa, thalidomide, PD-1 inhibitors, and tumor necrosis α inhibitors.18-30 Repigmentation also has been reported after local radiotherapy and herpes zoster infection.31,32

The underlying melanoma in our patient was removed by staged square excision. Excision was the treatment of choice for most similar reported cases. Radiotherapy was utilized in two different cases.3,4 In one case, radiotherapy was successfully used to treat melanoma in situ, lentigo maligna type; the patient’s hair grew back to its original color, which suggests that normal hair physiology was restored once melanoma cells were eliminated.3 One reported case demonstrated successful treatment of lentigo maligna type–melanoma with imiquimod cream 5% applied 6 times weekly for 9 months with a positive cosmetic result.9 The exact mechanism of imiquimod is not fully understood. Imiquimod induces cytokines to stimulate the production of IFN-α via activation of toll-like receptor 7.33 There was complete clearing of the lesion as well as the hair pigmentation,9 which suggests that the treatment also eliminated deeper cells influencing pigmentation. A case of malignant amelanotic melanoma was successfully treated with anti–PD-1 antibody pembrolizumab (2 mg/kg every 3 weeks), with no recurrence at 12 months. Pembrolizumab acts as an immune checkpoint inhibitor by binding to the PD-1 receptor and allowing the immune system to recognize and attack melanoma cells. After 5 doses of pembrolizumab, the patient was clear of disease and his hair color returned to gray.5

In 2022, melanoma was estimated to be the fifth most commonly diagnosed cancer among men and women in the United States.34 Early melanoma detection is a critical factor in achieving positive patient outcomes. Hair repigmentation is a potentially serious phenomenon that warrants a physician visit. Melanoma lesions under the hair may be overlooked because of limited visibility. Physicians must inspect spontaneous hair repigmentation with high suspicion and interpret the change as a possible indirect result of melanoma. Overall, it is important to increase public awareness of regular skin checks and melanoma warning signs.

References
  1. Gershenwald JE, Scolyer RA, Hess KR, et al. Melanoma staging: evidence‐based changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J Clin. 2017;67:472-492.
  2. Vetto JT, Hsueh EC, Gastman BR, et al. Guidance of sentinel lymph node biopsy decisions in patients with T1–T2 melanoma using gene expression profiling. Futur Oncol. 2019;15:1207-1217.
  3. Dummer R. Hair repigmentation in lentigo maligna. Lancet. 2001;357:598.
  4. Inzinger M, Massone C, Arzberger E, et al. Hair repigmentation in melanoma. Lancet. 2013;382:1224.
  5. Rahim RR, Husain A, Tobin DJ, et al. Desmoplastic melanoma presenting with localized hair repigmentation. Br J Dermatol. 2013;169:1371-1373.
  6. Tiger JB, Habeshian KA, Barton DT, et al. Repigmentation of hair associated with melanoma in situ of scalp. J Am Acad Dermatol. 2014;71:E144-E145.
  7. Amann VC, Dummer R. Localized hair repigmentation in a 91-year-old woman. JAMA Dermatol. 2016;152:81-82.
  8. Chan C, Magro CM, Pham AK, et al. Spontaneous hair repigmentation in an 80-year-old man: a case of melanoma-associated hair repigmentation and review of the literature. Am J Dermatopathol. 2019;41:671-674.
  9. Lackey AE, Glassman G, Grichnik J, et al. Repigmentation of gray hairs with lentigo maligna and response to topical imiquimod. JAAD Case Rep. 2019;5:1015-1017.
  10. Chew T, Pannell M, Jeeves A. Focal hair re-pigmentation associated with melanoma of the scalp. ANZ J Surg. 2019;90:1175-1176.
  11. López-Sánchez C, Collgros H. Hair repigmentation as a clue for scalp melanoma. Australas J Dermatol. 2019;61:179-180.
  12. Gessler J, Tejasvi T, Bresler SC. Repigmentation of scalp hair: a feature of early melanoma. Am J Med. 2023;136:E7-E8.
  13. Hasegawa T, Iino S, Kitakaze K, et al. Repigmentation of aging gray hair associated with unrecognized development and progression of amelanotic melanoma of the scalp: a physiological alert underlying hair rejuvenation. J Dermatol. 2021;48:E281-E283. doi:10.1111/1346-8138.15881
  14. D’Mello SAN, Finlay GJ, Baguley BC, et al. Signaling pathways in melanogenesis. Int J Mol Sci. 2016;17:1144.
  15. Hachiya A, Kobayashi A, Ohuchi A, et al. The paracrine role of stem cell factor/c-kit signaling in the activation of human melanocytes in ultraviolet-B-induced pigmentation. J Invest Dermatol. 2001;116:578-586.
  16. Slominski A, Wortsman J, Plonka PM, et al. Hair follicle pigmentation. J Invest Dermatol. 2005;124:13-21.
  17. Falabella R. Vitiligo and the melanocyte reservoir. Indian J Dermatol. 2009;54:313.
  18. Seckin D, Yildiz A. Repigmentation and curling of hair after acitretin therapy. Australas J Dermatol. 2009;50:214-216.
  19. Dasanu CA, Mitsis D, Alexandrescu DT. Hair repigmentation associated with the use of lenalidomide: graying may not be an irreversible process! J Oncol Pharm Pract. 2013;19:165-169.
  20. Sebaratnam DF, Rodríguez Bandera AI, Lowe PM. Hair repigmentation with anti–PD-1 and anti–PD-L1 immunotherapy: a novel hypothesis. JAMA Dermatol. 2018;154:112-113. doi:10.1001/jamadermatol.2017.4420
  21. Tintle SJ, Dabade TS, Kalish RA, et al. Repigmentation of hair following adalimumab therapy. Dermatol Online J. 2015;21:13030/qt6fn0t1xz.
  22. Penzi LR, Manatis-Lornell A, Saavedra A, et al. Hair repigmentation associated with the use of brentuximab. JAAD Case Rep. 2017;3:563-565.
  23. Khaled A, Trojjets S, Zeglaoui F, et al. Repigmentation of the white hair after systemic corticosteroids for bullous pemphigoid. J Eur Acad Dermatology Venereol. 2008;22:1018-1020.
  24. Cheng YP, Chen HJ, Chiu HC. Erlotinib-induced hair repigmentation. Int J Dermatol. 2014;53:E55-E57.
  25. Bellandi S, Amato L, Cipollini EM, et al. Repigmentation of hair after latanoprost therapy. J Eur Acad Dermatology Venereol. 2011;25:1485-1487.
  26. Read GM. Verapamil and hair colour change. Lancet. 1991;338:1520.
  27. Hampson JP, Donnelly A, Lewis‐Jones MS, et al. Tamoxifen‐induced hair colour change. Br J Dermatol. 1995;132:483-484.
  28. Reynolds NJ, Crossley J, Ferguson I, et al. Darkening of white hair in Parkinson’s disease. Clin Exp Dermatol. 1989;14:317-318.
  29. Lovering S, Miao W, Bailie T, et al. Hair repigmentation associated with thalidomide use for the treatment of multiple myeloma. BMJ Case Rep. 2016;2016:bcr2016215521.
  30. Rivera N, Boada A, Bielsa MI, et al. Hair repigmentation during immunotherapy treatment with an anti–programmed cell death 1 and anti–programmed cell death ligand 1 agent for lung cancer. JAMA Dermatol. 2017;153:1162-1165.
  31. Prasad S, Dougheney N, Hong A. Scalp hair repigmentation in the penumbral region of radiotherapy–a case series. Int J Radiol Radiat Ther. 2020;7:151-157.
  32. Adiga GU, Rehman KL, Wiernik PH. Permanent localized hair repigmentation following herpes zoster infection. Arch Dermatol. 2010;146:569-570.
  33. Hanna E, Abadi R, Abbas O. Imiquimod in dermatology: an overview. Int J Dermatol. 2016;55:831-844.
  34. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7-33.
Article PDF
Author and Disclosure Information

From the University of Arkansas for Medical Sciences, Little Rock. Ms. Ly is from the College of Medicine, Dr. Rollins is from the Department of Pathology, and Drs. Mohr and Jennings are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Sophia Ly, BA, 4301 W Markham St, Slot 576, Little Rock, AR 72205 (sly@uams.edu).

Issue
Cutis - 111(3)
Publications
Topics
Page Number
E18-E21
Sections
Author and Disclosure Information

From the University of Arkansas for Medical Sciences, Little Rock. Ms. Ly is from the College of Medicine, Dr. Rollins is from the Department of Pathology, and Drs. Mohr and Jennings are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Sophia Ly, BA, 4301 W Markham St, Slot 576, Little Rock, AR 72205 (sly@uams.edu).

Author and Disclosure Information

From the University of Arkansas for Medical Sciences, Little Rock. Ms. Ly is from the College of Medicine, Dr. Rollins is from the Department of Pathology, and Drs. Mohr and Jennings are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Sophia Ly, BA, 4301 W Markham St, Slot 576, Little Rock, AR 72205 (sly@uams.edu).

Article PDF
Article PDF

To the Editor:

An 85-year-old man with a history of hypertension and chronic kidney disease presented with a localized darkening patch of hair on the left parietal scalp that had progressed over the last 7 years (Figure 1A). He had no prior history of skin cancer. Physical examination revealed the remainder of the hair was gray. There was an irregularly pigmented plaque on the skin underlying the darkened hair measuring 5.0 cm in diameter that was confirmed to be melanoma (Figure 1B). He underwent a staged excision to remove the lesion. The surgical defect was closed via a 5.0×6.0-cm full-thickness skin graft. 

A, Localized darkening of hair on the left parietal scalp. B, An irregularly pigmented plaque measuring 5.0 cm in diameter was noted underlying the darkened hair.
FIGURE 1. A, Localized darkening of hair on the left parietal scalp. B, An irregularly pigmented plaque measuring 5.0 cm in diameter was noted underlying the darkened hair.

The initial biopsy showed melanoma in situ. However, the final pathology report following the excision revealed an invasive melanoma with a Breslow depth of 1.0 mm (Clark level IV; American Joint Committee on Cancer T1b).1 Histopathology showed pigment deposition with surrounding deep follicular extension of melanoma (Figure 2).

Staged excision of the pigmented area of the left parietal scalp revealed an emerging hair shaft with dark, variably chunky pigment deposition seen in association with surrounding melanoma
FIGURE 2. Staged excision of the pigmented area of the left parietal scalp revealed an emerging hair shaft with dark, variably chunky pigment deposition seen in association with surrounding melanoma (H&E, original magnification ×100).

The patient declined a sentinel lymph node biopsy and agreed to a genetic profile assessment.2 The results of the test identified the patient had a low probability of a positive sentinel lymph node and the lowest risk of melanoma recurrence within 5 years. The patient was clear of disease at 12-month follow-up.

Based on a PubMed search of articles indexed for MEDLINE using the terms hair repigmentation and melanoma, there have been 11 other reported cases of hair repigmentation associated with melanoma (Table).3-13 It initially was suspected that this rare phenomenon primarily existed in the female population, as the first 5 cases were reported solely in females,3-7 possibly due to the prevalence of androgenetic alopecia in males.11 However, 6 cases of repigmentation associated with melanoma were later reported in males8-13; our patient represents an additional reported case in a male. It is unknown if there is a higher prevalence of this phenomenon among males or females.

Summary of Reported Cases of Hair Repigmentation in Association With Melanoma

Most previously reported cases of repigmentation were associated with melanoma in situ, lentigo maligna type. Repigmentation also has been reported in malignant melanoma, as documented in our patient, as well as desmoplastic and amelanotic melanoma.5,6 In every case, the color of the repigmentation was darker than the rest of the patient’s hair; however, the repigmentation color can be different from the patient’s original hair color from their youth.4,5,11

The exact mechanism responsible for hair repigmentation in the setting of melanoma is unclear. It has been speculated from prior cases that repigmentation may be caused by paracrine stimulation from melanoma cells activating adjacent benign hair follicle melanocytes to produce melanin.7,14,15 This process likely is due to cytokines or growth factors, such as c-kit ligand.14,15 Several neural and immune networks and mediators activate the receptor tyrosine kinase KIT, which is thought to play a role in activating melanogenesis within the hair bulb.14 These signals also could originate from changes in the microenvironment instead of the melanoma cells themselves.6 Another possible mechanism is that repigmentation was caused by melanin-producing malignant melanocytes.4

Because this phenomenon typically occurs in older patients, the cause of repigmentation also could be related to chronic sun damage, which may result in upregulation of stem cell factor and α-melanocyte–stimulating hormone, as well as other molecules associated with melanogenesis, such as c-KIT receptor and tyrosinase.15,16 Upregulation of these molecules can lead to an increased number of melanocytes within the hair bulb. In addition, UVA and narrowband UVB have been recognized as major players in melanocyte stimulation. Phototherapy with UVA or narrowband UVB has been used for repigmentation in vitiligo patients.17

 

 

In cases without invasion of hair follicles by malignant cells, repigmentation more likely results from external signals stimulating benign bulbar melanocytes to produce melanin rather than melanoma cell growth extending into the hair bulb.6 In these cases, there is an increase in the number of hair bulbar melanocytes with a lack of malignant morphology in the hair bulb.8 If the signals are directly from melanoma cells in the hair bulb, it is unknown how the malignant cells upregulated melanogenesis in adjacent benign melanocytes or which specific signals required for normal pigmentation were involved in these repigmentation cases.6

Use of medications was ruled out as an underlying cause of the repigmentation in our patient. Drug-related repigmentation of the hair typically is observed in a diffuse generalized pattern. In our case, the repigmentation was localized to the area of the underlying dark patch, and the patient was not on any medications that could cause hair hyperpigmentation. Hyperpigmentation has been associated with acitretin, lenalidomide, corticosteroids, erlotinib, latanoprost, verapamil, tamoxifen, levodopa, thalidomide, PD-1 inhibitors, and tumor necrosis α inhibitors.18-30 Repigmentation also has been reported after local radiotherapy and herpes zoster infection.31,32

The underlying melanoma in our patient was removed by staged square excision. Excision was the treatment of choice for most similar reported cases. Radiotherapy was utilized in two different cases.3,4 In one case, radiotherapy was successfully used to treat melanoma in situ, lentigo maligna type; the patient’s hair grew back to its original color, which suggests that normal hair physiology was restored once melanoma cells were eliminated.3 One reported case demonstrated successful treatment of lentigo maligna type–melanoma with imiquimod cream 5% applied 6 times weekly for 9 months with a positive cosmetic result.9 The exact mechanism of imiquimod is not fully understood. Imiquimod induces cytokines to stimulate the production of IFN-α via activation of toll-like receptor 7.33 There was complete clearing of the lesion as well as the hair pigmentation,9 which suggests that the treatment also eliminated deeper cells influencing pigmentation. A case of malignant amelanotic melanoma was successfully treated with anti–PD-1 antibody pembrolizumab (2 mg/kg every 3 weeks), with no recurrence at 12 months. Pembrolizumab acts as an immune checkpoint inhibitor by binding to the PD-1 receptor and allowing the immune system to recognize and attack melanoma cells. After 5 doses of pembrolizumab, the patient was clear of disease and his hair color returned to gray.5

In 2022, melanoma was estimated to be the fifth most commonly diagnosed cancer among men and women in the United States.34 Early melanoma detection is a critical factor in achieving positive patient outcomes. Hair repigmentation is a potentially serious phenomenon that warrants a physician visit. Melanoma lesions under the hair may be overlooked because of limited visibility. Physicians must inspect spontaneous hair repigmentation with high suspicion and interpret the change as a possible indirect result of melanoma. Overall, it is important to increase public awareness of regular skin checks and melanoma warning signs.

To the Editor:

An 85-year-old man with a history of hypertension and chronic kidney disease presented with a localized darkening patch of hair on the left parietal scalp that had progressed over the last 7 years (Figure 1A). He had no prior history of skin cancer. Physical examination revealed the remainder of the hair was gray. There was an irregularly pigmented plaque on the skin underlying the darkened hair measuring 5.0 cm in diameter that was confirmed to be melanoma (Figure 1B). He underwent a staged excision to remove the lesion. The surgical defect was closed via a 5.0×6.0-cm full-thickness skin graft. 

A, Localized darkening of hair on the left parietal scalp. B, An irregularly pigmented plaque measuring 5.0 cm in diameter was noted underlying the darkened hair.
FIGURE 1. A, Localized darkening of hair on the left parietal scalp. B, An irregularly pigmented plaque measuring 5.0 cm in diameter was noted underlying the darkened hair.

The initial biopsy showed melanoma in situ. However, the final pathology report following the excision revealed an invasive melanoma with a Breslow depth of 1.0 mm (Clark level IV; American Joint Committee on Cancer T1b).1 Histopathology showed pigment deposition with surrounding deep follicular extension of melanoma (Figure 2).

Staged excision of the pigmented area of the left parietal scalp revealed an emerging hair shaft with dark, variably chunky pigment deposition seen in association with surrounding melanoma
FIGURE 2. Staged excision of the pigmented area of the left parietal scalp revealed an emerging hair shaft with dark, variably chunky pigment deposition seen in association with surrounding melanoma (H&E, original magnification ×100).

The patient declined a sentinel lymph node biopsy and agreed to a genetic profile assessment.2 The results of the test identified the patient had a low probability of a positive sentinel lymph node and the lowest risk of melanoma recurrence within 5 years. The patient was clear of disease at 12-month follow-up.

Based on a PubMed search of articles indexed for MEDLINE using the terms hair repigmentation and melanoma, there have been 11 other reported cases of hair repigmentation associated with melanoma (Table).3-13 It initially was suspected that this rare phenomenon primarily existed in the female population, as the first 5 cases were reported solely in females,3-7 possibly due to the prevalence of androgenetic alopecia in males.11 However, 6 cases of repigmentation associated with melanoma were later reported in males8-13; our patient represents an additional reported case in a male. It is unknown if there is a higher prevalence of this phenomenon among males or females.

Summary of Reported Cases of Hair Repigmentation in Association With Melanoma

Most previously reported cases of repigmentation were associated with melanoma in situ, lentigo maligna type. Repigmentation also has been reported in malignant melanoma, as documented in our patient, as well as desmoplastic and amelanotic melanoma.5,6 In every case, the color of the repigmentation was darker than the rest of the patient’s hair; however, the repigmentation color can be different from the patient’s original hair color from their youth.4,5,11

The exact mechanism responsible for hair repigmentation in the setting of melanoma is unclear. It has been speculated from prior cases that repigmentation may be caused by paracrine stimulation from melanoma cells activating adjacent benign hair follicle melanocytes to produce melanin.7,14,15 This process likely is due to cytokines or growth factors, such as c-kit ligand.14,15 Several neural and immune networks and mediators activate the receptor tyrosine kinase KIT, which is thought to play a role in activating melanogenesis within the hair bulb.14 These signals also could originate from changes in the microenvironment instead of the melanoma cells themselves.6 Another possible mechanism is that repigmentation was caused by melanin-producing malignant melanocytes.4

Because this phenomenon typically occurs in older patients, the cause of repigmentation also could be related to chronic sun damage, which may result in upregulation of stem cell factor and α-melanocyte–stimulating hormone, as well as other molecules associated with melanogenesis, such as c-KIT receptor and tyrosinase.15,16 Upregulation of these molecules can lead to an increased number of melanocytes within the hair bulb. In addition, UVA and narrowband UVB have been recognized as major players in melanocyte stimulation. Phototherapy with UVA or narrowband UVB has been used for repigmentation in vitiligo patients.17

 

 

In cases without invasion of hair follicles by malignant cells, repigmentation more likely results from external signals stimulating benign bulbar melanocytes to produce melanin rather than melanoma cell growth extending into the hair bulb.6 In these cases, there is an increase in the number of hair bulbar melanocytes with a lack of malignant morphology in the hair bulb.8 If the signals are directly from melanoma cells in the hair bulb, it is unknown how the malignant cells upregulated melanogenesis in adjacent benign melanocytes or which specific signals required for normal pigmentation were involved in these repigmentation cases.6

Use of medications was ruled out as an underlying cause of the repigmentation in our patient. Drug-related repigmentation of the hair typically is observed in a diffuse generalized pattern. In our case, the repigmentation was localized to the area of the underlying dark patch, and the patient was not on any medications that could cause hair hyperpigmentation. Hyperpigmentation has been associated with acitretin, lenalidomide, corticosteroids, erlotinib, latanoprost, verapamil, tamoxifen, levodopa, thalidomide, PD-1 inhibitors, and tumor necrosis α inhibitors.18-30 Repigmentation also has been reported after local radiotherapy and herpes zoster infection.31,32

The underlying melanoma in our patient was removed by staged square excision. Excision was the treatment of choice for most similar reported cases. Radiotherapy was utilized in two different cases.3,4 In one case, radiotherapy was successfully used to treat melanoma in situ, lentigo maligna type; the patient’s hair grew back to its original color, which suggests that normal hair physiology was restored once melanoma cells were eliminated.3 One reported case demonstrated successful treatment of lentigo maligna type–melanoma with imiquimod cream 5% applied 6 times weekly for 9 months with a positive cosmetic result.9 The exact mechanism of imiquimod is not fully understood. Imiquimod induces cytokines to stimulate the production of IFN-α via activation of toll-like receptor 7.33 There was complete clearing of the lesion as well as the hair pigmentation,9 which suggests that the treatment also eliminated deeper cells influencing pigmentation. A case of malignant amelanotic melanoma was successfully treated with anti–PD-1 antibody pembrolizumab (2 mg/kg every 3 weeks), with no recurrence at 12 months. Pembrolizumab acts as an immune checkpoint inhibitor by binding to the PD-1 receptor and allowing the immune system to recognize and attack melanoma cells. After 5 doses of pembrolizumab, the patient was clear of disease and his hair color returned to gray.5

In 2022, melanoma was estimated to be the fifth most commonly diagnosed cancer among men and women in the United States.34 Early melanoma detection is a critical factor in achieving positive patient outcomes. Hair repigmentation is a potentially serious phenomenon that warrants a physician visit. Melanoma lesions under the hair may be overlooked because of limited visibility. Physicians must inspect spontaneous hair repigmentation with high suspicion and interpret the change as a possible indirect result of melanoma. Overall, it is important to increase public awareness of regular skin checks and melanoma warning signs.

References
  1. Gershenwald JE, Scolyer RA, Hess KR, et al. Melanoma staging: evidence‐based changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J Clin. 2017;67:472-492.
  2. Vetto JT, Hsueh EC, Gastman BR, et al. Guidance of sentinel lymph node biopsy decisions in patients with T1–T2 melanoma using gene expression profiling. Futur Oncol. 2019;15:1207-1217.
  3. Dummer R. Hair repigmentation in lentigo maligna. Lancet. 2001;357:598.
  4. Inzinger M, Massone C, Arzberger E, et al. Hair repigmentation in melanoma. Lancet. 2013;382:1224.
  5. Rahim RR, Husain A, Tobin DJ, et al. Desmoplastic melanoma presenting with localized hair repigmentation. Br J Dermatol. 2013;169:1371-1373.
  6. Tiger JB, Habeshian KA, Barton DT, et al. Repigmentation of hair associated with melanoma in situ of scalp. J Am Acad Dermatol. 2014;71:E144-E145.
  7. Amann VC, Dummer R. Localized hair repigmentation in a 91-year-old woman. JAMA Dermatol. 2016;152:81-82.
  8. Chan C, Magro CM, Pham AK, et al. Spontaneous hair repigmentation in an 80-year-old man: a case of melanoma-associated hair repigmentation and review of the literature. Am J Dermatopathol. 2019;41:671-674.
  9. Lackey AE, Glassman G, Grichnik J, et al. Repigmentation of gray hairs with lentigo maligna and response to topical imiquimod. JAAD Case Rep. 2019;5:1015-1017.
  10. Chew T, Pannell M, Jeeves A. Focal hair re-pigmentation associated with melanoma of the scalp. ANZ J Surg. 2019;90:1175-1176.
  11. López-Sánchez C, Collgros H. Hair repigmentation as a clue for scalp melanoma. Australas J Dermatol. 2019;61:179-180.
  12. Gessler J, Tejasvi T, Bresler SC. Repigmentation of scalp hair: a feature of early melanoma. Am J Med. 2023;136:E7-E8.
  13. Hasegawa T, Iino S, Kitakaze K, et al. Repigmentation of aging gray hair associated with unrecognized development and progression of amelanotic melanoma of the scalp: a physiological alert underlying hair rejuvenation. J Dermatol. 2021;48:E281-E283. doi:10.1111/1346-8138.15881
  14. D’Mello SAN, Finlay GJ, Baguley BC, et al. Signaling pathways in melanogenesis. Int J Mol Sci. 2016;17:1144.
  15. Hachiya A, Kobayashi A, Ohuchi A, et al. The paracrine role of stem cell factor/c-kit signaling in the activation of human melanocytes in ultraviolet-B-induced pigmentation. J Invest Dermatol. 2001;116:578-586.
  16. Slominski A, Wortsman J, Plonka PM, et al. Hair follicle pigmentation. J Invest Dermatol. 2005;124:13-21.
  17. Falabella R. Vitiligo and the melanocyte reservoir. Indian J Dermatol. 2009;54:313.
  18. Seckin D, Yildiz A. Repigmentation and curling of hair after acitretin therapy. Australas J Dermatol. 2009;50:214-216.
  19. Dasanu CA, Mitsis D, Alexandrescu DT. Hair repigmentation associated with the use of lenalidomide: graying may not be an irreversible process! J Oncol Pharm Pract. 2013;19:165-169.
  20. Sebaratnam DF, Rodríguez Bandera AI, Lowe PM. Hair repigmentation with anti–PD-1 and anti–PD-L1 immunotherapy: a novel hypothesis. JAMA Dermatol. 2018;154:112-113. doi:10.1001/jamadermatol.2017.4420
  21. Tintle SJ, Dabade TS, Kalish RA, et al. Repigmentation of hair following adalimumab therapy. Dermatol Online J. 2015;21:13030/qt6fn0t1xz.
  22. Penzi LR, Manatis-Lornell A, Saavedra A, et al. Hair repigmentation associated with the use of brentuximab. JAAD Case Rep. 2017;3:563-565.
  23. Khaled A, Trojjets S, Zeglaoui F, et al. Repigmentation of the white hair after systemic corticosteroids for bullous pemphigoid. J Eur Acad Dermatology Venereol. 2008;22:1018-1020.
  24. Cheng YP, Chen HJ, Chiu HC. Erlotinib-induced hair repigmentation. Int J Dermatol. 2014;53:E55-E57.
  25. Bellandi S, Amato L, Cipollini EM, et al. Repigmentation of hair after latanoprost therapy. J Eur Acad Dermatology Venereol. 2011;25:1485-1487.
  26. Read GM. Verapamil and hair colour change. Lancet. 1991;338:1520.
  27. Hampson JP, Donnelly A, Lewis‐Jones MS, et al. Tamoxifen‐induced hair colour change. Br J Dermatol. 1995;132:483-484.
  28. Reynolds NJ, Crossley J, Ferguson I, et al. Darkening of white hair in Parkinson’s disease. Clin Exp Dermatol. 1989;14:317-318.
  29. Lovering S, Miao W, Bailie T, et al. Hair repigmentation associated with thalidomide use for the treatment of multiple myeloma. BMJ Case Rep. 2016;2016:bcr2016215521.
  30. Rivera N, Boada A, Bielsa MI, et al. Hair repigmentation during immunotherapy treatment with an anti–programmed cell death 1 and anti–programmed cell death ligand 1 agent for lung cancer. JAMA Dermatol. 2017;153:1162-1165.
  31. Prasad S, Dougheney N, Hong A. Scalp hair repigmentation in the penumbral region of radiotherapy–a case series. Int J Radiol Radiat Ther. 2020;7:151-157.
  32. Adiga GU, Rehman KL, Wiernik PH. Permanent localized hair repigmentation following herpes zoster infection. Arch Dermatol. 2010;146:569-570.
  33. Hanna E, Abadi R, Abbas O. Imiquimod in dermatology: an overview. Int J Dermatol. 2016;55:831-844.
  34. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7-33.
References
  1. Gershenwald JE, Scolyer RA, Hess KR, et al. Melanoma staging: evidence‐based changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J Clin. 2017;67:472-492.
  2. Vetto JT, Hsueh EC, Gastman BR, et al. Guidance of sentinel lymph node biopsy decisions in patients with T1–T2 melanoma using gene expression profiling. Futur Oncol. 2019;15:1207-1217.
  3. Dummer R. Hair repigmentation in lentigo maligna. Lancet. 2001;357:598.
  4. Inzinger M, Massone C, Arzberger E, et al. Hair repigmentation in melanoma. Lancet. 2013;382:1224.
  5. Rahim RR, Husain A, Tobin DJ, et al. Desmoplastic melanoma presenting with localized hair repigmentation. Br J Dermatol. 2013;169:1371-1373.
  6. Tiger JB, Habeshian KA, Barton DT, et al. Repigmentation of hair associated with melanoma in situ of scalp. J Am Acad Dermatol. 2014;71:E144-E145.
  7. Amann VC, Dummer R. Localized hair repigmentation in a 91-year-old woman. JAMA Dermatol. 2016;152:81-82.
  8. Chan C, Magro CM, Pham AK, et al. Spontaneous hair repigmentation in an 80-year-old man: a case of melanoma-associated hair repigmentation and review of the literature. Am J Dermatopathol. 2019;41:671-674.
  9. Lackey AE, Glassman G, Grichnik J, et al. Repigmentation of gray hairs with lentigo maligna and response to topical imiquimod. JAAD Case Rep. 2019;5:1015-1017.
  10. Chew T, Pannell M, Jeeves A. Focal hair re-pigmentation associated with melanoma of the scalp. ANZ J Surg. 2019;90:1175-1176.
  11. López-Sánchez C, Collgros H. Hair repigmentation as a clue for scalp melanoma. Australas J Dermatol. 2019;61:179-180.
  12. Gessler J, Tejasvi T, Bresler SC. Repigmentation of scalp hair: a feature of early melanoma. Am J Med. 2023;136:E7-E8.
  13. Hasegawa T, Iino S, Kitakaze K, et al. Repigmentation of aging gray hair associated with unrecognized development and progression of amelanotic melanoma of the scalp: a physiological alert underlying hair rejuvenation. J Dermatol. 2021;48:E281-E283. doi:10.1111/1346-8138.15881
  14. D’Mello SAN, Finlay GJ, Baguley BC, et al. Signaling pathways in melanogenesis. Int J Mol Sci. 2016;17:1144.
  15. Hachiya A, Kobayashi A, Ohuchi A, et al. The paracrine role of stem cell factor/c-kit signaling in the activation of human melanocytes in ultraviolet-B-induced pigmentation. J Invest Dermatol. 2001;116:578-586.
  16. Slominski A, Wortsman J, Plonka PM, et al. Hair follicle pigmentation. J Invest Dermatol. 2005;124:13-21.
  17. Falabella R. Vitiligo and the melanocyte reservoir. Indian J Dermatol. 2009;54:313.
  18. Seckin D, Yildiz A. Repigmentation and curling of hair after acitretin therapy. Australas J Dermatol. 2009;50:214-216.
  19. Dasanu CA, Mitsis D, Alexandrescu DT. Hair repigmentation associated with the use of lenalidomide: graying may not be an irreversible process! J Oncol Pharm Pract. 2013;19:165-169.
  20. Sebaratnam DF, Rodríguez Bandera AI, Lowe PM. Hair repigmentation with anti–PD-1 and anti–PD-L1 immunotherapy: a novel hypothesis. JAMA Dermatol. 2018;154:112-113. doi:10.1001/jamadermatol.2017.4420
  21. Tintle SJ, Dabade TS, Kalish RA, et al. Repigmentation of hair following adalimumab therapy. Dermatol Online J. 2015;21:13030/qt6fn0t1xz.
  22. Penzi LR, Manatis-Lornell A, Saavedra A, et al. Hair repigmentation associated with the use of brentuximab. JAAD Case Rep. 2017;3:563-565.
  23. Khaled A, Trojjets S, Zeglaoui F, et al. Repigmentation of the white hair after systemic corticosteroids for bullous pemphigoid. J Eur Acad Dermatology Venereol. 2008;22:1018-1020.
  24. Cheng YP, Chen HJ, Chiu HC. Erlotinib-induced hair repigmentation. Int J Dermatol. 2014;53:E55-E57.
  25. Bellandi S, Amato L, Cipollini EM, et al. Repigmentation of hair after latanoprost therapy. J Eur Acad Dermatology Venereol. 2011;25:1485-1487.
  26. Read GM. Verapamil and hair colour change. Lancet. 1991;338:1520.
  27. Hampson JP, Donnelly A, Lewis‐Jones MS, et al. Tamoxifen‐induced hair colour change. Br J Dermatol. 1995;132:483-484.
  28. Reynolds NJ, Crossley J, Ferguson I, et al. Darkening of white hair in Parkinson’s disease. Clin Exp Dermatol. 1989;14:317-318.
  29. Lovering S, Miao W, Bailie T, et al. Hair repigmentation associated with thalidomide use for the treatment of multiple myeloma. BMJ Case Rep. 2016;2016:bcr2016215521.
  30. Rivera N, Boada A, Bielsa MI, et al. Hair repigmentation during immunotherapy treatment with an anti–programmed cell death 1 and anti–programmed cell death ligand 1 agent for lung cancer. JAMA Dermatol. 2017;153:1162-1165.
  31. Prasad S, Dougheney N, Hong A. Scalp hair repigmentation in the penumbral region of radiotherapy–a case series. Int J Radiol Radiat Ther. 2020;7:151-157.
  32. Adiga GU, Rehman KL, Wiernik PH. Permanent localized hair repigmentation following herpes zoster infection. Arch Dermatol. 2010;146:569-570.
  33. Hanna E, Abadi R, Abbas O. Imiquimod in dermatology: an overview. Int J Dermatol. 2016;55:831-844.
  34. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7-33.
Issue
Cutis - 111(3)
Issue
Cutis - 111(3)
Page Number
E18-E21
Page Number
E18-E21
Publications
Publications
Topics
Article Type
Display Headline
Hair Repigmentation as a Melanoma Warning Sign
Display Headline
Hair Repigmentation as a Melanoma Warning Sign
Sections
Inside the Article

Practice Points

  • Localized repigmentation of the hair is a rare phenomenon that may indicate underlying melanoma.
  • Careful clinicopathologic correlation is necessary to appropriately diagnose and manage this unusual presentation of melanoma.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Generalized Essential Telangiectasia Treated With Pulsed Dye Laser

Article Type
Changed
Thu, 03/23/2023 - 07:37
Display Headline
Generalized Essential Telangiectasia Treated With Pulsed Dye Laser

To the Editor:

Generalized essential telangiectasia (GET) is a rare, benign, and progressive primary cutaneous disease manifesting as telangiectases of the skin without systemic symptoms. It is unique in that it has widespread distribution on the body. Generalized essential telangiectasia more commonly affects women, usually in the fourth decade of life. The telangiectases most frequently appear on the legs, advancing over time to involve the trunk and arms and presenting in several patterns, including diffuse, macular, plaquelike, discrete, or confluent. Although GET typically is asymptomatic, numbness, tingling, and burning of the involved areas have been reported.1 Treatment modalities for GET vary, though pulsed dye laser (PDL) therapy is most common. We report the case of a 40-year-old woman with a 5-year history of GET who was treated successfully with PDL.

A 40-year-old woman presented to our dermatology clinic with progressive prominence of blood vessels involving the dorsal aspects of the feet of 5 years’ duration. The prominent vessels had spread to involve the legs (Figure 1), buttocks, lower abdomen, forearms, and medial upper arms. The patient denied any personal history of bleeding disorders or family history of inherited conditions associated with visceral vascular malformations, such as hereditary hemorrhagic telangiectasia. Notably, magnetic resonance imaging of the liver approximately 3 weeks prior to initiating treatment with PDL demonstrated multiple hepatic lesions consistent with hemangiomas. The patient reported an occasional tingling sensation in the feet. She was otherwise asymptomatic but did report psychological distress associated with the skin changes.

Erythematous to purpuric telangiectases on the lower legs of a 40-year-old woman with generalized essential telangiectasia prior to starting pulsed dye laser therapy
FIGURE 1. Erythematous to purpuric telangiectases on the lower legs of a 40-year-old woman with generalized essential telangiectasia prior to starting pulsed dye laser therapy.

Punch biopsies from the right lower leg and right buttock demonstrated increased vascularity of the dermis, a mild superficial perivascular lymphocytic infiltrate, and mild edema of the upper dermis without evidence of vasculitis. Autoimmune and coagulopathy workups were negative. The clinical and pathological findings were most consistent with GET.

Over the next 2.5 years, the patient underwent treatment with doxycycline and a series of 16 treatments with PDL (fluence, 6–12 J/cm2; pulse width, 10 milliseconds) with a positive cosmetic response. Considerable improvement in the lower legs was noted after 2 years of treatment with PDL (Figure 2).

The patient’s condition improved considerably, albeit transiently, after 2 years of pulsed dye laser therapy (fluence, 6–12 J/cm2 ; pulse width, 10 milliseconds).
FIGURE 2. The patient’s condition improved considerably, albeit transiently, after 2 years of pulsed dye laser therapy (fluence, 6–12 J/cm2 ; pulse width, 10 milliseconds).

Recurrence of GET was noted between PDL treatments, which led to progression of the disease process; all treated sites showed slow recurrence of lesions within several months after treatment. After 2 years, doxycycline was discontinued because of a perceived lack of continued benefit and the patient’s desire for alternative therapy. She was started on a 3-month trial of supplementation with ascorbic acid and rutin (or rutoside, a bioflavinoid), without noticeable improvement.

The diffuse distribution of dramatic telangiectases in GET makes treatment difficult. Standard treatments are not well established or studied due to the rarity of the condition. A review of PubMed articles indexed for MEDLINE using the terms treatment and generalized essential telangiectasias demonstrated several attempted treatment modalities for GET with varying success. In 4 cases in which PDL was used,2-5 a positive cosmetic response was noted, similar to what was seen in our patient. In 1 of the 4 cases, conservative management with ascorbic acid and compression stockings was unsuccessful; however, 6-mercaptopurine, used to treat that patient’s ulcerative colitis, incidentally resulted in resolution of GET.2 In 2 cases, response was maintained at 1.5-year follow-up.3,5 Two cases noted successful treatment with acyclovir,6,7 and 2 more demonstrated successful treatment with systemic ketoconazole.6,8 Some improvement was reported with oral doxycycline or tetracycline in 2 cases.9,10 Sclerotherapy improved the cosmetic appearance of telangiectases in one patient but was unsustainable because of the pain associated with the procedure.11 Nd:YAG laser therapy was effective in one case12; however, the patient experienced relapse at 6-month follow-up—similar to what we observed in our patient. Three patients treated with intense pulsed light therapy experienced results that were maintained at 2-year follow-up.13

Generalized essential telangiectasia generally is considered a skin-limited disease without systemic manifestations, but 2 reports11,14 described its association with gastric antral vascular ectasia—known as watermelon stomach. Hepatic hemangiomas are the most common benign liver lesions; however, the findings on magnetic resonance imaging in our patient, in combination with the 2 reported cases of watermelon stomach, suggest that the vascular changes of GET might extend below the skin.

Of the cases we reviewed, our patient had the longest reported duration of PDL treatment and follow-up for GET in which a successful, albeit transient, response was demonstrated. Our review of the literature revealed other reports of success with PDL and intense pulsed light therapy; results were maintained in some patients, while disease relapsed in others. Further studies are needed to understand why results are maintained in some but not all patients.

Although the cost of PDL as a cosmetic procedure must be taken into consideration when planning treatment of GET, we conclude that it is a safe option that can be effective until other treatment options are established to control the disease.

References
  1. McGrae JD Jr, Winkelmann RK. Generalized essential telangiectasia: report of a clinical and histochemical study of 13 patients with acquired cutaneous lesions. JAMA. 1963;185:909-913. doi:10.1001/jama.1963.03060120019015
  2. Glazer AM, Sofen BD, Rigel DS, et al. Successful treatment of generalized essential telangiectasia with 6-mercaptopurine. J Drugs Dermatol. 2017;16:280-282.
  3. Pérez B, Núñez M, Boixeda P, et al. Progressive ascending telangiectasia treated with the 585 nm flashlamp-pumped pulsed dye laser. Lasers Surg Med. 1997;21:413-416. doi:10.1002/(sici)1096-9101(1997)21:5<413::aid-lsm1>3.0.co;2-t
  4. Buscaglia DA, Conte ET. Successful treatment of generalized essential telangiectasia with the 585-nm flashlamp-pumped pulsed dye laser. Cutis. 2001;67:107-108.
  5. Powell E, Markus R, Malone CH. Generalized essential telangiectasia treated with PDL. J Cosmet Dermatol. 2021;20:1086-1087. doi:10.1111/jocd.13938
  6. Ali MM, Teimory M, Sarhan M. Generalized essential telangiectasia with conjunctival involvement. Clin Exp Dermatol. 2006;31:781-782. doi:10.1111/j.1365-2230.2006.02217.x
  7. Shelley WB, Shelley ED. Essential progressive telangiectasia in an autoimmune setting: successful treatment with acyclovir. J Am Acad Dermatol. 1989;21(5 pt 2):1094-1096. doi:10.1016/s0190-9622(89)70303-0
  8. Shelley WB, Fierer JA. Focal intravascular coagulation in progressive ascending telangiectasia: ultrastructural studies of ketoconazole-induced involution of vessels. J Am Acad Dermatol. 1984;10(5 pt 2):876-887. doi:10.1016/s0190-9622(84)80439-9
  9. Wiznia LE, Steuer AB, Penn LA, et al. Generalized essential telangiectasia [published online December 15, 2018]. Dermatol Online J. doi:https://doi.org/10.5070/D32412042395
  10. Shelley WB. Essential progressive telangiectasia. successful treatment with tetracycline. JAMA. 1971;216:1343-1344.
  11. Checketts SR, Burton PS, Bjorkman DJ, et al. Generalized essential telangiectasia in the presence of gastrointestinal bleeding. J Am Acad Dermatol. 1997;37(2 pt 2):321-325.
  12. Gambichler T, Avermaete A, Wilmert M, et al. Generalized essential telangiectasia successfully treated with high-energy, long-pulse, frequency-doubled Nd:YAG laser. Dermatol Surg. 2001;27:355-357. doi:10.1046/j.1524-4725.2001.00307.x
  13. Fernández-Torres R, del Pozo J, de la Torre C, et al. Generalized essential telangiectasia: a report of three cases treated using an intense pulsed light system. Actas Dermosifiliogr. 2010;101:192-193.
  14. Tetart F, Lorthioir A, Girszyn N, et al. Watermelon stomach revealing generalized essential telangiectasia. Intern Med J. 2009;39:781-783. doi:10.1111/j.1445-5994.2009.02048.x
Article PDF
Author and Disclosure Information

Ms. Yu is from the School of Medicine, University of California, Riverside. Drs. Tawfik, Anderson, and Furukawa are from the Department of Dermatology, Loma Linda University Medical Center, California.

The authors report no conflict of interest.

Correspondence: Melanie Tawfik, MD, 25865 Barton Rd, Ste 101D, Loma Linda, CA 92354 (Melanie.masoud@gmail.com).

Issue
Cutis - 111(3)
Publications
Topics
Page Number
E12-E14
Sections
Author and Disclosure Information

Ms. Yu is from the School of Medicine, University of California, Riverside. Drs. Tawfik, Anderson, and Furukawa are from the Department of Dermatology, Loma Linda University Medical Center, California.

The authors report no conflict of interest.

Correspondence: Melanie Tawfik, MD, 25865 Barton Rd, Ste 101D, Loma Linda, CA 92354 (Melanie.masoud@gmail.com).

Author and Disclosure Information

Ms. Yu is from the School of Medicine, University of California, Riverside. Drs. Tawfik, Anderson, and Furukawa are from the Department of Dermatology, Loma Linda University Medical Center, California.

The authors report no conflict of interest.

Correspondence: Melanie Tawfik, MD, 25865 Barton Rd, Ste 101D, Loma Linda, CA 92354 (Melanie.masoud@gmail.com).

Article PDF
Article PDF

To the Editor:

Generalized essential telangiectasia (GET) is a rare, benign, and progressive primary cutaneous disease manifesting as telangiectases of the skin without systemic symptoms. It is unique in that it has widespread distribution on the body. Generalized essential telangiectasia more commonly affects women, usually in the fourth decade of life. The telangiectases most frequently appear on the legs, advancing over time to involve the trunk and arms and presenting in several patterns, including diffuse, macular, plaquelike, discrete, or confluent. Although GET typically is asymptomatic, numbness, tingling, and burning of the involved areas have been reported.1 Treatment modalities for GET vary, though pulsed dye laser (PDL) therapy is most common. We report the case of a 40-year-old woman with a 5-year history of GET who was treated successfully with PDL.

A 40-year-old woman presented to our dermatology clinic with progressive prominence of blood vessels involving the dorsal aspects of the feet of 5 years’ duration. The prominent vessels had spread to involve the legs (Figure 1), buttocks, lower abdomen, forearms, and medial upper arms. The patient denied any personal history of bleeding disorders or family history of inherited conditions associated with visceral vascular malformations, such as hereditary hemorrhagic telangiectasia. Notably, magnetic resonance imaging of the liver approximately 3 weeks prior to initiating treatment with PDL demonstrated multiple hepatic lesions consistent with hemangiomas. The patient reported an occasional tingling sensation in the feet. She was otherwise asymptomatic but did report psychological distress associated with the skin changes.

Erythematous to purpuric telangiectases on the lower legs of a 40-year-old woman with generalized essential telangiectasia prior to starting pulsed dye laser therapy
FIGURE 1. Erythematous to purpuric telangiectases on the lower legs of a 40-year-old woman with generalized essential telangiectasia prior to starting pulsed dye laser therapy.

Punch biopsies from the right lower leg and right buttock demonstrated increased vascularity of the dermis, a mild superficial perivascular lymphocytic infiltrate, and mild edema of the upper dermis without evidence of vasculitis. Autoimmune and coagulopathy workups were negative. The clinical and pathological findings were most consistent with GET.

Over the next 2.5 years, the patient underwent treatment with doxycycline and a series of 16 treatments with PDL (fluence, 6–12 J/cm2; pulse width, 10 milliseconds) with a positive cosmetic response. Considerable improvement in the lower legs was noted after 2 years of treatment with PDL (Figure 2).

The patient’s condition improved considerably, albeit transiently, after 2 years of pulsed dye laser therapy (fluence, 6–12 J/cm2 ; pulse width, 10 milliseconds).
FIGURE 2. The patient’s condition improved considerably, albeit transiently, after 2 years of pulsed dye laser therapy (fluence, 6–12 J/cm2 ; pulse width, 10 milliseconds).

Recurrence of GET was noted between PDL treatments, which led to progression of the disease process; all treated sites showed slow recurrence of lesions within several months after treatment. After 2 years, doxycycline was discontinued because of a perceived lack of continued benefit and the patient’s desire for alternative therapy. She was started on a 3-month trial of supplementation with ascorbic acid and rutin (or rutoside, a bioflavinoid), without noticeable improvement.

The diffuse distribution of dramatic telangiectases in GET makes treatment difficult. Standard treatments are not well established or studied due to the rarity of the condition. A review of PubMed articles indexed for MEDLINE using the terms treatment and generalized essential telangiectasias demonstrated several attempted treatment modalities for GET with varying success. In 4 cases in which PDL was used,2-5 a positive cosmetic response was noted, similar to what was seen in our patient. In 1 of the 4 cases, conservative management with ascorbic acid and compression stockings was unsuccessful; however, 6-mercaptopurine, used to treat that patient’s ulcerative colitis, incidentally resulted in resolution of GET.2 In 2 cases, response was maintained at 1.5-year follow-up.3,5 Two cases noted successful treatment with acyclovir,6,7 and 2 more demonstrated successful treatment with systemic ketoconazole.6,8 Some improvement was reported with oral doxycycline or tetracycline in 2 cases.9,10 Sclerotherapy improved the cosmetic appearance of telangiectases in one patient but was unsustainable because of the pain associated with the procedure.11 Nd:YAG laser therapy was effective in one case12; however, the patient experienced relapse at 6-month follow-up—similar to what we observed in our patient. Three patients treated with intense pulsed light therapy experienced results that were maintained at 2-year follow-up.13

Generalized essential telangiectasia generally is considered a skin-limited disease without systemic manifestations, but 2 reports11,14 described its association with gastric antral vascular ectasia—known as watermelon stomach. Hepatic hemangiomas are the most common benign liver lesions; however, the findings on magnetic resonance imaging in our patient, in combination with the 2 reported cases of watermelon stomach, suggest that the vascular changes of GET might extend below the skin.

Of the cases we reviewed, our patient had the longest reported duration of PDL treatment and follow-up for GET in which a successful, albeit transient, response was demonstrated. Our review of the literature revealed other reports of success with PDL and intense pulsed light therapy; results were maintained in some patients, while disease relapsed in others. Further studies are needed to understand why results are maintained in some but not all patients.

Although the cost of PDL as a cosmetic procedure must be taken into consideration when planning treatment of GET, we conclude that it is a safe option that can be effective until other treatment options are established to control the disease.

To the Editor:

Generalized essential telangiectasia (GET) is a rare, benign, and progressive primary cutaneous disease manifesting as telangiectases of the skin without systemic symptoms. It is unique in that it has widespread distribution on the body. Generalized essential telangiectasia more commonly affects women, usually in the fourth decade of life. The telangiectases most frequently appear on the legs, advancing over time to involve the trunk and arms and presenting in several patterns, including diffuse, macular, plaquelike, discrete, or confluent. Although GET typically is asymptomatic, numbness, tingling, and burning of the involved areas have been reported.1 Treatment modalities for GET vary, though pulsed dye laser (PDL) therapy is most common. We report the case of a 40-year-old woman with a 5-year history of GET who was treated successfully with PDL.

A 40-year-old woman presented to our dermatology clinic with progressive prominence of blood vessels involving the dorsal aspects of the feet of 5 years’ duration. The prominent vessels had spread to involve the legs (Figure 1), buttocks, lower abdomen, forearms, and medial upper arms. The patient denied any personal history of bleeding disorders or family history of inherited conditions associated with visceral vascular malformations, such as hereditary hemorrhagic telangiectasia. Notably, magnetic resonance imaging of the liver approximately 3 weeks prior to initiating treatment with PDL demonstrated multiple hepatic lesions consistent with hemangiomas. The patient reported an occasional tingling sensation in the feet. She was otherwise asymptomatic but did report psychological distress associated with the skin changes.

Erythematous to purpuric telangiectases on the lower legs of a 40-year-old woman with generalized essential telangiectasia prior to starting pulsed dye laser therapy
FIGURE 1. Erythematous to purpuric telangiectases on the lower legs of a 40-year-old woman with generalized essential telangiectasia prior to starting pulsed dye laser therapy.

Punch biopsies from the right lower leg and right buttock demonstrated increased vascularity of the dermis, a mild superficial perivascular lymphocytic infiltrate, and mild edema of the upper dermis without evidence of vasculitis. Autoimmune and coagulopathy workups were negative. The clinical and pathological findings were most consistent with GET.

Over the next 2.5 years, the patient underwent treatment with doxycycline and a series of 16 treatments with PDL (fluence, 6–12 J/cm2; pulse width, 10 milliseconds) with a positive cosmetic response. Considerable improvement in the lower legs was noted after 2 years of treatment with PDL (Figure 2).

The patient’s condition improved considerably, albeit transiently, after 2 years of pulsed dye laser therapy (fluence, 6–12 J/cm2 ; pulse width, 10 milliseconds).
FIGURE 2. The patient’s condition improved considerably, albeit transiently, after 2 years of pulsed dye laser therapy (fluence, 6–12 J/cm2 ; pulse width, 10 milliseconds).

Recurrence of GET was noted between PDL treatments, which led to progression of the disease process; all treated sites showed slow recurrence of lesions within several months after treatment. After 2 years, doxycycline was discontinued because of a perceived lack of continued benefit and the patient’s desire for alternative therapy. She was started on a 3-month trial of supplementation with ascorbic acid and rutin (or rutoside, a bioflavinoid), without noticeable improvement.

The diffuse distribution of dramatic telangiectases in GET makes treatment difficult. Standard treatments are not well established or studied due to the rarity of the condition. A review of PubMed articles indexed for MEDLINE using the terms treatment and generalized essential telangiectasias demonstrated several attempted treatment modalities for GET with varying success. In 4 cases in which PDL was used,2-5 a positive cosmetic response was noted, similar to what was seen in our patient. In 1 of the 4 cases, conservative management with ascorbic acid and compression stockings was unsuccessful; however, 6-mercaptopurine, used to treat that patient’s ulcerative colitis, incidentally resulted in resolution of GET.2 In 2 cases, response was maintained at 1.5-year follow-up.3,5 Two cases noted successful treatment with acyclovir,6,7 and 2 more demonstrated successful treatment with systemic ketoconazole.6,8 Some improvement was reported with oral doxycycline or tetracycline in 2 cases.9,10 Sclerotherapy improved the cosmetic appearance of telangiectases in one patient but was unsustainable because of the pain associated with the procedure.11 Nd:YAG laser therapy was effective in one case12; however, the patient experienced relapse at 6-month follow-up—similar to what we observed in our patient. Three patients treated with intense pulsed light therapy experienced results that were maintained at 2-year follow-up.13

Generalized essential telangiectasia generally is considered a skin-limited disease without systemic manifestations, but 2 reports11,14 described its association with gastric antral vascular ectasia—known as watermelon stomach. Hepatic hemangiomas are the most common benign liver lesions; however, the findings on magnetic resonance imaging in our patient, in combination with the 2 reported cases of watermelon stomach, suggest that the vascular changes of GET might extend below the skin.

Of the cases we reviewed, our patient had the longest reported duration of PDL treatment and follow-up for GET in which a successful, albeit transient, response was demonstrated. Our review of the literature revealed other reports of success with PDL and intense pulsed light therapy; results were maintained in some patients, while disease relapsed in others. Further studies are needed to understand why results are maintained in some but not all patients.

Although the cost of PDL as a cosmetic procedure must be taken into consideration when planning treatment of GET, we conclude that it is a safe option that can be effective until other treatment options are established to control the disease.

References
  1. McGrae JD Jr, Winkelmann RK. Generalized essential telangiectasia: report of a clinical and histochemical study of 13 patients with acquired cutaneous lesions. JAMA. 1963;185:909-913. doi:10.1001/jama.1963.03060120019015
  2. Glazer AM, Sofen BD, Rigel DS, et al. Successful treatment of generalized essential telangiectasia with 6-mercaptopurine. J Drugs Dermatol. 2017;16:280-282.
  3. Pérez B, Núñez M, Boixeda P, et al. Progressive ascending telangiectasia treated with the 585 nm flashlamp-pumped pulsed dye laser. Lasers Surg Med. 1997;21:413-416. doi:10.1002/(sici)1096-9101(1997)21:5<413::aid-lsm1>3.0.co;2-t
  4. Buscaglia DA, Conte ET. Successful treatment of generalized essential telangiectasia with the 585-nm flashlamp-pumped pulsed dye laser. Cutis. 2001;67:107-108.
  5. Powell E, Markus R, Malone CH. Generalized essential telangiectasia treated with PDL. J Cosmet Dermatol. 2021;20:1086-1087. doi:10.1111/jocd.13938
  6. Ali MM, Teimory M, Sarhan M. Generalized essential telangiectasia with conjunctival involvement. Clin Exp Dermatol. 2006;31:781-782. doi:10.1111/j.1365-2230.2006.02217.x
  7. Shelley WB, Shelley ED. Essential progressive telangiectasia in an autoimmune setting: successful treatment with acyclovir. J Am Acad Dermatol. 1989;21(5 pt 2):1094-1096. doi:10.1016/s0190-9622(89)70303-0
  8. Shelley WB, Fierer JA. Focal intravascular coagulation in progressive ascending telangiectasia: ultrastructural studies of ketoconazole-induced involution of vessels. J Am Acad Dermatol. 1984;10(5 pt 2):876-887. doi:10.1016/s0190-9622(84)80439-9
  9. Wiznia LE, Steuer AB, Penn LA, et al. Generalized essential telangiectasia [published online December 15, 2018]. Dermatol Online J. doi:https://doi.org/10.5070/D32412042395
  10. Shelley WB. Essential progressive telangiectasia. successful treatment with tetracycline. JAMA. 1971;216:1343-1344.
  11. Checketts SR, Burton PS, Bjorkman DJ, et al. Generalized essential telangiectasia in the presence of gastrointestinal bleeding. J Am Acad Dermatol. 1997;37(2 pt 2):321-325.
  12. Gambichler T, Avermaete A, Wilmert M, et al. Generalized essential telangiectasia successfully treated with high-energy, long-pulse, frequency-doubled Nd:YAG laser. Dermatol Surg. 2001;27:355-357. doi:10.1046/j.1524-4725.2001.00307.x
  13. Fernández-Torres R, del Pozo J, de la Torre C, et al. Generalized essential telangiectasia: a report of three cases treated using an intense pulsed light system. Actas Dermosifiliogr. 2010;101:192-193.
  14. Tetart F, Lorthioir A, Girszyn N, et al. Watermelon stomach revealing generalized essential telangiectasia. Intern Med J. 2009;39:781-783. doi:10.1111/j.1445-5994.2009.02048.x
References
  1. McGrae JD Jr, Winkelmann RK. Generalized essential telangiectasia: report of a clinical and histochemical study of 13 patients with acquired cutaneous lesions. JAMA. 1963;185:909-913. doi:10.1001/jama.1963.03060120019015
  2. Glazer AM, Sofen BD, Rigel DS, et al. Successful treatment of generalized essential telangiectasia with 6-mercaptopurine. J Drugs Dermatol. 2017;16:280-282.
  3. Pérez B, Núñez M, Boixeda P, et al. Progressive ascending telangiectasia treated with the 585 nm flashlamp-pumped pulsed dye laser. Lasers Surg Med. 1997;21:413-416. doi:10.1002/(sici)1096-9101(1997)21:5<413::aid-lsm1>3.0.co;2-t
  4. Buscaglia DA, Conte ET. Successful treatment of generalized essential telangiectasia with the 585-nm flashlamp-pumped pulsed dye laser. Cutis. 2001;67:107-108.
  5. Powell E, Markus R, Malone CH. Generalized essential telangiectasia treated with PDL. J Cosmet Dermatol. 2021;20:1086-1087. doi:10.1111/jocd.13938
  6. Ali MM, Teimory M, Sarhan M. Generalized essential telangiectasia with conjunctival involvement. Clin Exp Dermatol. 2006;31:781-782. doi:10.1111/j.1365-2230.2006.02217.x
  7. Shelley WB, Shelley ED. Essential progressive telangiectasia in an autoimmune setting: successful treatment with acyclovir. J Am Acad Dermatol. 1989;21(5 pt 2):1094-1096. doi:10.1016/s0190-9622(89)70303-0
  8. Shelley WB, Fierer JA. Focal intravascular coagulation in progressive ascending telangiectasia: ultrastructural studies of ketoconazole-induced involution of vessels. J Am Acad Dermatol. 1984;10(5 pt 2):876-887. doi:10.1016/s0190-9622(84)80439-9
  9. Wiznia LE, Steuer AB, Penn LA, et al. Generalized essential telangiectasia [published online December 15, 2018]. Dermatol Online J. doi:https://doi.org/10.5070/D32412042395
  10. Shelley WB. Essential progressive telangiectasia. successful treatment with tetracycline. JAMA. 1971;216:1343-1344.
  11. Checketts SR, Burton PS, Bjorkman DJ, et al. Generalized essential telangiectasia in the presence of gastrointestinal bleeding. J Am Acad Dermatol. 1997;37(2 pt 2):321-325.
  12. Gambichler T, Avermaete A, Wilmert M, et al. Generalized essential telangiectasia successfully treated with high-energy, long-pulse, frequency-doubled Nd:YAG laser. Dermatol Surg. 2001;27:355-357. doi:10.1046/j.1524-4725.2001.00307.x
  13. Fernández-Torres R, del Pozo J, de la Torre C, et al. Generalized essential telangiectasia: a report of three cases treated using an intense pulsed light system. Actas Dermosifiliogr. 2010;101:192-193.
  14. Tetart F, Lorthioir A, Girszyn N, et al. Watermelon stomach revealing generalized essential telangiectasia. Intern Med J. 2009;39:781-783. doi:10.1111/j.1445-5994.2009.02048.x
Issue
Cutis - 111(3)
Issue
Cutis - 111(3)
Page Number
E12-E14
Page Number
E12-E14
Publications
Publications
Topics
Article Type
Display Headline
Generalized Essential Telangiectasia Treated With Pulsed Dye Laser
Display Headline
Generalized Essential Telangiectasia Treated With Pulsed Dye Laser
Sections
Inside the Article

Practice Points

  • Generalized essential telangiectasia (GET) is a primary benign skin condition in which there is progressive development of telangiectases but a lack of systemic symptoms.
  • Although patients should be assured that GET is a benign disease, its manifestation on the skin may cause negative psychologic impacts that should not be overlooked.
  • Pulsed dye laser therapy does lead to improvement of the condition, but it does not prevent progression.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Fat Necrosis of the Breast Mimicking Breast Cancer in a Male Patient Following Wax Hair Removal

Article Type
Changed
Tue, 03/21/2023 - 07:52
Display Headline
Fat Necrosis of the Breast Mimicking Breast Cancer in a Male Patient Following Wax Hair Removal

To the Editor:

Fat necrosis of the breast is a benign inflammatory disease of adipose tissue commonly observed after trauma in the female breast during the perimenopausal period.1 Fat necrosis of the male breast is rare, first described by Silverstone2 in 1949; the condition usually presents with unilateral, painful or asymptomatic, firm nodules, which in rare cases are observed as skin retraction and thickening, ecchymosis, erythematous plaque–like cellulitis, local depression, and/or discoloration of the breast skin.3-5

Diagnosis of fat necrosis of the male breast may need to be confirmed via biopsy in conjunction with clinical and radiologic findings because the condition can mimic breast cancer.1 We report a case of bilateral fat necrosis of the breast mimicking breast cancer following wax hair removal.

A 42-year-old man presented to our outpatient dermatology clinic for evaluation of redness, swelling, and hardness of the skin of both breasts of 3 weeks’ duration. The patient had a history of wax hair removal of the entire anterior aspect of the body. He reported an erythematous, edematous, warm plaque that developed on the breasts 2 days after waxing. The plaque did not respond to antibiotics. The swelling and induration progressed over the 2 weeks after the patient was waxed. The patient had no family history of breast cancer. He had a standing diagnosis of gynecomastia. He denied any history of fat or filler injection in the affected area.

Dermatologic examination revealed erythematous, edematous, indurated, asymptomatic plaques with a peau d’orange appearance on the bilateral pectoral and presternal region. Minimal retraction of the right areola was noted (Figure 1). The bilateral axillary lymph nodes were palpable.

Erythematous, edematous, indurated, asymptomatic plaques with a peau d’orange appearance on the bilateral pectoral and presternal regions with minimal retraction of the right areola.
FIGURE 1. Erythematous, edematous, indurated, asymptomatic plaques with a peau d’orange appearance on the bilateral pectoral and presternal regions with minimal retraction of the right areola.

Laboratory results including erythrocyte sedimentation rate (108 mm/h [reference range, 2–20 mm/h]), C-reactive protein (9.2 mg/dL [reference range, >0.5 mg/dL]), and ferritin levels (645μg/L [reference range, 13–500 μg/L]) were consistent with inflammation; testing also included white blood cell count (8.5×103/μL [reference range, 4–10×103/μL]), hemoglobin (9.6 g/dL [reference range, 12–16 g/dL]), platelet count (437×103/μL [reference range, 100–400×103/μL]), procalcitonin (0.2 ng/mL [reference range, <0.3 ng/mL]), vitamin B12 (159 ng/L [reference range, 197–771 ng/L]), and folate (4.57 μg/L [reference range, 3.89–26.8 μg/L]). Other biochemical values were within reference range.

Mammography of both breasts revealed a Breast Imaging Reporting and Data System (BI-RADS) score of 4 with a suspicious abnormality (ie, diffuse edema of the breast, multiple calcifications in a nonspecific pattern, oil cysts with calcifications, and bilateral axillary lymphadenopathy with a diameter of 2.5 cm and a thick and irregular cortex)(Figure 2A). Ultrasonography of both breasts revealed an inflammatory breast. Magnetic resonance imaging showed similar findings with diffuse edema and a heterogeneous appearance. Contrast-enhanced magnetic resonance imaging showed diffuse contrast enhancement in both breasts extending to the pectoral muscles and axillary regions, consistent with inflammatory changes (Figure 2B).

Mammography revealed diffuse edema of the breast tissue, multiple calcifications in a nonspecific pattern, oil cysts with calcifications, and bilateral axillary lymphadenopathy with a diameter of 2.5 cm and a thick irregular cortex.
FIGURE 2. A, Mammography revealed diffuse edema of the breast tissue, multiple calcifications in a nonspecific pattern, oil cysts with calcifications, and bilateral axillary lymphadenopathy with a diameter of 2.5 cm and a thick irregular cortex. B, Contrast-enhanced magnetic resonance imaging revealed diffuse edema, a heterogeneous appearance, and diffuse contrast enhancement in both breasts extending to the pectoral muscles and axillary regions.

Because of difficulty differentiating inflammation and an infiltrating tumor, histopathologic examination was recommended by radiology. Results from a 5-mm punch biopsy from the right breast yielded the following differential diagnoses: cellulitis, panniculitis, inflammatory breast cancer, subcutaneous fat necrosis, and paraffinoma. Histopathologic examination of the skin revealed a normal epidermis and a dense inflammatory cell infiltrate comprising lymphocytes and monocytes in the dermis and subcutaneous tissue. Marked fibrosis also was noted in the dermis and subcutaneous tissue. Lipophagic fat necrosis accompanied by a variable inflammatory cell infiltrate consisted of histiocytes and neutrophils (Figure 3A). Pankeratin immunostaining was negative. Fat necrosis was present in a biopsy specimen obtained from the right breast; no signs of malignancy were present (Figure 3B). Fine-needle aspiration of the axillary lymph nodes was benign. Given these histopathologic findings, malignancy was excluded from the differential diagnosis. Paraffinoma also was ruled out because the patient insistently denied any history of fat or filler injection.

Skin biopsy and histopathology
FIGURE 3. A, Skin biopsy and histopathology demonstrated a normal epidermis, a dense inflammatory-cell infiltrate comprised of lymphocytes and monocytes as well as marked fibrosis in the dermis and subcutaneous tissue and lipophagic fat necrosis with an inflammatorycell infiltrate that contained histiocytes and neutrophils (H&E, original magnification ×10). B, Areas of fat necrosis were seen in a biopsy specimen (H&E, original magnification ×40).

Based on the clinical, histopathologic, and radiologic findings, as well as the history of minor trauma due to wax hair removal, a diagnosis of fat necrosis of the breast was made. Intervention was not recommended by the plastic surgeons who subsequently evaluated the patient, because the additional trauma may aggravate the lesion. He was treated with nonsteroidal anti-inflammatory drugs.

At 6-month follow-up, there was marked reduction in the erythema and edema but no notable improvement of the induration. A potent topical steroid was added to the treatment, but only slight regression of the induration was observed.

The normal male breast is comprised of fat and a few secretory ducts.6 Gynecomastia and breast cancer are the 2 most common conditions of the male breast; fat necrosis of the male breast is rare. In a study of 236 male patients with breast disease, only 5 had fat necrosis.7

Fat necrosis of the breast can be observed with various clinical and radiological presentations. Subcutaneous nodules, skin retraction and thickening, local skin depression, and ecchymosis are the more common presentations of fat necrosis.3-5 In our case, the first symptoms of disease were similar to those seen in cellulitis. The presentation of fat necrosis–like cellulitis has been described only rarely in the medical literature. Haikin et al5 reported a case of fat necrosis of the leg in a child that presented with cellulitis followed by induration, which did not respond to antibiotics, as was the case with our patient.5

Blunt trauma, breast reduction surgery, and breast augmentation surgery can cause fat necrosis of the breast1,4; in some cases, the cause cannot be determined.8 The only pertinent history in our patient was wax hair removal. Fat necrosis was an unexpected complication, but hair removal can be considered minor trauma; however, this is not commonly reported in the literature following hair removal with wax. In a study that reviewed diseases of the male breast, the investigators observed that all male patients with fat necrosis had pseudogynecomastia (adipomastia).7 Although our patient’s entire anterior trunk was epilated, only the breast was affected. This situation might be explained by underlying gynecomastia because fat necrosis is common in areas of the body where subcutaneous fat tissue is dense.

Fat necrosis of the breast can be mistaken—both clinically and radiologically—for malignancy, such as in our case. Diagnosis of fat necrosis of the breast should be a diagnosis of exclusion; therefore, histopathologic confirmation of the lesion is imperative.9

In conclusion, fat necrosis of the male breast is rare. The condition can present as cellulitis. Hair removal with wax might be a cause of fat necrosis. Because breast cancer and fat necrosis can exhibit clinical and radiologic similarities, the diagnosis of fat necrosis should be confirmed by histopathologic analysis in conjunction with clinical and radiologic findings.

References
  1. Tan PH, Lai LM, Carrington EV, et al. Fat necrosis of the breast—a review. Breast. 2006;15:313-318. doi:10.1016/j.breast.2005.07.003
  2. Silverstone M. Fat necrosis of the breast with report of a case in a male. Br J Surg. 1949;37:49-52. doi:10.1002/bjs.18003714508
  3. Akyol M, Kayali A, Yildirim N. Traumatic fat necrosis of male breast. Clin Imaging. 2013;37:954-956. doi:10.1016/j.clinimag.2013.05.009
  4. Crawford EA, King JJ, Fox EJ, et al. Symptomatic fat necrosis and lipoatrophy of the posterior pelvis following trauma. Orthopedics. 2009;32:444. doi:10.3928/01477447-20090511-25
  5. Haikin Herzberger E, Aviner S, Cherniavsky E. Posttraumatic fat necrosis presented as cellulitis of the leg. Case Rep Pediatr. 2012;2012:672397. doi:10.1155/2012/672397
  6. Michels LG, Gold RH, Arndt RD. Radiography of gynecomastia and other disorders of the male breast. Radiology. 1977;122:117-122. doi:10.1148/122.1.117
  7. Günhan-Bilgen I, Bozkaya H, Ustün E, et al. Male breast disease: clinical, mammographic, and ultrasonographic features. Eur J Radiol. 2002;43:246-255. doi:10.1016/s0720-048x(01)00483-1
  8. Chala LF, de Barros N, de Camargo Moraes P, et al. Fat necrosis of the breast: mammographic, sonographic, computed tomography, and magnetic resonance imaging findings. Curr Probl Diagn Radiol. 2004;33:106-126. doi:10.1067/j.cpradiol.2004.01.001
  9. Pullyblank AM, Davies JD, Basten J, et al. Fat necrosis of the female breast—Hadfield re-visited. Breast. 2001;10:388-391. doi:10.1054/brst.2000.0287
Article PDF
Author and Disclosure Information

Dr. Gore Karaali is from the Department of Dermatology, Irmet International Hospital, Turkey. Drs. N.D. Sarı, Altunkaynak, Leblebici, and Koku Aksu are from Istanbul Training and Research Hospital, University of Health Sciences, Turkey. Drs. N.D. Sarı and Altunkaynak are from the Department of Infectious Diseases and Clinical Microbiology, Dr. Leblebici is from the Department of Pathology, and Dr. Koku Aksu is from the Department of Dermatology. Dr. Y. Sarı is from the Department of Dermatology, Ankara Halil S¸ivgın Çubuk State Hospital, Turkey.

The authors report no conflict of interest.

Correspondence: Muge Gore Karaali, MD, Department of Dermatology, Irmet International Hospital, GOP Mah. Namık Kemal Bulvarı, No:17-21, Çerkezköy, Tekirdag˘, Turkey (mugegore@hotmail.com).

Issue
Cutis - 111(3)
Publications
Topics
Page Number
E9-E11
Sections
Author and Disclosure Information

Dr. Gore Karaali is from the Department of Dermatology, Irmet International Hospital, Turkey. Drs. N.D. Sarı, Altunkaynak, Leblebici, and Koku Aksu are from Istanbul Training and Research Hospital, University of Health Sciences, Turkey. Drs. N.D. Sarı and Altunkaynak are from the Department of Infectious Diseases and Clinical Microbiology, Dr. Leblebici is from the Department of Pathology, and Dr. Koku Aksu is from the Department of Dermatology. Dr. Y. Sarı is from the Department of Dermatology, Ankara Halil S¸ivgın Çubuk State Hospital, Turkey.

The authors report no conflict of interest.

Correspondence: Muge Gore Karaali, MD, Department of Dermatology, Irmet International Hospital, GOP Mah. Namık Kemal Bulvarı, No:17-21, Çerkezköy, Tekirdag˘, Turkey (mugegore@hotmail.com).

Author and Disclosure Information

Dr. Gore Karaali is from the Department of Dermatology, Irmet International Hospital, Turkey. Drs. N.D. Sarı, Altunkaynak, Leblebici, and Koku Aksu are from Istanbul Training and Research Hospital, University of Health Sciences, Turkey. Drs. N.D. Sarı and Altunkaynak are from the Department of Infectious Diseases and Clinical Microbiology, Dr. Leblebici is from the Department of Pathology, and Dr. Koku Aksu is from the Department of Dermatology. Dr. Y. Sarı is from the Department of Dermatology, Ankara Halil S¸ivgın Çubuk State Hospital, Turkey.

The authors report no conflict of interest.

Correspondence: Muge Gore Karaali, MD, Department of Dermatology, Irmet International Hospital, GOP Mah. Namık Kemal Bulvarı, No:17-21, Çerkezköy, Tekirdag˘, Turkey (mugegore@hotmail.com).

Article PDF
Article PDF

To the Editor:

Fat necrosis of the breast is a benign inflammatory disease of adipose tissue commonly observed after trauma in the female breast during the perimenopausal period.1 Fat necrosis of the male breast is rare, first described by Silverstone2 in 1949; the condition usually presents with unilateral, painful or asymptomatic, firm nodules, which in rare cases are observed as skin retraction and thickening, ecchymosis, erythematous plaque–like cellulitis, local depression, and/or discoloration of the breast skin.3-5

Diagnosis of fat necrosis of the male breast may need to be confirmed via biopsy in conjunction with clinical and radiologic findings because the condition can mimic breast cancer.1 We report a case of bilateral fat necrosis of the breast mimicking breast cancer following wax hair removal.

A 42-year-old man presented to our outpatient dermatology clinic for evaluation of redness, swelling, and hardness of the skin of both breasts of 3 weeks’ duration. The patient had a history of wax hair removal of the entire anterior aspect of the body. He reported an erythematous, edematous, warm plaque that developed on the breasts 2 days after waxing. The plaque did not respond to antibiotics. The swelling and induration progressed over the 2 weeks after the patient was waxed. The patient had no family history of breast cancer. He had a standing diagnosis of gynecomastia. He denied any history of fat or filler injection in the affected area.

Dermatologic examination revealed erythematous, edematous, indurated, asymptomatic plaques with a peau d’orange appearance on the bilateral pectoral and presternal region. Minimal retraction of the right areola was noted (Figure 1). The bilateral axillary lymph nodes were palpable.

Erythematous, edematous, indurated, asymptomatic plaques with a peau d’orange appearance on the bilateral pectoral and presternal regions with minimal retraction of the right areola.
FIGURE 1. Erythematous, edematous, indurated, asymptomatic plaques with a peau d’orange appearance on the bilateral pectoral and presternal regions with minimal retraction of the right areola.

Laboratory results including erythrocyte sedimentation rate (108 mm/h [reference range, 2–20 mm/h]), C-reactive protein (9.2 mg/dL [reference range, >0.5 mg/dL]), and ferritin levels (645μg/L [reference range, 13–500 μg/L]) were consistent with inflammation; testing also included white blood cell count (8.5×103/μL [reference range, 4–10×103/μL]), hemoglobin (9.6 g/dL [reference range, 12–16 g/dL]), platelet count (437×103/μL [reference range, 100–400×103/μL]), procalcitonin (0.2 ng/mL [reference range, <0.3 ng/mL]), vitamin B12 (159 ng/L [reference range, 197–771 ng/L]), and folate (4.57 μg/L [reference range, 3.89–26.8 μg/L]). Other biochemical values were within reference range.

Mammography of both breasts revealed a Breast Imaging Reporting and Data System (BI-RADS) score of 4 with a suspicious abnormality (ie, diffuse edema of the breast, multiple calcifications in a nonspecific pattern, oil cysts with calcifications, and bilateral axillary lymphadenopathy with a diameter of 2.5 cm and a thick and irregular cortex)(Figure 2A). Ultrasonography of both breasts revealed an inflammatory breast. Magnetic resonance imaging showed similar findings with diffuse edema and a heterogeneous appearance. Contrast-enhanced magnetic resonance imaging showed diffuse contrast enhancement in both breasts extending to the pectoral muscles and axillary regions, consistent with inflammatory changes (Figure 2B).

Mammography revealed diffuse edema of the breast tissue, multiple calcifications in a nonspecific pattern, oil cysts with calcifications, and bilateral axillary lymphadenopathy with a diameter of 2.5 cm and a thick irregular cortex.
FIGURE 2. A, Mammography revealed diffuse edema of the breast tissue, multiple calcifications in a nonspecific pattern, oil cysts with calcifications, and bilateral axillary lymphadenopathy with a diameter of 2.5 cm and a thick irregular cortex. B, Contrast-enhanced magnetic resonance imaging revealed diffuse edema, a heterogeneous appearance, and diffuse contrast enhancement in both breasts extending to the pectoral muscles and axillary regions.

Because of difficulty differentiating inflammation and an infiltrating tumor, histopathologic examination was recommended by radiology. Results from a 5-mm punch biopsy from the right breast yielded the following differential diagnoses: cellulitis, panniculitis, inflammatory breast cancer, subcutaneous fat necrosis, and paraffinoma. Histopathologic examination of the skin revealed a normal epidermis and a dense inflammatory cell infiltrate comprising lymphocytes and monocytes in the dermis and subcutaneous tissue. Marked fibrosis also was noted in the dermis and subcutaneous tissue. Lipophagic fat necrosis accompanied by a variable inflammatory cell infiltrate consisted of histiocytes and neutrophils (Figure 3A). Pankeratin immunostaining was negative. Fat necrosis was present in a biopsy specimen obtained from the right breast; no signs of malignancy were present (Figure 3B). Fine-needle aspiration of the axillary lymph nodes was benign. Given these histopathologic findings, malignancy was excluded from the differential diagnosis. Paraffinoma also was ruled out because the patient insistently denied any history of fat or filler injection.

Skin biopsy and histopathology
FIGURE 3. A, Skin biopsy and histopathology demonstrated a normal epidermis, a dense inflammatory-cell infiltrate comprised of lymphocytes and monocytes as well as marked fibrosis in the dermis and subcutaneous tissue and lipophagic fat necrosis with an inflammatorycell infiltrate that contained histiocytes and neutrophils (H&E, original magnification ×10). B, Areas of fat necrosis were seen in a biopsy specimen (H&E, original magnification ×40).

Based on the clinical, histopathologic, and radiologic findings, as well as the history of minor trauma due to wax hair removal, a diagnosis of fat necrosis of the breast was made. Intervention was not recommended by the plastic surgeons who subsequently evaluated the patient, because the additional trauma may aggravate the lesion. He was treated with nonsteroidal anti-inflammatory drugs.

At 6-month follow-up, there was marked reduction in the erythema and edema but no notable improvement of the induration. A potent topical steroid was added to the treatment, but only slight regression of the induration was observed.

The normal male breast is comprised of fat and a few secretory ducts.6 Gynecomastia and breast cancer are the 2 most common conditions of the male breast; fat necrosis of the male breast is rare. In a study of 236 male patients with breast disease, only 5 had fat necrosis.7

Fat necrosis of the breast can be observed with various clinical and radiological presentations. Subcutaneous nodules, skin retraction and thickening, local skin depression, and ecchymosis are the more common presentations of fat necrosis.3-5 In our case, the first symptoms of disease were similar to those seen in cellulitis. The presentation of fat necrosis–like cellulitis has been described only rarely in the medical literature. Haikin et al5 reported a case of fat necrosis of the leg in a child that presented with cellulitis followed by induration, which did not respond to antibiotics, as was the case with our patient.5

Blunt trauma, breast reduction surgery, and breast augmentation surgery can cause fat necrosis of the breast1,4; in some cases, the cause cannot be determined.8 The only pertinent history in our patient was wax hair removal. Fat necrosis was an unexpected complication, but hair removal can be considered minor trauma; however, this is not commonly reported in the literature following hair removal with wax. In a study that reviewed diseases of the male breast, the investigators observed that all male patients with fat necrosis had pseudogynecomastia (adipomastia).7 Although our patient’s entire anterior trunk was epilated, only the breast was affected. This situation might be explained by underlying gynecomastia because fat necrosis is common in areas of the body where subcutaneous fat tissue is dense.

Fat necrosis of the breast can be mistaken—both clinically and radiologically—for malignancy, such as in our case. Diagnosis of fat necrosis of the breast should be a diagnosis of exclusion; therefore, histopathologic confirmation of the lesion is imperative.9

In conclusion, fat necrosis of the male breast is rare. The condition can present as cellulitis. Hair removal with wax might be a cause of fat necrosis. Because breast cancer and fat necrosis can exhibit clinical and radiologic similarities, the diagnosis of fat necrosis should be confirmed by histopathologic analysis in conjunction with clinical and radiologic findings.

To the Editor:

Fat necrosis of the breast is a benign inflammatory disease of adipose tissue commonly observed after trauma in the female breast during the perimenopausal period.1 Fat necrosis of the male breast is rare, first described by Silverstone2 in 1949; the condition usually presents with unilateral, painful or asymptomatic, firm nodules, which in rare cases are observed as skin retraction and thickening, ecchymosis, erythematous plaque–like cellulitis, local depression, and/or discoloration of the breast skin.3-5

Diagnosis of fat necrosis of the male breast may need to be confirmed via biopsy in conjunction with clinical and radiologic findings because the condition can mimic breast cancer.1 We report a case of bilateral fat necrosis of the breast mimicking breast cancer following wax hair removal.

A 42-year-old man presented to our outpatient dermatology clinic for evaluation of redness, swelling, and hardness of the skin of both breasts of 3 weeks’ duration. The patient had a history of wax hair removal of the entire anterior aspect of the body. He reported an erythematous, edematous, warm plaque that developed on the breasts 2 days after waxing. The plaque did not respond to antibiotics. The swelling and induration progressed over the 2 weeks after the patient was waxed. The patient had no family history of breast cancer. He had a standing diagnosis of gynecomastia. He denied any history of fat or filler injection in the affected area.

Dermatologic examination revealed erythematous, edematous, indurated, asymptomatic plaques with a peau d’orange appearance on the bilateral pectoral and presternal region. Minimal retraction of the right areola was noted (Figure 1). The bilateral axillary lymph nodes were palpable.

Erythematous, edematous, indurated, asymptomatic plaques with a peau d’orange appearance on the bilateral pectoral and presternal regions with minimal retraction of the right areola.
FIGURE 1. Erythematous, edematous, indurated, asymptomatic plaques with a peau d’orange appearance on the bilateral pectoral and presternal regions with minimal retraction of the right areola.

Laboratory results including erythrocyte sedimentation rate (108 mm/h [reference range, 2–20 mm/h]), C-reactive protein (9.2 mg/dL [reference range, >0.5 mg/dL]), and ferritin levels (645μg/L [reference range, 13–500 μg/L]) were consistent with inflammation; testing also included white blood cell count (8.5×103/μL [reference range, 4–10×103/μL]), hemoglobin (9.6 g/dL [reference range, 12–16 g/dL]), platelet count (437×103/μL [reference range, 100–400×103/μL]), procalcitonin (0.2 ng/mL [reference range, <0.3 ng/mL]), vitamin B12 (159 ng/L [reference range, 197–771 ng/L]), and folate (4.57 μg/L [reference range, 3.89–26.8 μg/L]). Other biochemical values were within reference range.

Mammography of both breasts revealed a Breast Imaging Reporting and Data System (BI-RADS) score of 4 with a suspicious abnormality (ie, diffuse edema of the breast, multiple calcifications in a nonspecific pattern, oil cysts with calcifications, and bilateral axillary lymphadenopathy with a diameter of 2.5 cm and a thick and irregular cortex)(Figure 2A). Ultrasonography of both breasts revealed an inflammatory breast. Magnetic resonance imaging showed similar findings with diffuse edema and a heterogeneous appearance. Contrast-enhanced magnetic resonance imaging showed diffuse contrast enhancement in both breasts extending to the pectoral muscles and axillary regions, consistent with inflammatory changes (Figure 2B).

Mammography revealed diffuse edema of the breast tissue, multiple calcifications in a nonspecific pattern, oil cysts with calcifications, and bilateral axillary lymphadenopathy with a diameter of 2.5 cm and a thick irregular cortex.
FIGURE 2. A, Mammography revealed diffuse edema of the breast tissue, multiple calcifications in a nonspecific pattern, oil cysts with calcifications, and bilateral axillary lymphadenopathy with a diameter of 2.5 cm and a thick irregular cortex. B, Contrast-enhanced magnetic resonance imaging revealed diffuse edema, a heterogeneous appearance, and diffuse contrast enhancement in both breasts extending to the pectoral muscles and axillary regions.

Because of difficulty differentiating inflammation and an infiltrating tumor, histopathologic examination was recommended by radiology. Results from a 5-mm punch biopsy from the right breast yielded the following differential diagnoses: cellulitis, panniculitis, inflammatory breast cancer, subcutaneous fat necrosis, and paraffinoma. Histopathologic examination of the skin revealed a normal epidermis and a dense inflammatory cell infiltrate comprising lymphocytes and monocytes in the dermis and subcutaneous tissue. Marked fibrosis also was noted in the dermis and subcutaneous tissue. Lipophagic fat necrosis accompanied by a variable inflammatory cell infiltrate consisted of histiocytes and neutrophils (Figure 3A). Pankeratin immunostaining was negative. Fat necrosis was present in a biopsy specimen obtained from the right breast; no signs of malignancy were present (Figure 3B). Fine-needle aspiration of the axillary lymph nodes was benign. Given these histopathologic findings, malignancy was excluded from the differential diagnosis. Paraffinoma also was ruled out because the patient insistently denied any history of fat or filler injection.

Skin biopsy and histopathology
FIGURE 3. A, Skin biopsy and histopathology demonstrated a normal epidermis, a dense inflammatory-cell infiltrate comprised of lymphocytes and monocytes as well as marked fibrosis in the dermis and subcutaneous tissue and lipophagic fat necrosis with an inflammatorycell infiltrate that contained histiocytes and neutrophils (H&E, original magnification ×10). B, Areas of fat necrosis were seen in a biopsy specimen (H&E, original magnification ×40).

Based on the clinical, histopathologic, and radiologic findings, as well as the history of minor trauma due to wax hair removal, a diagnosis of fat necrosis of the breast was made. Intervention was not recommended by the plastic surgeons who subsequently evaluated the patient, because the additional trauma may aggravate the lesion. He was treated with nonsteroidal anti-inflammatory drugs.

At 6-month follow-up, there was marked reduction in the erythema and edema but no notable improvement of the induration. A potent topical steroid was added to the treatment, but only slight regression of the induration was observed.

The normal male breast is comprised of fat and a few secretory ducts.6 Gynecomastia and breast cancer are the 2 most common conditions of the male breast; fat necrosis of the male breast is rare. In a study of 236 male patients with breast disease, only 5 had fat necrosis.7

Fat necrosis of the breast can be observed with various clinical and radiological presentations. Subcutaneous nodules, skin retraction and thickening, local skin depression, and ecchymosis are the more common presentations of fat necrosis.3-5 In our case, the first symptoms of disease were similar to those seen in cellulitis. The presentation of fat necrosis–like cellulitis has been described only rarely in the medical literature. Haikin et al5 reported a case of fat necrosis of the leg in a child that presented with cellulitis followed by induration, which did not respond to antibiotics, as was the case with our patient.5

Blunt trauma, breast reduction surgery, and breast augmentation surgery can cause fat necrosis of the breast1,4; in some cases, the cause cannot be determined.8 The only pertinent history in our patient was wax hair removal. Fat necrosis was an unexpected complication, but hair removal can be considered minor trauma; however, this is not commonly reported in the literature following hair removal with wax. In a study that reviewed diseases of the male breast, the investigators observed that all male patients with fat necrosis had pseudogynecomastia (adipomastia).7 Although our patient’s entire anterior trunk was epilated, only the breast was affected. This situation might be explained by underlying gynecomastia because fat necrosis is common in areas of the body where subcutaneous fat tissue is dense.

Fat necrosis of the breast can be mistaken—both clinically and radiologically—for malignancy, such as in our case. Diagnosis of fat necrosis of the breast should be a diagnosis of exclusion; therefore, histopathologic confirmation of the lesion is imperative.9

In conclusion, fat necrosis of the male breast is rare. The condition can present as cellulitis. Hair removal with wax might be a cause of fat necrosis. Because breast cancer and fat necrosis can exhibit clinical and radiologic similarities, the diagnosis of fat necrosis should be confirmed by histopathologic analysis in conjunction with clinical and radiologic findings.

References
  1. Tan PH, Lai LM, Carrington EV, et al. Fat necrosis of the breast—a review. Breast. 2006;15:313-318. doi:10.1016/j.breast.2005.07.003
  2. Silverstone M. Fat necrosis of the breast with report of a case in a male. Br J Surg. 1949;37:49-52. doi:10.1002/bjs.18003714508
  3. Akyol M, Kayali A, Yildirim N. Traumatic fat necrosis of male breast. Clin Imaging. 2013;37:954-956. doi:10.1016/j.clinimag.2013.05.009
  4. Crawford EA, King JJ, Fox EJ, et al. Symptomatic fat necrosis and lipoatrophy of the posterior pelvis following trauma. Orthopedics. 2009;32:444. doi:10.3928/01477447-20090511-25
  5. Haikin Herzberger E, Aviner S, Cherniavsky E. Posttraumatic fat necrosis presented as cellulitis of the leg. Case Rep Pediatr. 2012;2012:672397. doi:10.1155/2012/672397
  6. Michels LG, Gold RH, Arndt RD. Radiography of gynecomastia and other disorders of the male breast. Radiology. 1977;122:117-122. doi:10.1148/122.1.117
  7. Günhan-Bilgen I, Bozkaya H, Ustün E, et al. Male breast disease: clinical, mammographic, and ultrasonographic features. Eur J Radiol. 2002;43:246-255. doi:10.1016/s0720-048x(01)00483-1
  8. Chala LF, de Barros N, de Camargo Moraes P, et al. Fat necrosis of the breast: mammographic, sonographic, computed tomography, and magnetic resonance imaging findings. Curr Probl Diagn Radiol. 2004;33:106-126. doi:10.1067/j.cpradiol.2004.01.001
  9. Pullyblank AM, Davies JD, Basten J, et al. Fat necrosis of the female breast—Hadfield re-visited. Breast. 2001;10:388-391. doi:10.1054/brst.2000.0287
References
  1. Tan PH, Lai LM, Carrington EV, et al. Fat necrosis of the breast—a review. Breast. 2006;15:313-318. doi:10.1016/j.breast.2005.07.003
  2. Silverstone M. Fat necrosis of the breast with report of a case in a male. Br J Surg. 1949;37:49-52. doi:10.1002/bjs.18003714508
  3. Akyol M, Kayali A, Yildirim N. Traumatic fat necrosis of male breast. Clin Imaging. 2013;37:954-956. doi:10.1016/j.clinimag.2013.05.009
  4. Crawford EA, King JJ, Fox EJ, et al. Symptomatic fat necrosis and lipoatrophy of the posterior pelvis following trauma. Orthopedics. 2009;32:444. doi:10.3928/01477447-20090511-25
  5. Haikin Herzberger E, Aviner S, Cherniavsky E. Posttraumatic fat necrosis presented as cellulitis of the leg. Case Rep Pediatr. 2012;2012:672397. doi:10.1155/2012/672397
  6. Michels LG, Gold RH, Arndt RD. Radiography of gynecomastia and other disorders of the male breast. Radiology. 1977;122:117-122. doi:10.1148/122.1.117
  7. Günhan-Bilgen I, Bozkaya H, Ustün E, et al. Male breast disease: clinical, mammographic, and ultrasonographic features. Eur J Radiol. 2002;43:246-255. doi:10.1016/s0720-048x(01)00483-1
  8. Chala LF, de Barros N, de Camargo Moraes P, et al. Fat necrosis of the breast: mammographic, sonographic, computed tomography, and magnetic resonance imaging findings. Curr Probl Diagn Radiol. 2004;33:106-126. doi:10.1067/j.cpradiol.2004.01.001
  9. Pullyblank AM, Davies JD, Basten J, et al. Fat necrosis of the female breast—Hadfield re-visited. Breast. 2001;10:388-391. doi:10.1054/brst.2000.0287
Issue
Cutis - 111(3)
Issue
Cutis - 111(3)
Page Number
E9-E11
Page Number
E9-E11
Publications
Publications
Topics
Article Type
Display Headline
Fat Necrosis of the Breast Mimicking Breast Cancer in a Male Patient Following Wax Hair Removal
Display Headline
Fat Necrosis of the Breast Mimicking Breast Cancer in a Male Patient Following Wax Hair Removal
Sections
Inside the Article

Practice Points

  • Fat necrosis of the breast can be mistaken—both clinically and radiologically—for malignancy; therefore, diagnosis should be confirmed by histopathology in conjunction with clinical and radiologic findings.
  • Fat necrosis of the male breast is rare, and hair removal with wax may be a rare cause of the disease.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Cyclosporine-Induced Posterior Reversible Encephalopathy Syndrome: An Adverse Effect in a Patient With Atopic Dermatitis

Article Type
Changed
Mon, 03/20/2023 - 16:26
Display Headline
Cyclosporine-Induced Posterior Reversible Encephalopathy Syndrome: An Adverse Effect in a Patient With Atopic Dermatitis

To the Editor:

Cyclosporine is an immunomodulatory medication that impacts T-lymphocyte function through calcineurin inhibition and suppression of IL-2 expression. Oral cyclosporine at low doses (1–3 mg/kg/d) is one of the more common systemic treatment options for moderate to severe atopic dermatitis. At these doses it has been shown to have therapeutic benefit in several skin conditions, including chronic spontaneous urticaria,1 psoriasis,2 and atopic dermatitis.3 When used at higher doses for conditions such as glomerulonephritis or transplantation, adverse effects may be notable, and close monitoring of drug metabolism as well as end-organ function is required. In contrast, severe adverse effects are uncommon with the lower doses of cyclosporine used for cutaneous conditions, and monitoring serum drug levels is not routinely practiced.4

A 58-year-old man was referred to clinic with severe atopic dermatitis refractory to maximal topical therapy prescribed by an outside physician. He was started on cyclosporine as an anticipated bridge to dupilumab biologic therapy. He had no history of hypertension, renal disease, or hepatic insufficiency prior to starting therapy. He demonstrated notable clinical improvement at a cyclosporine dosage of 300 mg/d (equating to 3.7 mg/kg/d). Three months after initiation of therapy, the patient presented to a local emergency department with new-onset seizurelike activity, confusion, and agitation. He was normotensive with clinical concern for status epilepticus. An initial laboratory assessment included a complete blood cell count, serum electrolyte panel, and urine toxicology screen, which were unremarkable. Computed tomography of the head showed confluent white-matter hypodensities in the left parietal-temporal-occipital lobes. Magnetic resonance imaging (MRI) of the brain showed innumerable peripherally distributed foci of microhemorrhage and vasogenic edema within the left parietal-temporal-occipital lobes (Figure).

Magnetic resonance imaging of the brain obtained in the emergency department at the time of presentation showed innumerable peripherally distributed foci of microhemorrhage and vasogenic edema within the left parietal-occipital-temporal lobes.
Magnetic resonance imaging of the brain obtained in the emergency department at the time of presentation showed innumerable peripherally distributed foci of microhemorrhage and vasogenic edema within the left parietal-occipital-temporal lobes.

He was intubated and sedated with admission to the medical intensive care unit, where a random cyclosporine level drawn approximately 9 hours after the prior dose was noted to be 263 ng/mL. Although target therapeutic levels for cyclosporine vary based on indication, toxic supratherapeutic levels generally are considered to be greater than 400 ng/mL.5 He had no evidence of acute kidney injury, uremia, or hypertension throughout hospitalization. An electroencephalogram showed left parieto-occipital periodic epileptiform discharges with generalized slowing. Cyclosporine was discontinued, and he was started on levetiracetam. His clinical and neuroimaging findings improved over the course of the 1-week hospitalization without any further intervention. Four weeks after hospitalization, he had full neurologic, electroencephalogram, and imaging recovery. Based on the presenting symptoms, transient neuroimaging findings, and full recovery with discontinuation of cyclosporine, the patient was diagnosed with cyclosporine-induced posterior reversible encephalopathy syndrome (PRES).

The diagnosis of PRES requires evidence of acute neurologic symptoms and radiographic findings of cortical/subcortical white-matter changes on computed tomography or MRI consistent with edema. The pathophysiology is not fully understood but appears to be related to vasogenic edema, primarily impacting the posterior aspect of the brain. There have been many reported offending agents, and symptoms typically resolve following cessation of these medications. Cases of cyclosporine-induced PRES have been reported, but most occurred at higher doses within weeks of medication initiation. Two cases of cyclosporine-induced PRES treated with cutaneous dosing have been reported; neither patient was taking it for atopic dermatitis.6

Cyclosporine-induced PRES remains a pathophysiologic conundrum. However, there is evidence to support direct endothelial damage causing cellular apoptosis in the brain of mouse models that is medication specific and not necessarily related to the dosages used.7 Our case highlights a rare but important adverse event associated with even low-dose cyclosporine use that should be considered in patients currently taking cyclosporine who present with acute neurologic changes.

References
  1. Kulthanan K, Chaweekulrat P, Komoltri C, et al. Cyclosporine for chronic spontaneous urticaria: a meta-analysis and systematic review. J Allergy Clin Immunol Pract. 2018;6:586-599. doi:10.1016/j.jaip.2017.07.017
  2. Armstrong AW, Read C. Pathophysiology, clinical presentation, and treatment of psoriasis: a review. JAMA. 2020;323:1945-1960. doi:10.1001/jama.2020.4006
  3. Seger EW, Wechter T, Strowd L, et al. Relative efficacy of systemic treatments for atopic dermatitis [published online October 6, 2018]. J Am Acad Dermatol. 2019;80:411-416.e4. doi:10.1016/j.jaad.2018.09.053
  4. Blake SC, Murrell DF. Monitoring trough levels in cyclosporine for atopic dermatitis: a systematic review. Pediatr Dermatol. 2019;36:843-853. doi:10.1111/pde.13999
  5. Tapia C, Nessel TA, Zito PM. Cyclosporine. StatPearls Publishing: 2022. https://www.ncbi.nlm.nih.gov/books/NBK482450/
  6. Cosottini M, Lazzarotti G, Ceravolo R, et al. Cyclosporine‐related posterior reversible encephalopathy syndrome (PRES) in non‐transplant patient: a case report and literature review. Eur J Neurol. 2003;10:461-462. doi:10.1046/j.1468-1331.2003.00608_1.x
  7. Kochi S, Takanaga H, Matsuo H, et al. Induction of apoptosis in mouse brain capillary endothelial cells by cyclosporin A and tacrolimus. Life Sci. 2000;66:2255-2260. doi:10.1016/s0024-3205(00)00554-3
Article PDF
Author and Disclosure Information

Dr. C.P. Dunn is from the Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle. Dr. C.L. Dunn is from the Department of Dermatology, KCU–GME/ADCS Consortium, Maitland, Florida. Dr. Petroni is from the Northwest Allergy and Asthma Center, University of Washington, Seattle.

The authors report no conflict of interest.

Correspondence: Clinton P. Dunn, MD, University of Washington Division of Allergy and Infectious Diseases, Department of Medicine, 750 Republican St, Box 358061, Seattle, WA 98109 (Cpdunn08@uw.edu).

Issue
Cutis - 111(3)
Publications
Topics
Page Number
E5-E6
Sections
Author and Disclosure Information

Dr. C.P. Dunn is from the Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle. Dr. C.L. Dunn is from the Department of Dermatology, KCU–GME/ADCS Consortium, Maitland, Florida. Dr. Petroni is from the Northwest Allergy and Asthma Center, University of Washington, Seattle.

The authors report no conflict of interest.

Correspondence: Clinton P. Dunn, MD, University of Washington Division of Allergy and Infectious Diseases, Department of Medicine, 750 Republican St, Box 358061, Seattle, WA 98109 (Cpdunn08@uw.edu).

Author and Disclosure Information

Dr. C.P. Dunn is from the Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle. Dr. C.L. Dunn is from the Department of Dermatology, KCU–GME/ADCS Consortium, Maitland, Florida. Dr. Petroni is from the Northwest Allergy and Asthma Center, University of Washington, Seattle.

The authors report no conflict of interest.

Correspondence: Clinton P. Dunn, MD, University of Washington Division of Allergy and Infectious Diseases, Department of Medicine, 750 Republican St, Box 358061, Seattle, WA 98109 (Cpdunn08@uw.edu).

Article PDF
Article PDF

To the Editor:

Cyclosporine is an immunomodulatory medication that impacts T-lymphocyte function through calcineurin inhibition and suppression of IL-2 expression. Oral cyclosporine at low doses (1–3 mg/kg/d) is one of the more common systemic treatment options for moderate to severe atopic dermatitis. At these doses it has been shown to have therapeutic benefit in several skin conditions, including chronic spontaneous urticaria,1 psoriasis,2 and atopic dermatitis.3 When used at higher doses for conditions such as glomerulonephritis or transplantation, adverse effects may be notable, and close monitoring of drug metabolism as well as end-organ function is required. In contrast, severe adverse effects are uncommon with the lower doses of cyclosporine used for cutaneous conditions, and monitoring serum drug levels is not routinely practiced.4

A 58-year-old man was referred to clinic with severe atopic dermatitis refractory to maximal topical therapy prescribed by an outside physician. He was started on cyclosporine as an anticipated bridge to dupilumab biologic therapy. He had no history of hypertension, renal disease, or hepatic insufficiency prior to starting therapy. He demonstrated notable clinical improvement at a cyclosporine dosage of 300 mg/d (equating to 3.7 mg/kg/d). Three months after initiation of therapy, the patient presented to a local emergency department with new-onset seizurelike activity, confusion, and agitation. He was normotensive with clinical concern for status epilepticus. An initial laboratory assessment included a complete blood cell count, serum electrolyte panel, and urine toxicology screen, which were unremarkable. Computed tomography of the head showed confluent white-matter hypodensities in the left parietal-temporal-occipital lobes. Magnetic resonance imaging (MRI) of the brain showed innumerable peripherally distributed foci of microhemorrhage and vasogenic edema within the left parietal-temporal-occipital lobes (Figure).

Magnetic resonance imaging of the brain obtained in the emergency department at the time of presentation showed innumerable peripherally distributed foci of microhemorrhage and vasogenic edema within the left parietal-occipital-temporal lobes.
Magnetic resonance imaging of the brain obtained in the emergency department at the time of presentation showed innumerable peripherally distributed foci of microhemorrhage and vasogenic edema within the left parietal-occipital-temporal lobes.

He was intubated and sedated with admission to the medical intensive care unit, where a random cyclosporine level drawn approximately 9 hours after the prior dose was noted to be 263 ng/mL. Although target therapeutic levels for cyclosporine vary based on indication, toxic supratherapeutic levels generally are considered to be greater than 400 ng/mL.5 He had no evidence of acute kidney injury, uremia, or hypertension throughout hospitalization. An electroencephalogram showed left parieto-occipital periodic epileptiform discharges with generalized slowing. Cyclosporine was discontinued, and he was started on levetiracetam. His clinical and neuroimaging findings improved over the course of the 1-week hospitalization without any further intervention. Four weeks after hospitalization, he had full neurologic, electroencephalogram, and imaging recovery. Based on the presenting symptoms, transient neuroimaging findings, and full recovery with discontinuation of cyclosporine, the patient was diagnosed with cyclosporine-induced posterior reversible encephalopathy syndrome (PRES).

The diagnosis of PRES requires evidence of acute neurologic symptoms and radiographic findings of cortical/subcortical white-matter changes on computed tomography or MRI consistent with edema. The pathophysiology is not fully understood but appears to be related to vasogenic edema, primarily impacting the posterior aspect of the brain. There have been many reported offending agents, and symptoms typically resolve following cessation of these medications. Cases of cyclosporine-induced PRES have been reported, but most occurred at higher doses within weeks of medication initiation. Two cases of cyclosporine-induced PRES treated with cutaneous dosing have been reported; neither patient was taking it for atopic dermatitis.6

Cyclosporine-induced PRES remains a pathophysiologic conundrum. However, there is evidence to support direct endothelial damage causing cellular apoptosis in the brain of mouse models that is medication specific and not necessarily related to the dosages used.7 Our case highlights a rare but important adverse event associated with even low-dose cyclosporine use that should be considered in patients currently taking cyclosporine who present with acute neurologic changes.

To the Editor:

Cyclosporine is an immunomodulatory medication that impacts T-lymphocyte function through calcineurin inhibition and suppression of IL-2 expression. Oral cyclosporine at low doses (1–3 mg/kg/d) is one of the more common systemic treatment options for moderate to severe atopic dermatitis. At these doses it has been shown to have therapeutic benefit in several skin conditions, including chronic spontaneous urticaria,1 psoriasis,2 and atopic dermatitis.3 When used at higher doses for conditions such as glomerulonephritis or transplantation, adverse effects may be notable, and close monitoring of drug metabolism as well as end-organ function is required. In contrast, severe adverse effects are uncommon with the lower doses of cyclosporine used for cutaneous conditions, and monitoring serum drug levels is not routinely practiced.4

A 58-year-old man was referred to clinic with severe atopic dermatitis refractory to maximal topical therapy prescribed by an outside physician. He was started on cyclosporine as an anticipated bridge to dupilumab biologic therapy. He had no history of hypertension, renal disease, or hepatic insufficiency prior to starting therapy. He demonstrated notable clinical improvement at a cyclosporine dosage of 300 mg/d (equating to 3.7 mg/kg/d). Three months after initiation of therapy, the patient presented to a local emergency department with new-onset seizurelike activity, confusion, and agitation. He was normotensive with clinical concern for status epilepticus. An initial laboratory assessment included a complete blood cell count, serum electrolyte panel, and urine toxicology screen, which were unremarkable. Computed tomography of the head showed confluent white-matter hypodensities in the left parietal-temporal-occipital lobes. Magnetic resonance imaging (MRI) of the brain showed innumerable peripherally distributed foci of microhemorrhage and vasogenic edema within the left parietal-temporal-occipital lobes (Figure).

Magnetic resonance imaging of the brain obtained in the emergency department at the time of presentation showed innumerable peripherally distributed foci of microhemorrhage and vasogenic edema within the left parietal-occipital-temporal lobes.
Magnetic resonance imaging of the brain obtained in the emergency department at the time of presentation showed innumerable peripherally distributed foci of microhemorrhage and vasogenic edema within the left parietal-occipital-temporal lobes.

He was intubated and sedated with admission to the medical intensive care unit, where a random cyclosporine level drawn approximately 9 hours after the prior dose was noted to be 263 ng/mL. Although target therapeutic levels for cyclosporine vary based on indication, toxic supratherapeutic levels generally are considered to be greater than 400 ng/mL.5 He had no evidence of acute kidney injury, uremia, or hypertension throughout hospitalization. An electroencephalogram showed left parieto-occipital periodic epileptiform discharges with generalized slowing. Cyclosporine was discontinued, and he was started on levetiracetam. His clinical and neuroimaging findings improved over the course of the 1-week hospitalization without any further intervention. Four weeks after hospitalization, he had full neurologic, electroencephalogram, and imaging recovery. Based on the presenting symptoms, transient neuroimaging findings, and full recovery with discontinuation of cyclosporine, the patient was diagnosed with cyclosporine-induced posterior reversible encephalopathy syndrome (PRES).

The diagnosis of PRES requires evidence of acute neurologic symptoms and radiographic findings of cortical/subcortical white-matter changes on computed tomography or MRI consistent with edema. The pathophysiology is not fully understood but appears to be related to vasogenic edema, primarily impacting the posterior aspect of the brain. There have been many reported offending agents, and symptoms typically resolve following cessation of these medications. Cases of cyclosporine-induced PRES have been reported, but most occurred at higher doses within weeks of medication initiation. Two cases of cyclosporine-induced PRES treated with cutaneous dosing have been reported; neither patient was taking it for atopic dermatitis.6

Cyclosporine-induced PRES remains a pathophysiologic conundrum. However, there is evidence to support direct endothelial damage causing cellular apoptosis in the brain of mouse models that is medication specific and not necessarily related to the dosages used.7 Our case highlights a rare but important adverse event associated with even low-dose cyclosporine use that should be considered in patients currently taking cyclosporine who present with acute neurologic changes.

References
  1. Kulthanan K, Chaweekulrat P, Komoltri C, et al. Cyclosporine for chronic spontaneous urticaria: a meta-analysis and systematic review. J Allergy Clin Immunol Pract. 2018;6:586-599. doi:10.1016/j.jaip.2017.07.017
  2. Armstrong AW, Read C. Pathophysiology, clinical presentation, and treatment of psoriasis: a review. JAMA. 2020;323:1945-1960. doi:10.1001/jama.2020.4006
  3. Seger EW, Wechter T, Strowd L, et al. Relative efficacy of systemic treatments for atopic dermatitis [published online October 6, 2018]. J Am Acad Dermatol. 2019;80:411-416.e4. doi:10.1016/j.jaad.2018.09.053
  4. Blake SC, Murrell DF. Monitoring trough levels in cyclosporine for atopic dermatitis: a systematic review. Pediatr Dermatol. 2019;36:843-853. doi:10.1111/pde.13999
  5. Tapia C, Nessel TA, Zito PM. Cyclosporine. StatPearls Publishing: 2022. https://www.ncbi.nlm.nih.gov/books/NBK482450/
  6. Cosottini M, Lazzarotti G, Ceravolo R, et al. Cyclosporine‐related posterior reversible encephalopathy syndrome (PRES) in non‐transplant patient: a case report and literature review. Eur J Neurol. 2003;10:461-462. doi:10.1046/j.1468-1331.2003.00608_1.x
  7. Kochi S, Takanaga H, Matsuo H, et al. Induction of apoptosis in mouse brain capillary endothelial cells by cyclosporin A and tacrolimus. Life Sci. 2000;66:2255-2260. doi:10.1016/s0024-3205(00)00554-3
References
  1. Kulthanan K, Chaweekulrat P, Komoltri C, et al. Cyclosporine for chronic spontaneous urticaria: a meta-analysis and systematic review. J Allergy Clin Immunol Pract. 2018;6:586-599. doi:10.1016/j.jaip.2017.07.017
  2. Armstrong AW, Read C. Pathophysiology, clinical presentation, and treatment of psoriasis: a review. JAMA. 2020;323:1945-1960. doi:10.1001/jama.2020.4006
  3. Seger EW, Wechter T, Strowd L, et al. Relative efficacy of systemic treatments for atopic dermatitis [published online October 6, 2018]. J Am Acad Dermatol. 2019;80:411-416.e4. doi:10.1016/j.jaad.2018.09.053
  4. Blake SC, Murrell DF. Monitoring trough levels in cyclosporine for atopic dermatitis: a systematic review. Pediatr Dermatol. 2019;36:843-853. doi:10.1111/pde.13999
  5. Tapia C, Nessel TA, Zito PM. Cyclosporine. StatPearls Publishing: 2022. https://www.ncbi.nlm.nih.gov/books/NBK482450/
  6. Cosottini M, Lazzarotti G, Ceravolo R, et al. Cyclosporine‐related posterior reversible encephalopathy syndrome (PRES) in non‐transplant patient: a case report and literature review. Eur J Neurol. 2003;10:461-462. doi:10.1046/j.1468-1331.2003.00608_1.x
  7. Kochi S, Takanaga H, Matsuo H, et al. Induction of apoptosis in mouse brain capillary endothelial cells by cyclosporin A and tacrolimus. Life Sci. 2000;66:2255-2260. doi:10.1016/s0024-3205(00)00554-3
Issue
Cutis - 111(3)
Issue
Cutis - 111(3)
Page Number
E5-E6
Page Number
E5-E6
Publications
Publications
Topics
Article Type
Display Headline
Cyclosporine-Induced Posterior Reversible Encephalopathy Syndrome: An Adverse Effect in a Patient With Atopic Dermatitis
Display Headline
Cyclosporine-Induced Posterior Reversible Encephalopathy Syndrome: An Adverse Effect in a Patient With Atopic Dermatitis
Sections
Inside the Article

Practice Points

  • Cyclosporine is an immunomodulatory therapeutic utilized for several indications in dermatology practice, most commonly in low doses.
  • Posterior reversible encephalopathy syndrome (PRES) is a known but rare adverse effect of cyclosporine presenting with acute encephalopathic changes and radiographic findings on central imaging.
  • Knowledge of this association is critical, as symptoms are reversible with prompt recognition, appropriate inpatient supportive care, and discontinuation of offending medications.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media