LayerRx Mapping ID
245
Slot System
Featured Buckets
Featured Buckets Admin

Erythematous Flaky Rash on the Toe

Article Type
Changed
Tue, 06/11/2024 - 11:59
Display Headline
Erythematous Flaky Rash on the Toe

The Diagnosis: Necrolytic Migratory Erythema

Necrolytic migratory erythema (NME) is a waxing and waning rash associated with rare pancreatic neuroendocrine tumors called glucagonomas. It is characterized by pruritic and painful, well-demarcated, erythematous plaques that manifest in the intertriginous areas and on the perineum and buttocks.1 Due to the evolving nature of the rash, the histopathologic findings in NME vary depending on the stage of the cutaneous lesions at the time of biopsy.2 Multiple dyskeratotic keratinocytes spanning all epidermal layers may be a diagnostic clue in early lesions of NME.3 Typical features of longstanding lesions include confluent parakeratosis, psoriasiform hyperplasia with mild or absent spongiosis, and upper epidermal necrosis with keratinocyte vacuolization and pallor.4 Morphologic features that are present prior to the development of epidermal vacuolation and necrosis frequently are misattributed to psoriasis or eczema. Long-standing lesions also may develop a neutrophilic infiltrate with subcorneal and intraepidermal pustules.2 Other common features include a discrete perivascular lymphocytic infiltrate and an erosive or encrusted epidermis.5 Although direct immunofluorescence typically is negative, nonspecific findings can be seen, including apoptotic keratinocytes labeling with fibrinogen and C3, as well as scattered, clumped, IgM-positive cytoid bodies present at the dermal-epidermal junction (DEJ).6 Biopsies also have shown scattered, clumped, IgM-positive cytoid bodies present at the DEJ.5

Psoriasis is a chronic relapsing papulosquamous disorder characterized by scaly erythematous plaques often overlying the extensor surfaces of the extremities. Histopathology shows a psoriasiform pattern of inflammation with thinning of the suprapapillary plates and elongation of the rete ridges. Further diagnostic clues of psoriasis include regular acanthosis, characteristic Munro microabscesses with neutrophils in a hyperkeratotic stratum corneum (Figure 1), hypogranulosis, and neutrophilic spongiform pustules of Kogoj in the stratum spinosum. Generally, there is a lack of the epidermal necrosis seen with NME.7,8

Lichen simplex chronicus manifests as pruritic, often hyperpigmented, well-defined, lichenified plaques with excoriation following repetitive mechanical trauma, commonly on the lower lateral legs, posterior neck, and flexural areas.9 The histologic landscape is marked by well-developed lesions evolving to show compact orthokeratosis, hypergranulosis, irregularly elongated rete ridges (ie, irregular acanthosis), and papillary dermal fibrosis with vertical streaking of collagen (Figure 2).9,10

Subacute cutaneous lupus erythematosus (SCLE) is recognized clinically by scaly/psoriasiform and annular lesions with mild or absent systemic involvement. Common histopathologic findings include epidermal atrophy, vacuolar interface dermatitis with hydropic degeneration of the basal layer, a subepidermal lymphocytic infiltrate, and a periadnexal and perivascular infiltrate (Figure 3).11 Upper dermal edema, spotty necrosis of individual cells in the epidermis, dermal-epidermal separation caused by prominent basal cell degeneration, and accumulation of acid mucopolysaccharides (mucin) are other histologic features associated with SCLE.12,13

fruprububiuubrodiwrasavothihapulauedrotojafredisugiphavipranomuciswiprowowachochemuwadrokespimusotruchipuspistajesibaspuluspilivashaspesheciwrasteswedredilurabeprobaphachubishemislimumoclo
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Psoriasis%20shows%20hyperkeratosis%20with%20neutrophils%20in%20the%20stratum%20corneum%20on%20histopathology%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%3C%2Fp%3E

mecrifrigugadrijuwrabujudehauilokowutruvidrubuspebaslima
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Lichen%20simplex%20chronicus%20shows%20a%20compact%20stratum%20corneum%2C%20irregular%20acanthosis%2C%20and%20papillary%20dermal%20fibrosis%20on%20biopsy%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9710).%3C%2Fp%3E

The immunofluorescence pattern in SCLE features dustlike particles of IgG deposition in the epidermis, subepidermal region, and dermal cellular infiltrate. Lesions also may have granular deposition of immunoreactions at the DEJ.11,13

racruhecresocosisletribruvesteprospiphenukogitaclerouasloslojibem
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Subacute%20cutaneous%20lupus%20erythematosus%20shows%20vacuolar%20interface%20dermatitis%20with%20epidermal%20atrophy%2C%20subepidermal%20lymphocytes%2C%20and%20perivascular%20inflammation%20on%20biopsy%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9710).%3C%2Fp%3E

cutrewristicluu
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Drug%20reaction%20with%20eosinophilia%20and%20systemic%20symptoms%20(DRESS)%20syndrome%20shows%20spongiosis%20with%20a%20perivascular%20infiltrate%20on%20biopsy%3B%20eosinophils%20are%20variably%20observed%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9710).%3C%2Fp%3E

The manifestation of drug reaction with eosinophilia and systemic symptoms (DRESS) syndrome (also known as drug-induced hypersensitivity syndrome) is variable, with a morbilliform rash that spreads from the face to the entire body, urticaria, atypical target lesions, purpuriform lesions, lymphadenopathy, and exfoliative dermatitis.14 The nonspecific morphologic features of DRESS syndrome lesions are associated with variable histologic features, which include focal interface changes with vacuolar alteration of the basal layer; atypical lymphocytes with hyperchromic nuclei; and a superficial, inconsistently dense, perivascular lymphocytic infiltrate. Other relatively common histopathologic patterns include an upper dermis with dilated blood vessels, spongiosis with exocytosis of lymphocytes (Figure 4), and necrotic keratinocytes. Although peripheral eosinophilia is an important diagnostic criterion and is observed consistently, eosinophils are variably present on skin biopsy.15,16 Given the histopathologic variability and nonspecific findings, clinical correlation is required when diagnosing DRESS syndrome.

References
  1. Halvorson SA, Gilbert E, Hopkins RS, et al. Putting the pieces together: necrolytic migratory erythema and the glucagonoma syndrome. J Gen Intern Med. 2013;28:1525-1529. doi:10.1007 /s11606-013-2490-5
  2. Toberer F, Hartschuh W, Wiedemeyer K. Glucagonoma-associated necrolytic migratory erythema: the broad spectrum of the clinical and histopathological findings and clues to the diagnosis. Am J Dermatopathol. 2019;41:E29-E32. doi:10.1097DAD .0000000000001219
  3. Hunt SJ, Narus VT, Abell E. Necrolytic migratory erythema: dyskeratotic dermatitis, a clue to early diagnosis. J Am Acad Dermatol. 1991; 24:473-477. doi:10.1016/0190-9622(91)70076-e
  4. van Beek AP, de Haas ER, van Vloten WA, et al. The glucagonoma syndrome and necrolytic migratory erythema: a clinical review. Eur J Endocrinol. 2004;151:531-537. doi:10.1530/eje.0.1510531
  5. Pujol RM, Wang C-Y E, el-Azhary RA, et al. Necrolytic migratory erythema: clinicopathologic study of 13 cases. Int J Dermatol. 2004;43:12- 18. doi:10.1111/j.1365-4632.2004.01844.x
  6. Johnson SM, Smoller BR, Lamps LW, et al. Necrolytic migratory erythema as the only presenting sign of a glucagonoma. J Am Acad Dermatol. 2003;49:325-328. doi:10.1067/s0190-9622(02)61774-8
  7. De Rosa G, Mignogna C. The histopathology of psoriasis. Reumatismo. 2007;59(suppl 1):46-48. doi:10.4081/reumatismo.2007.1s.46
  8. Kimmel GW, Lebwohl M. Psoriasis: overview and diagnosis. In: Bhutani T, Liao W, Nakamura M, eds. Evidence-Based Psoriasis. Springer; 2018:1-16. doi:10.1007/978-3-319-90107-7_1
  9. Balan R, Grigoras¸ A, Popovici D, et al. The histopathological landscape of the major psoriasiform dermatoses. Arch Clin Cases. 2021;6:59-68. doi:10.22551/2019.24.0603.10155
  10. O’Keefe RJ, Scurry JP, Dennerstein G, et al. Audit of 114 nonneoplastic vulvar biopsies. Br J Obstet Gynaecol. 1995;102:780-786. doi:10.1111/j.1471-0528.1995.tb10842.x
  11. Parodi A, Caproni M, Cardinali C, et al P. Clinical, histological and immunopathological features of 58 patients with subacute cutaneous lupus erythematosus. Dermatology. 2000;200:6-10. doi:10.1159/000018307
  12. Lyon CC, Blewitt R, Harrison PV. Subacute cutaneous lupus erythematosus: two cases of delayed diagnosis. Acta Derm Venereol. 1998;78:57-59. doi:10.1080/00015559850135869
  13. David-Bajar KM. Subacute cutaneous lupus erythematosus. J Invest Dermatol. 1993;100:2S-8S. doi:10.1111/1523-1747.ep12355164
  14. Paulmann M, Mockenhaupt M. Severe drug-induced skin reactions: clinical features, diagnosis, etiology, and therapy. J Dtsch Dermatol Ges. 2015;13:625-643. doi:10.1111/ddg.12747
  15. Borroni G, Torti S, Pezzini C, et al. Histopathologic spectrum of drug reaction with eosinophilia and systemic symptoms (DRESS): a diagnosis that needs clinico-pathological correlation. G Ital Dermatol Venereol. 2014;149:291-300.
  16. Ortonne N, Valeyrie-Allanore L, Bastuji-Garin S, et al. Histopathology of drug rash with eosinophilia and systemic symptoms syndrome: a morphological and phenotypical study. Br J Dermatol. 2015;173:50-58. doi:10.1111/bjd.13683
Article PDF
Author and Disclosure Information

Sarah K. Friske is from the School of Medicine, Baylor College of Medicine, Houston, Texas. Drs. Wiggins, Carrigg, and Bohlke are from Good Samaritan Health Services/Frontier Derm, Salem, Oregon. Dr. Seervai is from Oregon Health & Science University, Portland.

The authors report no conflict of interest.

Correspondence: Riyad N.H. Seervai, MD, PhD, 3303 S Bond Ave, Bldg 1, Portland, OR 97239 (seervai@ohsu.edu).

Issue
Cutis - 113(6)
Publications
Topics
Page Number
234,258-259
Sections
Author and Disclosure Information

Sarah K. Friske is from the School of Medicine, Baylor College of Medicine, Houston, Texas. Drs. Wiggins, Carrigg, and Bohlke are from Good Samaritan Health Services/Frontier Derm, Salem, Oregon. Dr. Seervai is from Oregon Health & Science University, Portland.

The authors report no conflict of interest.

Correspondence: Riyad N.H. Seervai, MD, PhD, 3303 S Bond Ave, Bldg 1, Portland, OR 97239 (seervai@ohsu.edu).

Author and Disclosure Information

Sarah K. Friske is from the School of Medicine, Baylor College of Medicine, Houston, Texas. Drs. Wiggins, Carrigg, and Bohlke are from Good Samaritan Health Services/Frontier Derm, Salem, Oregon. Dr. Seervai is from Oregon Health & Science University, Portland.

The authors report no conflict of interest.

Correspondence: Riyad N.H. Seervai, MD, PhD, 3303 S Bond Ave, Bldg 1, Portland, OR 97239 (seervai@ohsu.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Necrolytic Migratory Erythema

Necrolytic migratory erythema (NME) is a waxing and waning rash associated with rare pancreatic neuroendocrine tumors called glucagonomas. It is characterized by pruritic and painful, well-demarcated, erythematous plaques that manifest in the intertriginous areas and on the perineum and buttocks.1 Due to the evolving nature of the rash, the histopathologic findings in NME vary depending on the stage of the cutaneous lesions at the time of biopsy.2 Multiple dyskeratotic keratinocytes spanning all epidermal layers may be a diagnostic clue in early lesions of NME.3 Typical features of longstanding lesions include confluent parakeratosis, psoriasiform hyperplasia with mild or absent spongiosis, and upper epidermal necrosis with keratinocyte vacuolization and pallor.4 Morphologic features that are present prior to the development of epidermal vacuolation and necrosis frequently are misattributed to psoriasis or eczema. Long-standing lesions also may develop a neutrophilic infiltrate with subcorneal and intraepidermal pustules.2 Other common features include a discrete perivascular lymphocytic infiltrate and an erosive or encrusted epidermis.5 Although direct immunofluorescence typically is negative, nonspecific findings can be seen, including apoptotic keratinocytes labeling with fibrinogen and C3, as well as scattered, clumped, IgM-positive cytoid bodies present at the dermal-epidermal junction (DEJ).6 Biopsies also have shown scattered, clumped, IgM-positive cytoid bodies present at the DEJ.5

Psoriasis is a chronic relapsing papulosquamous disorder characterized by scaly erythematous plaques often overlying the extensor surfaces of the extremities. Histopathology shows a psoriasiform pattern of inflammation with thinning of the suprapapillary plates and elongation of the rete ridges. Further diagnostic clues of psoriasis include regular acanthosis, characteristic Munro microabscesses with neutrophils in a hyperkeratotic stratum corneum (Figure 1), hypogranulosis, and neutrophilic spongiform pustules of Kogoj in the stratum spinosum. Generally, there is a lack of the epidermal necrosis seen with NME.7,8

Lichen simplex chronicus manifests as pruritic, often hyperpigmented, well-defined, lichenified plaques with excoriation following repetitive mechanical trauma, commonly on the lower lateral legs, posterior neck, and flexural areas.9 The histologic landscape is marked by well-developed lesions evolving to show compact orthokeratosis, hypergranulosis, irregularly elongated rete ridges (ie, irregular acanthosis), and papillary dermal fibrosis with vertical streaking of collagen (Figure 2).9,10

Subacute cutaneous lupus erythematosus (SCLE) is recognized clinically by scaly/psoriasiform and annular lesions with mild or absent systemic involvement. Common histopathologic findings include epidermal atrophy, vacuolar interface dermatitis with hydropic degeneration of the basal layer, a subepidermal lymphocytic infiltrate, and a periadnexal and perivascular infiltrate (Figure 3).11 Upper dermal edema, spotty necrosis of individual cells in the epidermis, dermal-epidermal separation caused by prominent basal cell degeneration, and accumulation of acid mucopolysaccharides (mucin) are other histologic features associated with SCLE.12,13

fruprububiuubrodiwrasavothihapulauedrotojafredisugiphavipranomuciswiprowowachochemuwadrokespimusotruchipuspistajesibaspuluspilivashaspesheciwrasteswedredilurabeprobaphachubishemislimumoclo
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Psoriasis%20shows%20hyperkeratosis%20with%20neutrophils%20in%20the%20stratum%20corneum%20on%20histopathology%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%3C%2Fp%3E

mecrifrigugadrijuwrabujudehauilokowutruvidrubuspebaslima
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Lichen%20simplex%20chronicus%20shows%20a%20compact%20stratum%20corneum%2C%20irregular%20acanthosis%2C%20and%20papillary%20dermal%20fibrosis%20on%20biopsy%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9710).%3C%2Fp%3E

The immunofluorescence pattern in SCLE features dustlike particles of IgG deposition in the epidermis, subepidermal region, and dermal cellular infiltrate. Lesions also may have granular deposition of immunoreactions at the DEJ.11,13

racruhecresocosisletribruvesteprospiphenukogitaclerouasloslojibem
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Subacute%20cutaneous%20lupus%20erythematosus%20shows%20vacuolar%20interface%20dermatitis%20with%20epidermal%20atrophy%2C%20subepidermal%20lymphocytes%2C%20and%20perivascular%20inflammation%20on%20biopsy%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9710).%3C%2Fp%3E

cutrewristicluu
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Drug%20reaction%20with%20eosinophilia%20and%20systemic%20symptoms%20(DRESS)%20syndrome%20shows%20spongiosis%20with%20a%20perivascular%20infiltrate%20on%20biopsy%3B%20eosinophils%20are%20variably%20observed%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9710).%3C%2Fp%3E

The manifestation of drug reaction with eosinophilia and systemic symptoms (DRESS) syndrome (also known as drug-induced hypersensitivity syndrome) is variable, with a morbilliform rash that spreads from the face to the entire body, urticaria, atypical target lesions, purpuriform lesions, lymphadenopathy, and exfoliative dermatitis.14 The nonspecific morphologic features of DRESS syndrome lesions are associated with variable histologic features, which include focal interface changes with vacuolar alteration of the basal layer; atypical lymphocytes with hyperchromic nuclei; and a superficial, inconsistently dense, perivascular lymphocytic infiltrate. Other relatively common histopathologic patterns include an upper dermis with dilated blood vessels, spongiosis with exocytosis of lymphocytes (Figure 4), and necrotic keratinocytes. Although peripheral eosinophilia is an important diagnostic criterion and is observed consistently, eosinophils are variably present on skin biopsy.15,16 Given the histopathologic variability and nonspecific findings, clinical correlation is required when diagnosing DRESS syndrome.

The Diagnosis: Necrolytic Migratory Erythema

Necrolytic migratory erythema (NME) is a waxing and waning rash associated with rare pancreatic neuroendocrine tumors called glucagonomas. It is characterized by pruritic and painful, well-demarcated, erythematous plaques that manifest in the intertriginous areas and on the perineum and buttocks.1 Due to the evolving nature of the rash, the histopathologic findings in NME vary depending on the stage of the cutaneous lesions at the time of biopsy.2 Multiple dyskeratotic keratinocytes spanning all epidermal layers may be a diagnostic clue in early lesions of NME.3 Typical features of longstanding lesions include confluent parakeratosis, psoriasiform hyperplasia with mild or absent spongiosis, and upper epidermal necrosis with keratinocyte vacuolization and pallor.4 Morphologic features that are present prior to the development of epidermal vacuolation and necrosis frequently are misattributed to psoriasis or eczema. Long-standing lesions also may develop a neutrophilic infiltrate with subcorneal and intraepidermal pustules.2 Other common features include a discrete perivascular lymphocytic infiltrate and an erosive or encrusted epidermis.5 Although direct immunofluorescence typically is negative, nonspecific findings can be seen, including apoptotic keratinocytes labeling with fibrinogen and C3, as well as scattered, clumped, IgM-positive cytoid bodies present at the dermal-epidermal junction (DEJ).6 Biopsies also have shown scattered, clumped, IgM-positive cytoid bodies present at the DEJ.5

Psoriasis is a chronic relapsing papulosquamous disorder characterized by scaly erythematous plaques often overlying the extensor surfaces of the extremities. Histopathology shows a psoriasiform pattern of inflammation with thinning of the suprapapillary plates and elongation of the rete ridges. Further diagnostic clues of psoriasis include regular acanthosis, characteristic Munro microabscesses with neutrophils in a hyperkeratotic stratum corneum (Figure 1), hypogranulosis, and neutrophilic spongiform pustules of Kogoj in the stratum spinosum. Generally, there is a lack of the epidermal necrosis seen with NME.7,8

Lichen simplex chronicus manifests as pruritic, often hyperpigmented, well-defined, lichenified plaques with excoriation following repetitive mechanical trauma, commonly on the lower lateral legs, posterior neck, and flexural areas.9 The histologic landscape is marked by well-developed lesions evolving to show compact orthokeratosis, hypergranulosis, irregularly elongated rete ridges (ie, irregular acanthosis), and papillary dermal fibrosis with vertical streaking of collagen (Figure 2).9,10

Subacute cutaneous lupus erythematosus (SCLE) is recognized clinically by scaly/psoriasiform and annular lesions with mild or absent systemic involvement. Common histopathologic findings include epidermal atrophy, vacuolar interface dermatitis with hydropic degeneration of the basal layer, a subepidermal lymphocytic infiltrate, and a periadnexal and perivascular infiltrate (Figure 3).11 Upper dermal edema, spotty necrosis of individual cells in the epidermis, dermal-epidermal separation caused by prominent basal cell degeneration, and accumulation of acid mucopolysaccharides (mucin) are other histologic features associated with SCLE.12,13

fruprububiuubrodiwrasavothihapulauedrotojafredisugiphavipranomuciswiprowowachochemuwadrokespimusotruchipuspistajesibaspuluspilivashaspesheciwrasteswedredilurabeprobaphachubishemislimumoclo
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Psoriasis%20shows%20hyperkeratosis%20with%20neutrophils%20in%20the%20stratum%20corneum%20on%20histopathology%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%3C%2Fp%3E

mecrifrigugadrijuwrabujudehauilokowutruvidrubuspebaslima
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Lichen%20simplex%20chronicus%20shows%20a%20compact%20stratum%20corneum%2C%20irregular%20acanthosis%2C%20and%20papillary%20dermal%20fibrosis%20on%20biopsy%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9710).%3C%2Fp%3E

The immunofluorescence pattern in SCLE features dustlike particles of IgG deposition in the epidermis, subepidermal region, and dermal cellular infiltrate. Lesions also may have granular deposition of immunoreactions at the DEJ.11,13

racruhecresocosisletribruvesteprospiphenukogitaclerouasloslojibem
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Subacute%20cutaneous%20lupus%20erythematosus%20shows%20vacuolar%20interface%20dermatitis%20with%20epidermal%20atrophy%2C%20subepidermal%20lymphocytes%2C%20and%20perivascular%20inflammation%20on%20biopsy%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9710).%3C%2Fp%3E

cutrewristicluu
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Drug%20reaction%20with%20eosinophilia%20and%20systemic%20symptoms%20(DRESS)%20syndrome%20shows%20spongiosis%20with%20a%20perivascular%20infiltrate%20on%20biopsy%3B%20eosinophils%20are%20variably%20observed%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9710).%3C%2Fp%3E

The manifestation of drug reaction with eosinophilia and systemic symptoms (DRESS) syndrome (also known as drug-induced hypersensitivity syndrome) is variable, with a morbilliform rash that spreads from the face to the entire body, urticaria, atypical target lesions, purpuriform lesions, lymphadenopathy, and exfoliative dermatitis.14 The nonspecific morphologic features of DRESS syndrome lesions are associated with variable histologic features, which include focal interface changes with vacuolar alteration of the basal layer; atypical lymphocytes with hyperchromic nuclei; and a superficial, inconsistently dense, perivascular lymphocytic infiltrate. Other relatively common histopathologic patterns include an upper dermis with dilated blood vessels, spongiosis with exocytosis of lymphocytes (Figure 4), and necrotic keratinocytes. Although peripheral eosinophilia is an important diagnostic criterion and is observed consistently, eosinophils are variably present on skin biopsy.15,16 Given the histopathologic variability and nonspecific findings, clinical correlation is required when diagnosing DRESS syndrome.

References
  1. Halvorson SA, Gilbert E, Hopkins RS, et al. Putting the pieces together: necrolytic migratory erythema and the glucagonoma syndrome. J Gen Intern Med. 2013;28:1525-1529. doi:10.1007 /s11606-013-2490-5
  2. Toberer F, Hartschuh W, Wiedemeyer K. Glucagonoma-associated necrolytic migratory erythema: the broad spectrum of the clinical and histopathological findings and clues to the diagnosis. Am J Dermatopathol. 2019;41:E29-E32. doi:10.1097DAD .0000000000001219
  3. Hunt SJ, Narus VT, Abell E. Necrolytic migratory erythema: dyskeratotic dermatitis, a clue to early diagnosis. J Am Acad Dermatol. 1991; 24:473-477. doi:10.1016/0190-9622(91)70076-e
  4. van Beek AP, de Haas ER, van Vloten WA, et al. The glucagonoma syndrome and necrolytic migratory erythema: a clinical review. Eur J Endocrinol. 2004;151:531-537. doi:10.1530/eje.0.1510531
  5. Pujol RM, Wang C-Y E, el-Azhary RA, et al. Necrolytic migratory erythema: clinicopathologic study of 13 cases. Int J Dermatol. 2004;43:12- 18. doi:10.1111/j.1365-4632.2004.01844.x
  6. Johnson SM, Smoller BR, Lamps LW, et al. Necrolytic migratory erythema as the only presenting sign of a glucagonoma. J Am Acad Dermatol. 2003;49:325-328. doi:10.1067/s0190-9622(02)61774-8
  7. De Rosa G, Mignogna C. The histopathology of psoriasis. Reumatismo. 2007;59(suppl 1):46-48. doi:10.4081/reumatismo.2007.1s.46
  8. Kimmel GW, Lebwohl M. Psoriasis: overview and diagnosis. In: Bhutani T, Liao W, Nakamura M, eds. Evidence-Based Psoriasis. Springer; 2018:1-16. doi:10.1007/978-3-319-90107-7_1
  9. Balan R, Grigoras¸ A, Popovici D, et al. The histopathological landscape of the major psoriasiform dermatoses. Arch Clin Cases. 2021;6:59-68. doi:10.22551/2019.24.0603.10155
  10. O’Keefe RJ, Scurry JP, Dennerstein G, et al. Audit of 114 nonneoplastic vulvar biopsies. Br J Obstet Gynaecol. 1995;102:780-786. doi:10.1111/j.1471-0528.1995.tb10842.x
  11. Parodi A, Caproni M, Cardinali C, et al P. Clinical, histological and immunopathological features of 58 patients with subacute cutaneous lupus erythematosus. Dermatology. 2000;200:6-10. doi:10.1159/000018307
  12. Lyon CC, Blewitt R, Harrison PV. Subacute cutaneous lupus erythematosus: two cases of delayed diagnosis. Acta Derm Venereol. 1998;78:57-59. doi:10.1080/00015559850135869
  13. David-Bajar KM. Subacute cutaneous lupus erythematosus. J Invest Dermatol. 1993;100:2S-8S. doi:10.1111/1523-1747.ep12355164
  14. Paulmann M, Mockenhaupt M. Severe drug-induced skin reactions: clinical features, diagnosis, etiology, and therapy. J Dtsch Dermatol Ges. 2015;13:625-643. doi:10.1111/ddg.12747
  15. Borroni G, Torti S, Pezzini C, et al. Histopathologic spectrum of drug reaction with eosinophilia and systemic symptoms (DRESS): a diagnosis that needs clinico-pathological correlation. G Ital Dermatol Venereol. 2014;149:291-300.
  16. Ortonne N, Valeyrie-Allanore L, Bastuji-Garin S, et al. Histopathology of drug rash with eosinophilia and systemic symptoms syndrome: a morphological and phenotypical study. Br J Dermatol. 2015;173:50-58. doi:10.1111/bjd.13683
References
  1. Halvorson SA, Gilbert E, Hopkins RS, et al. Putting the pieces together: necrolytic migratory erythema and the glucagonoma syndrome. J Gen Intern Med. 2013;28:1525-1529. doi:10.1007 /s11606-013-2490-5
  2. Toberer F, Hartschuh W, Wiedemeyer K. Glucagonoma-associated necrolytic migratory erythema: the broad spectrum of the clinical and histopathological findings and clues to the diagnosis. Am J Dermatopathol. 2019;41:E29-E32. doi:10.1097DAD .0000000000001219
  3. Hunt SJ, Narus VT, Abell E. Necrolytic migratory erythema: dyskeratotic dermatitis, a clue to early diagnosis. J Am Acad Dermatol. 1991; 24:473-477. doi:10.1016/0190-9622(91)70076-e
  4. van Beek AP, de Haas ER, van Vloten WA, et al. The glucagonoma syndrome and necrolytic migratory erythema: a clinical review. Eur J Endocrinol. 2004;151:531-537. doi:10.1530/eje.0.1510531
  5. Pujol RM, Wang C-Y E, el-Azhary RA, et al. Necrolytic migratory erythema: clinicopathologic study of 13 cases. Int J Dermatol. 2004;43:12- 18. doi:10.1111/j.1365-4632.2004.01844.x
  6. Johnson SM, Smoller BR, Lamps LW, et al. Necrolytic migratory erythema as the only presenting sign of a glucagonoma. J Am Acad Dermatol. 2003;49:325-328. doi:10.1067/s0190-9622(02)61774-8
  7. De Rosa G, Mignogna C. The histopathology of psoriasis. Reumatismo. 2007;59(suppl 1):46-48. doi:10.4081/reumatismo.2007.1s.46
  8. Kimmel GW, Lebwohl M. Psoriasis: overview and diagnosis. In: Bhutani T, Liao W, Nakamura M, eds. Evidence-Based Psoriasis. Springer; 2018:1-16. doi:10.1007/978-3-319-90107-7_1
  9. Balan R, Grigoras¸ A, Popovici D, et al. The histopathological landscape of the major psoriasiform dermatoses. Arch Clin Cases. 2021;6:59-68. doi:10.22551/2019.24.0603.10155
  10. O’Keefe RJ, Scurry JP, Dennerstein G, et al. Audit of 114 nonneoplastic vulvar biopsies. Br J Obstet Gynaecol. 1995;102:780-786. doi:10.1111/j.1471-0528.1995.tb10842.x
  11. Parodi A, Caproni M, Cardinali C, et al P. Clinical, histological and immunopathological features of 58 patients with subacute cutaneous lupus erythematosus. Dermatology. 2000;200:6-10. doi:10.1159/000018307
  12. Lyon CC, Blewitt R, Harrison PV. Subacute cutaneous lupus erythematosus: two cases of delayed diagnosis. Acta Derm Venereol. 1998;78:57-59. doi:10.1080/00015559850135869
  13. David-Bajar KM. Subacute cutaneous lupus erythematosus. J Invest Dermatol. 1993;100:2S-8S. doi:10.1111/1523-1747.ep12355164
  14. Paulmann M, Mockenhaupt M. Severe drug-induced skin reactions: clinical features, diagnosis, etiology, and therapy. J Dtsch Dermatol Ges. 2015;13:625-643. doi:10.1111/ddg.12747
  15. Borroni G, Torti S, Pezzini C, et al. Histopathologic spectrum of drug reaction with eosinophilia and systemic symptoms (DRESS): a diagnosis that needs clinico-pathological correlation. G Ital Dermatol Venereol. 2014;149:291-300.
  16. Ortonne N, Valeyrie-Allanore L, Bastuji-Garin S, et al. Histopathology of drug rash with eosinophilia and systemic symptoms syndrome: a morphological and phenotypical study. Br J Dermatol. 2015;173:50-58. doi:10.1111/bjd.13683
Issue
Cutis - 113(6)
Issue
Cutis - 113(6)
Page Number
234,258-259
Page Number
234,258-259
Publications
Publications
Topics
Article Type
Display Headline
Erythematous Flaky Rash on the Toe
Display Headline
Erythematous Flaky Rash on the Toe
Sections
Questionnaire Body

A 62-year-old man presented with an erythematous flaky rash associated with burning pain on the right medial second toe that persisted for several months. Prior treatment with econazole, ciclopirox, and oral amoxicillin had failed. A shave biopsy was performed.

friprunethecrojoswosucliphatreuenuvomophedalitonispifraspiwrajifretrapusliphubrouojechishimulafrolilispashuvekeg
%3Cp%3EH%26amp%3BE%2C%20original%20magnifications%20%C3%9720.%3C%2Fp%3E

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 06/11/2024 - 11:00
Un-Gate On Date
Tue, 06/11/2024 - 11:00
Use ProPublica
CFC Schedule Remove Status
Tue, 06/11/2024 - 11:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Disable zoom
Off

Need a Wood Lamp Alternative? Grab Your Smartphone

Article Type
Changed
Fri, 06/14/2024 - 12:37
Display Headline
Need a Wood Lamp Alternative? Grab Your Smartphone

Practice Gap

The Wood lamp commonly is used as a diagnostic tool for pigmentary skin conditions (eg, vitiligo) or skin conditions that exhibit fluorescence (eg, erythrasma).1 Recently, its diagnostic efficacy has extended to scabies, in which it unveils a distinctive wavy, bluish-white, linear fluorescence upon illumination.2

Functionally, the Wood lamp operates by subjecting phosphors to UV light within the wavelength range of 320 to 400 nm, inducing fluorescence in substances such as collagen and elastin. In the context of vitiligo, this process manifests as a preferential chalk white fluorescence in areas lacking melanin.1

Despite its demonstrated effectiveness, the Wood lamp is not without limitations. It comes with a notable financial investment ranging from $70 to $500, requires periodic maintenance such as light bulb replacements, and can be unwieldy.3 Furthermore, its reliance on a power source poses a challenge in settings where immediate access to convenient power outlets is limited, such as inpatient and rural dermatology clinics. These limitations underscore the need for alternative solutions and innovations to address challenges and ensure accessibility in diverse health care environments.

The Tools

Free smartphone applications (apps), such as Ultraviolet Light-UV Lamp by AppBrain or Blacklight UV Light Simulator by That Smile, can simulate UV light and functionally serve as a Wood lamp.

The Technique

UV light apps use LED or organic LED screen pixels to emit a blue light equivalent at 467 nm.4 Although these apps are not designed specifically for dermatologic uses, they are mostly free, widely available for Android and iPhone users, and portable. Importantly, they can demonstrate good performance in visualizing vitiligo, as shown in Figure 1—albeit perhaps not reaching the same level as the Wood lamp (Figure 2).

ruvujepachanunaspukustunisloseslopripraspahelothulathurisikibralutauipheswapipheuispovestitredrachoclechujefrachiwibamuwrobepenithusabrovosuwr
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20and%20B%2C%20Depigmented%20patches%20of%20vitiligo%20on%20the%20skin%20are%20visualized%20with%20a%20free%20UV%20light%20smartphone%20application%2C%20respectively.%3C%2Fp%3E

trapreuechikibrefrithashaslotadrouushowepafraceshilesugastedrigicloswaphewuspasperecledosteratrunidrucludamagubupricluswiswehathiswogasephobrejuspedrijahastothopresosluvurogawafribra
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%20and%20B%2C%20The%20same%20depigmented%20patches%20of%20vitiligo%20are%20visualized%20with%20a%20free%20UV%20light%20smartphone%20application%20vs%20a%20Wood%20lamp%2C%20respectively.%3C%2Fp%3E

Because these UV light apps are not regulated and their efficacy for medical use has not been firmly established, the Wood lamp remains the gold standard. Therefore, we propose the use of UV light apps in situations when a Wood lamp is not available or convenient, such as in rural, inpatient, or international health care settings.

Practice Implications

Exploring and adopting these free alternatives can contribute to improved accessibility and diagnostic capabilities in diverse health care environments, particularly for communities facing financial constraints. Continued research and validation of these apps in clinical settings will be essential to establish their reliability and effectiveness in enhancing diagnostic practices.

References
  1. Dyer JM, Foy VM. Revealing the unseen: a review of Wood’s lamp in dermatology. J Clin Aesthet Dermatol. 2022;15:25-30.
  2. Scanni G. Facilitations in the clinical diagnosis of human scabies through the use of ultraviolet light (UV-scab scanning): a case-series study. Trop Med Infect Dis. 2022;7:422. doi:10.3390/tropicalmed7120422
  3. USA Medical and Surgical Supplies. Top 9 medical diagnostic applications for a Woods lamp. February 26, 2019. Accessed May 20, 2024.
  4. Huang Y, Hsiang E-L, Deng M-Y, et al. Mini-led, micro-led and OLED displays: present status and future perspectives. Light Sci Appl. 2020;9:105. doi:10.1038/s41377-020-0341-9
Article PDF
Author and Disclosure Information

 

Ruth Zagales is from the Indiana University School of Medicine, Indianapolis. Dr. Korman is from the Department of Dermatology, The Ohio State University Wexner Medical Center, Columbus.

The authors report no conflict of interest.

Correspondence: Abraham M. Korman, MD, 540 Office Center Pl, Ste 240, Columbus, OH 43230 (Abraham.Korman@osumc.edu).

Cutis. 2024 June;113(6):271-272. doi:10.12788/cutis.1026

Issue
Cutis - 113(6)
Publications
Topics
Page Number
271-272
Sections
Author and Disclosure Information

 

Ruth Zagales is from the Indiana University School of Medicine, Indianapolis. Dr. Korman is from the Department of Dermatology, The Ohio State University Wexner Medical Center, Columbus.

The authors report no conflict of interest.

Correspondence: Abraham M. Korman, MD, 540 Office Center Pl, Ste 240, Columbus, OH 43230 (Abraham.Korman@osumc.edu).

Cutis. 2024 June;113(6):271-272. doi:10.12788/cutis.1026

Author and Disclosure Information

 

Ruth Zagales is from the Indiana University School of Medicine, Indianapolis. Dr. Korman is from the Department of Dermatology, The Ohio State University Wexner Medical Center, Columbus.

The authors report no conflict of interest.

Correspondence: Abraham M. Korman, MD, 540 Office Center Pl, Ste 240, Columbus, OH 43230 (Abraham.Korman@osumc.edu).

Cutis. 2024 June;113(6):271-272. doi:10.12788/cutis.1026

Article PDF
Article PDF

Practice Gap

The Wood lamp commonly is used as a diagnostic tool for pigmentary skin conditions (eg, vitiligo) or skin conditions that exhibit fluorescence (eg, erythrasma).1 Recently, its diagnostic efficacy has extended to scabies, in which it unveils a distinctive wavy, bluish-white, linear fluorescence upon illumination.2

Functionally, the Wood lamp operates by subjecting phosphors to UV light within the wavelength range of 320 to 400 nm, inducing fluorescence in substances such as collagen and elastin. In the context of vitiligo, this process manifests as a preferential chalk white fluorescence in areas lacking melanin.1

Despite its demonstrated effectiveness, the Wood lamp is not without limitations. It comes with a notable financial investment ranging from $70 to $500, requires periodic maintenance such as light bulb replacements, and can be unwieldy.3 Furthermore, its reliance on a power source poses a challenge in settings where immediate access to convenient power outlets is limited, such as inpatient and rural dermatology clinics. These limitations underscore the need for alternative solutions and innovations to address challenges and ensure accessibility in diverse health care environments.

The Tools

Free smartphone applications (apps), such as Ultraviolet Light-UV Lamp by AppBrain or Blacklight UV Light Simulator by That Smile, can simulate UV light and functionally serve as a Wood lamp.

The Technique

UV light apps use LED or organic LED screen pixels to emit a blue light equivalent at 467 nm.4 Although these apps are not designed specifically for dermatologic uses, they are mostly free, widely available for Android and iPhone users, and portable. Importantly, they can demonstrate good performance in visualizing vitiligo, as shown in Figure 1—albeit perhaps not reaching the same level as the Wood lamp (Figure 2).

ruvujepachanunaspukustunisloseslopripraspahelothulathurisikibralutauipheswapipheuispovestitredrachoclechujefrachiwibamuwrobepenithusabrovosuwr
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20and%20B%2C%20Depigmented%20patches%20of%20vitiligo%20on%20the%20skin%20are%20visualized%20with%20a%20free%20UV%20light%20smartphone%20application%2C%20respectively.%3C%2Fp%3E

trapreuechikibrefrithashaslotadrouushowepafraceshilesugastedrigicloswaphewuspasperecledosteratrunidrucludamagubupricluswiswehathiswogasephobrejuspedrijahastothopresosluvurogawafribra
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%20and%20B%2C%20The%20same%20depigmented%20patches%20of%20vitiligo%20are%20visualized%20with%20a%20free%20UV%20light%20smartphone%20application%20vs%20a%20Wood%20lamp%2C%20respectively.%3C%2Fp%3E

Because these UV light apps are not regulated and their efficacy for medical use has not been firmly established, the Wood lamp remains the gold standard. Therefore, we propose the use of UV light apps in situations when a Wood lamp is not available or convenient, such as in rural, inpatient, or international health care settings.

Practice Implications

Exploring and adopting these free alternatives can contribute to improved accessibility and diagnostic capabilities in diverse health care environments, particularly for communities facing financial constraints. Continued research and validation of these apps in clinical settings will be essential to establish their reliability and effectiveness in enhancing diagnostic practices.

Practice Gap

The Wood lamp commonly is used as a diagnostic tool for pigmentary skin conditions (eg, vitiligo) or skin conditions that exhibit fluorescence (eg, erythrasma).1 Recently, its diagnostic efficacy has extended to scabies, in which it unveils a distinctive wavy, bluish-white, linear fluorescence upon illumination.2

Functionally, the Wood lamp operates by subjecting phosphors to UV light within the wavelength range of 320 to 400 nm, inducing fluorescence in substances such as collagen and elastin. In the context of vitiligo, this process manifests as a preferential chalk white fluorescence in areas lacking melanin.1

Despite its demonstrated effectiveness, the Wood lamp is not without limitations. It comes with a notable financial investment ranging from $70 to $500, requires periodic maintenance such as light bulb replacements, and can be unwieldy.3 Furthermore, its reliance on a power source poses a challenge in settings where immediate access to convenient power outlets is limited, such as inpatient and rural dermatology clinics. These limitations underscore the need for alternative solutions and innovations to address challenges and ensure accessibility in diverse health care environments.

The Tools

Free smartphone applications (apps), such as Ultraviolet Light-UV Lamp by AppBrain or Blacklight UV Light Simulator by That Smile, can simulate UV light and functionally serve as a Wood lamp.

The Technique

UV light apps use LED or organic LED screen pixels to emit a blue light equivalent at 467 nm.4 Although these apps are not designed specifically for dermatologic uses, they are mostly free, widely available for Android and iPhone users, and portable. Importantly, they can demonstrate good performance in visualizing vitiligo, as shown in Figure 1—albeit perhaps not reaching the same level as the Wood lamp (Figure 2).

ruvujepachanunaspukustunisloseslopripraspahelothulathurisikibralutauipheswapipheuispovestitredrachoclechujefrachiwibamuwrobepenithusabrovosuwr
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20and%20B%2C%20Depigmented%20patches%20of%20vitiligo%20on%20the%20skin%20are%20visualized%20with%20a%20free%20UV%20light%20smartphone%20application%2C%20respectively.%3C%2Fp%3E

trapreuechikibrefrithashaslotadrouushowepafraceshilesugastedrigicloswaphewuspasperecledosteratrunidrucludamagubupricluswiswehathiswogasephobrejuspedrijahastothopresosluvurogawafribra
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%20and%20B%2C%20The%20same%20depigmented%20patches%20of%20vitiligo%20are%20visualized%20with%20a%20free%20UV%20light%20smartphone%20application%20vs%20a%20Wood%20lamp%2C%20respectively.%3C%2Fp%3E

Because these UV light apps are not regulated and their efficacy for medical use has not been firmly established, the Wood lamp remains the gold standard. Therefore, we propose the use of UV light apps in situations when a Wood lamp is not available or convenient, such as in rural, inpatient, or international health care settings.

Practice Implications

Exploring and adopting these free alternatives can contribute to improved accessibility and diagnostic capabilities in diverse health care environments, particularly for communities facing financial constraints. Continued research and validation of these apps in clinical settings will be essential to establish their reliability and effectiveness in enhancing diagnostic practices.

References
  1. Dyer JM, Foy VM. Revealing the unseen: a review of Wood’s lamp in dermatology. J Clin Aesthet Dermatol. 2022;15:25-30.
  2. Scanni G. Facilitations in the clinical diagnosis of human scabies through the use of ultraviolet light (UV-scab scanning): a case-series study. Trop Med Infect Dis. 2022;7:422. doi:10.3390/tropicalmed7120422
  3. USA Medical and Surgical Supplies. Top 9 medical diagnostic applications for a Woods lamp. February 26, 2019. Accessed May 20, 2024.
  4. Huang Y, Hsiang E-L, Deng M-Y, et al. Mini-led, micro-led and OLED displays: present status and future perspectives. Light Sci Appl. 2020;9:105. doi:10.1038/s41377-020-0341-9
References
  1. Dyer JM, Foy VM. Revealing the unseen: a review of Wood’s lamp in dermatology. J Clin Aesthet Dermatol. 2022;15:25-30.
  2. Scanni G. Facilitations in the clinical diagnosis of human scabies through the use of ultraviolet light (UV-scab scanning): a case-series study. Trop Med Infect Dis. 2022;7:422. doi:10.3390/tropicalmed7120422
  3. USA Medical and Surgical Supplies. Top 9 medical diagnostic applications for a Woods lamp. February 26, 2019. Accessed May 20, 2024.
  4. Huang Y, Hsiang E-L, Deng M-Y, et al. Mini-led, micro-led and OLED displays: present status and future perspectives. Light Sci Appl. 2020;9:105. doi:10.1038/s41377-020-0341-9
Issue
Cutis - 113(6)
Issue
Cutis - 113(6)
Page Number
271-272
Page Number
271-272
Publications
Publications
Topics
Article Type
Display Headline
Need a Wood Lamp Alternative? Grab Your Smartphone
Display Headline
Need a Wood Lamp Alternative? Grab Your Smartphone
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Zagales</fileName> <TBEID>0C02F79E.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F79E</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Pearls</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240610T104052</firstPublished> <LastPublished>20240610T104052</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240610T104051</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Ruth Zagales, BS; Abraham M. Korman, MD</byline> <bylineText>Ruth Zagales, BS; Abraham M. Korman, MD</bylineText> <bylineFull>Ruth Zagales, BS; Abraham M. Korman, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>271-272</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>The Wood lamp commonly is used as a diagnostic tool for pigmentary skin conditions (eg, vitiligo) or skin conditions that exhibit fluorescence (eg, erythrasma).</metaDescription> <articlePDF>301784</articlePDF> <teaserImage/> <title>Need a Wood Lamp Alternative? Grab Your Smartphone</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>June</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>6</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>19497</CMSID> <CMSID>2159</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>June 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Practice Pearls | 19497<pubSubsection/></pubSection> </pubSections> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">39452</term> </sections> <topics> <term canonical="true">204</term> <term>276</term> <term>281</term> <term>66772</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/1800274f.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Need a Wood Lamp Alternative? Grab Your Smartphone</title> <deck/> </itemMeta> <itemContent> <p class="abstract">The Wood lamp is an effective diagnostic tool for skin conditions such as vitiligo, erythrasma, and scabies. However, it can be an expensive and cumbersome tool or may be unavailable in some settings. Smartphone applications that simulate UV light may be helpful in these instances.</p> <h3>Practice Gap</h3> <p>The Wood lamp commonly is used as a diagnostic tool for pigmentary skin conditions (eg, vitiligo) or skin conditions that exhibit fluorescence (eg, erythrasma).<sup>1</sup> Recently, its diagnostic efficacy has extended to scabies, in which it unveils a distinctive wavy, bluish-white, linear fluorescence upon illumination.<sup>2</sup> </p> <p>Functionally, the Wood lamp operates by subjecting phosphors to UV light within the wavelength range of 320 to 400 nm, inducing fluorescence in substances such as collagen and elastin. In the context of vitiligo, this process manifests as a preferential chalk white fluorescence in areas lacking melanin.<sup>1<br/><br/></sup>Despite its demonstrated effectiveness, the Wood lamp is not without limitations. It comes with a notable financial investment ranging from $70 to $500, requires periodic maintenance such as light bulb replacements, and can be unwieldy.<sup>3</sup> Furthermore, its reliance on a power source poses a challenge in settings where immediate access to convenient power outlets is limited, such as inpatient and rural dermatology clinics. These limitations underscore the need for alternative solutions and innovations to address challenges and ensure accessibility in diverse health care environments.</p> <h3>The Tools</h3> <p>Free smartphone applications (apps), such as Ultraviolet Light-UV Lamp by AppBrain or Blacklight UV Light Simulator by That Smile, can simulate UV light and functionally serve as a Wood lamp. </p> <h3>The Technique</h3> <p>UV light apps use LED or organic LED screen pixels to emit a blue light equivalent at 467 nm.<sup>4</sup> Although these apps are not designed specifically for dermatologic uses, they are mostly free, widely available for Android and iPhone users, and portable. Importantly, they can demonstrate good performance in visualizing vitiligo, as shown in Figure 1—albeit perhaps not reaching the same level as the Wood lamp (Figure 2). </p> <p>Because these UV light apps are not regulated and their efficacy for medical use has not been firmly established, the Wood lamp remains the gold standard. Therefore, we propose the use of UV light apps in situations when a Wood lamp is not available or convenient, such as in rural, inpatient, or international health care settings.</p> <h3>Practice Implications</h3> <p>Exploring and adopting these free alternatives can contribute to improved accessibility and diagnostic capabilities in diverse health care environments, particularly for communities facing financial constraints. Continued research and validation of these apps in clinical settings will be essential to establish their reliability and effectiveness in enhancing diagnostic practices.</p> <h2>References</h2> <p class="reference"> 1. Dyer JM, Foy VM. Revealing the unseen: a review of Wood’s lamp in dermatology. <i>J Clin Aesthet Dermatol</i>. 2022;15:25-30.<br/><br/> 2. Scanni G. Facilitations in the clinical diagnosis of human scabies through the use of ultraviolet light (UV-scab scanning): a case-series study. <i>Trop Med Infect Dis</i>. 2022;7:422. doi:10.3390/tropicalmed7120422 <br/><br/> 3. USA Medical and Surgical Supplies. Top 9 medical diagnostic applications for a Woods lamp. February 26, 2019. Accessed May 20, 2024. <br/><br/> 4. Huang Y, Hsiang E-L, Deng M-Y, et al. Mini-led, micro-led and OLED displays: present status and future perspectives. <i>Light Sci Appl</i>. 2020;9:105. doi:10.1038/s41377-020-0341-9 </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Ruth Zagales is from the Indiana University School of Medicine, Indianapolis. Dr. Korman is from the Department of Dermatology, The Ohio State University Wexner Medical Center, Columbus. </p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>Correspondence: Abraham M. Korman, MD, 540 Office Center Pl, Ste 240, Columbus, OH 43230 (Abraham.Korman@osumc.edu). <br/><br/><em>Cutis.</em> 2024 June;113(6):271-272. doi:10.12788/cutis.1026</p> </itemContent> </newsItem> </itemSet></root>
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
Image
Disable zoom
Off

Celiac Disease: Five Things to Know

Article Type
Changed
Fri, 06/07/2024 - 16:34

Celiac disease is a chronic, immune-mediated, systemic disorder caused by intolerance to gluten — a protein present in rye, barley, and wheat grains — that affects genetically predisposed individuals.

Due to its wide spectrum of clinical manifestations, celiac disease resembles a multisystemic disorder. Its most common gastrointestinal (GI) symptoms include chronic diarrhea, weight loss, and abdominal distention. However, celiac disease can also manifest in myriad extraintestinal symptoms, ranging from headache and fatigue to delayed puberty and psychiatric disorders, with differing presentations in children and adults.

To date, the only treatment is adopting a gluten-free diet (GFD). Although key to preventing persistent villous atrophy, the main cause of complications in celiac disease, lifelong adherence to GFD is challenging and may not resolve all clinical issues. These shortcomings have driven recent efforts to develop novel therapeutic options for patients with this disease.

Here are five things to know about celiac disease.
 

1. Rising Prevalence of Celiac Disease and Other Autoimmune Disorders Suggests Environmental Factors May Be at Play

Gluten was first identified as the cause of celiac disease in the 1950s. At that time, the condition was thought to be a relatively rare GI disease of childhood that primarily affected people of European descent, but it is now known to be a common disease affecting those of various ages, races, and ethnicities.

2018 meta-analysis found the pooled global prevalence of celiac disease was 1.4%. Incidence has increased by as much as 7.5% annually over the past several decades.

Increased awareness among clinicians and improved detection likely play a role in the trend. However, the growth in celiac disease is consistent with that seen for other autoimmune disorders, according to a 2024 update of evidence surrounding celiac disease. Shared environmental factors have been proposed as triggers for celiac disease and other autoimmune diseases and appear to be influencing their rise, the authors noted. These factors include migration and population growth, changing dietary patterns and food processing practices, and altered wheat consumption.
 

2. No-Biopsy Diagnosis Is Accepted for Children and Shows Promise for Adults

It is estimated that almost 60 million people worldwide have celiac disease, but most remain undiagnosed or misdiagnosed, or they experience significant diagnostic delays.

Prospective data indicate that children with first-degree relatives with celiac disease are at a significantly higher risk of developing the condition, which should prompt screening efforts in this population.

The 2023 updated guidelines from the American College of Gastroenterology (ACG) state that serology testing plays a central role in screening. This commonly involves serological testing for positive serological markers of the disease, including immunoglobulin A (IgA), anti-tissue transglutaminase IgA (tTG-IgA), anti-deamidated gliadin peptide, or endomysial antibodies.

To confirm diagnosis, clinicians have relied on intestinal biopsy since the late 1950s. The ACG still recommends esophagogastroduodenoscopy with multiple duodenal biopsies for confirmation of diagnosis in both children and adults with suspicion of celiac disease. However, recent years have seen a shift toward a no-biopsy approach.

For more than a decade in Europe, a no-biopsy approach has been established practice in pediatric patients, for whom the burden of obtaining a histological confirmation is understandably greater. Most guidelines now permit children to be diagnosed with celiac disease in the absence of a biopsy under specific circumstances (eg, characteristic symptoms of celiac disease and tTG-IgA levels > 10 times the upper limit of normal). The ACG guidelines state that “this approach is a reasonable alternative to the standard approach to a [celiac disease] diagnosis in selected children.”

The ACG does not recommend a no-biopsy approach in adults, noting that, in comparison with children, there is a relative lack of data indicating that serology is predictive in this population. However, it does recognize that physicians may encounter patients for whom a biopsy diagnosis may not be safe or practical. In such cases, an “after-the-fact” diagnosis of likely celiac disease can be given to symptomatic adult patients with a ≥ 10-fold elevation of tTG-IgA and a positive endomysial antibody in a second blood sample.

A 2024 meta-analysis of 18 studies involving over 12,103 adult patients from 15 countries concluded that a no-biopsy approach using tTG-IgA antibody levels ≥ 10 times the upper limit of normal was highly specific and predictive of celiac disease.
 

 

 

3. Celiac Disease Is Associated With Several Life-Threatening Conditions

Emerging data indicate that gastroenterologists should be vigilant in screening patients with celiac disease for several other GI conditions.

Inflammatory bowel disease and celiac disease have a strong bidirectional association, suggesting a possible genetic link between the conditions and indicating that physicians should consider the alternate diagnosis when symptoms persist after treatment.

Given the hypervigilance around food and diet inherent to celiac disease, patients are at an increased risk of developing avoidant/restrictive food intake disorder, according to a 2022 retrospective study.

In 2023, Italian investigators showed that children with celiac disease have an elevated prevalence of functional GI disorders even after adopting a GFD for a year, regardless of whether they consumed processed or natural foods. It was unclear whether this was due to a chronic inflammatory process or to nutritional factors.

Complications resulting from celiac disease are not limited to GI disorders. For a variety of underlying pathophysiological reasons, including intestinal permeability, hyposplenism, and malabsorption of nutrients, patients with celiac disease may be at a higher risk for non-GI conditions, such as osteopeniawomen’s health disorders (eg, ovarian failure, endometriosis, or pregnancy loss), juvenile idiopathic arthritis in children and rheumatoid arthritis in adultscertain forms of cancerinfectious diseases, and cardiomyopathy.
 

4. GFD Is the Only Treatment, but It’s Imperfect and Frustrating for Patients

GFD is the only treatment for celiac disease and must be adhered to without deviation throughout a patient’s life.

Maintaining unwavering adherence reaps considerable benefits: Improved clinical symptoms, robust mucosal healing, and normalization of serological markers. Yet it also takes a considerable toll on patients. Patients with celiac disease struggle with a host of negative physical, psychological, and social impacts. They also report a higher treatment burden than those with gastroesophageal reflux disease or hypertension, and comparable with end-stage renal disease.

GFD also poses financial challenges. Although the price of gluten-free products has decreased in recent years, they still cost significantly more than items with gluten.

Adherence to GFD does not always equate to complete mucosal recovery. While mucosal recovery is achieved in 95% of children within 2 years of the diet’s adoption, only 34% and 66% of adults obtain it within 2 and 5 years, respectively.

GFD may lead to nutrient imbalances because gluten-free foods are typically low in alimentary fiber, micronutrients (eg, vitamin D, vitamin B12, or folate), and minerals (eg, iron, zinc, magnesium, or calcium). With higher sugar and fat content, GFD may leave patients susceptible to unwanted weight gain.

The pervasiveness of gluten in the food production system makes the risk for cross-contamination high. Gluten is often found in both naturally gluten-free foods and products labeled as such. Gluten-sensing technologies, some of which can be used via smartphone apps, have been developed to help patients identify possible cross-contamination. However, the ACG guidelines recommend against the use of these technologies until there is sufficient evidence supporting their ability to improve adherence and clinical outcomes.
 

5. Novel Therapies for Celiac Disease Are in the Pipeline

The limitations of GFD as the standard treatment for celiac disease have led to an increased focus on developing novel therapeutic interventions. They can be sorted into five key categories: Modulation of the immunostimulatory effects of toxic gluten peptides, elimination of toxic gluten peptides before they reach the intestine, induction of gluten tolerance, modulation of intestinal permeability, and restoration of gut microbiota balance.

Three therapies designed to block antigen presentation by HLA-DQ2/8, the gene alleles that predispose people to celiac disease, show promise: TPM502, an agent that contains three gluten-specific antigenic peptides with overlapping T-cell epitopes for the HLA-DQ2.5 gene; KAN-101, designed to induce gluten tolerance by targeting receptors on the liver; and DONQ52, a multi-specific antibody that targets HLA-DQ2. The KAN-101 therapy received Fast Track designation by the US Food and Drug Administration in 2022.

These and several other agents in clinical and preclinical development are discussed in detail in a 2024 review article. Although no therapies have reached phase 3 testing, when they do, it will undoubtedly be welcomed by those with celiac disease.

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

Celiac disease is a chronic, immune-mediated, systemic disorder caused by intolerance to gluten — a protein present in rye, barley, and wheat grains — that affects genetically predisposed individuals.

Due to its wide spectrum of clinical manifestations, celiac disease resembles a multisystemic disorder. Its most common gastrointestinal (GI) symptoms include chronic diarrhea, weight loss, and abdominal distention. However, celiac disease can also manifest in myriad extraintestinal symptoms, ranging from headache and fatigue to delayed puberty and psychiatric disorders, with differing presentations in children and adults.

To date, the only treatment is adopting a gluten-free diet (GFD). Although key to preventing persistent villous atrophy, the main cause of complications in celiac disease, lifelong adherence to GFD is challenging and may not resolve all clinical issues. These shortcomings have driven recent efforts to develop novel therapeutic options for patients with this disease.

Here are five things to know about celiac disease.
 

1. Rising Prevalence of Celiac Disease and Other Autoimmune Disorders Suggests Environmental Factors May Be at Play

Gluten was first identified as the cause of celiac disease in the 1950s. At that time, the condition was thought to be a relatively rare GI disease of childhood that primarily affected people of European descent, but it is now known to be a common disease affecting those of various ages, races, and ethnicities.

2018 meta-analysis found the pooled global prevalence of celiac disease was 1.4%. Incidence has increased by as much as 7.5% annually over the past several decades.

Increased awareness among clinicians and improved detection likely play a role in the trend. However, the growth in celiac disease is consistent with that seen for other autoimmune disorders, according to a 2024 update of evidence surrounding celiac disease. Shared environmental factors have been proposed as triggers for celiac disease and other autoimmune diseases and appear to be influencing their rise, the authors noted. These factors include migration and population growth, changing dietary patterns and food processing practices, and altered wheat consumption.
 

2. No-Biopsy Diagnosis Is Accepted for Children and Shows Promise for Adults

It is estimated that almost 60 million people worldwide have celiac disease, but most remain undiagnosed or misdiagnosed, or they experience significant diagnostic delays.

Prospective data indicate that children with first-degree relatives with celiac disease are at a significantly higher risk of developing the condition, which should prompt screening efforts in this population.

The 2023 updated guidelines from the American College of Gastroenterology (ACG) state that serology testing plays a central role in screening. This commonly involves serological testing for positive serological markers of the disease, including immunoglobulin A (IgA), anti-tissue transglutaminase IgA (tTG-IgA), anti-deamidated gliadin peptide, or endomysial antibodies.

To confirm diagnosis, clinicians have relied on intestinal biopsy since the late 1950s. The ACG still recommends esophagogastroduodenoscopy with multiple duodenal biopsies for confirmation of diagnosis in both children and adults with suspicion of celiac disease. However, recent years have seen a shift toward a no-biopsy approach.

For more than a decade in Europe, a no-biopsy approach has been established practice in pediatric patients, for whom the burden of obtaining a histological confirmation is understandably greater. Most guidelines now permit children to be diagnosed with celiac disease in the absence of a biopsy under specific circumstances (eg, characteristic symptoms of celiac disease and tTG-IgA levels > 10 times the upper limit of normal). The ACG guidelines state that “this approach is a reasonable alternative to the standard approach to a [celiac disease] diagnosis in selected children.”

The ACG does not recommend a no-biopsy approach in adults, noting that, in comparison with children, there is a relative lack of data indicating that serology is predictive in this population. However, it does recognize that physicians may encounter patients for whom a biopsy diagnosis may not be safe or practical. In such cases, an “after-the-fact” diagnosis of likely celiac disease can be given to symptomatic adult patients with a ≥ 10-fold elevation of tTG-IgA and a positive endomysial antibody in a second blood sample.

A 2024 meta-analysis of 18 studies involving over 12,103 adult patients from 15 countries concluded that a no-biopsy approach using tTG-IgA antibody levels ≥ 10 times the upper limit of normal was highly specific and predictive of celiac disease.
 

 

 

3. Celiac Disease Is Associated With Several Life-Threatening Conditions

Emerging data indicate that gastroenterologists should be vigilant in screening patients with celiac disease for several other GI conditions.

Inflammatory bowel disease and celiac disease have a strong bidirectional association, suggesting a possible genetic link between the conditions and indicating that physicians should consider the alternate diagnosis when symptoms persist after treatment.

Given the hypervigilance around food and diet inherent to celiac disease, patients are at an increased risk of developing avoidant/restrictive food intake disorder, according to a 2022 retrospective study.

In 2023, Italian investigators showed that children with celiac disease have an elevated prevalence of functional GI disorders even after adopting a GFD for a year, regardless of whether they consumed processed or natural foods. It was unclear whether this was due to a chronic inflammatory process or to nutritional factors.

Complications resulting from celiac disease are not limited to GI disorders. For a variety of underlying pathophysiological reasons, including intestinal permeability, hyposplenism, and malabsorption of nutrients, patients with celiac disease may be at a higher risk for non-GI conditions, such as osteopeniawomen’s health disorders (eg, ovarian failure, endometriosis, or pregnancy loss), juvenile idiopathic arthritis in children and rheumatoid arthritis in adultscertain forms of cancerinfectious diseases, and cardiomyopathy.
 

4. GFD Is the Only Treatment, but It’s Imperfect and Frustrating for Patients

GFD is the only treatment for celiac disease and must be adhered to without deviation throughout a patient’s life.

Maintaining unwavering adherence reaps considerable benefits: Improved clinical symptoms, robust mucosal healing, and normalization of serological markers. Yet it also takes a considerable toll on patients. Patients with celiac disease struggle with a host of negative physical, psychological, and social impacts. They also report a higher treatment burden than those with gastroesophageal reflux disease or hypertension, and comparable with end-stage renal disease.

GFD also poses financial challenges. Although the price of gluten-free products has decreased in recent years, they still cost significantly more than items with gluten.

Adherence to GFD does not always equate to complete mucosal recovery. While mucosal recovery is achieved in 95% of children within 2 years of the diet’s adoption, only 34% and 66% of adults obtain it within 2 and 5 years, respectively.

GFD may lead to nutrient imbalances because gluten-free foods are typically low in alimentary fiber, micronutrients (eg, vitamin D, vitamin B12, or folate), and minerals (eg, iron, zinc, magnesium, or calcium). With higher sugar and fat content, GFD may leave patients susceptible to unwanted weight gain.

The pervasiveness of gluten in the food production system makes the risk for cross-contamination high. Gluten is often found in both naturally gluten-free foods and products labeled as such. Gluten-sensing technologies, some of which can be used via smartphone apps, have been developed to help patients identify possible cross-contamination. However, the ACG guidelines recommend against the use of these technologies until there is sufficient evidence supporting their ability to improve adherence and clinical outcomes.
 

5. Novel Therapies for Celiac Disease Are in the Pipeline

The limitations of GFD as the standard treatment for celiac disease have led to an increased focus on developing novel therapeutic interventions. They can be sorted into five key categories: Modulation of the immunostimulatory effects of toxic gluten peptides, elimination of toxic gluten peptides before they reach the intestine, induction of gluten tolerance, modulation of intestinal permeability, and restoration of gut microbiota balance.

Three therapies designed to block antigen presentation by HLA-DQ2/8, the gene alleles that predispose people to celiac disease, show promise: TPM502, an agent that contains three gluten-specific antigenic peptides with overlapping T-cell epitopes for the HLA-DQ2.5 gene; KAN-101, designed to induce gluten tolerance by targeting receptors on the liver; and DONQ52, a multi-specific antibody that targets HLA-DQ2. The KAN-101 therapy received Fast Track designation by the US Food and Drug Administration in 2022.

These and several other agents in clinical and preclinical development are discussed in detail in a 2024 review article. Although no therapies have reached phase 3 testing, when they do, it will undoubtedly be welcomed by those with celiac disease.

A version of this article first appeared on Medscape.com.

Celiac disease is a chronic, immune-mediated, systemic disorder caused by intolerance to gluten — a protein present in rye, barley, and wheat grains — that affects genetically predisposed individuals.

Due to its wide spectrum of clinical manifestations, celiac disease resembles a multisystemic disorder. Its most common gastrointestinal (GI) symptoms include chronic diarrhea, weight loss, and abdominal distention. However, celiac disease can also manifest in myriad extraintestinal symptoms, ranging from headache and fatigue to delayed puberty and psychiatric disorders, with differing presentations in children and adults.

To date, the only treatment is adopting a gluten-free diet (GFD). Although key to preventing persistent villous atrophy, the main cause of complications in celiac disease, lifelong adherence to GFD is challenging and may not resolve all clinical issues. These shortcomings have driven recent efforts to develop novel therapeutic options for patients with this disease.

Here are five things to know about celiac disease.
 

1. Rising Prevalence of Celiac Disease and Other Autoimmune Disorders Suggests Environmental Factors May Be at Play

Gluten was first identified as the cause of celiac disease in the 1950s. At that time, the condition was thought to be a relatively rare GI disease of childhood that primarily affected people of European descent, but it is now known to be a common disease affecting those of various ages, races, and ethnicities.

2018 meta-analysis found the pooled global prevalence of celiac disease was 1.4%. Incidence has increased by as much as 7.5% annually over the past several decades.

Increased awareness among clinicians and improved detection likely play a role in the trend. However, the growth in celiac disease is consistent with that seen for other autoimmune disorders, according to a 2024 update of evidence surrounding celiac disease. Shared environmental factors have been proposed as triggers for celiac disease and other autoimmune diseases and appear to be influencing their rise, the authors noted. These factors include migration and population growth, changing dietary patterns and food processing practices, and altered wheat consumption.
 

2. No-Biopsy Diagnosis Is Accepted for Children and Shows Promise for Adults

It is estimated that almost 60 million people worldwide have celiac disease, but most remain undiagnosed or misdiagnosed, or they experience significant diagnostic delays.

Prospective data indicate that children with first-degree relatives with celiac disease are at a significantly higher risk of developing the condition, which should prompt screening efforts in this population.

The 2023 updated guidelines from the American College of Gastroenterology (ACG) state that serology testing plays a central role in screening. This commonly involves serological testing for positive serological markers of the disease, including immunoglobulin A (IgA), anti-tissue transglutaminase IgA (tTG-IgA), anti-deamidated gliadin peptide, or endomysial antibodies.

To confirm diagnosis, clinicians have relied on intestinal biopsy since the late 1950s. The ACG still recommends esophagogastroduodenoscopy with multiple duodenal biopsies for confirmation of diagnosis in both children and adults with suspicion of celiac disease. However, recent years have seen a shift toward a no-biopsy approach.

For more than a decade in Europe, a no-biopsy approach has been established practice in pediatric patients, for whom the burden of obtaining a histological confirmation is understandably greater. Most guidelines now permit children to be diagnosed with celiac disease in the absence of a biopsy under specific circumstances (eg, characteristic symptoms of celiac disease and tTG-IgA levels > 10 times the upper limit of normal). The ACG guidelines state that “this approach is a reasonable alternative to the standard approach to a [celiac disease] diagnosis in selected children.”

The ACG does not recommend a no-biopsy approach in adults, noting that, in comparison with children, there is a relative lack of data indicating that serology is predictive in this population. However, it does recognize that physicians may encounter patients for whom a biopsy diagnosis may not be safe or practical. In such cases, an “after-the-fact” diagnosis of likely celiac disease can be given to symptomatic adult patients with a ≥ 10-fold elevation of tTG-IgA and a positive endomysial antibody in a second blood sample.

A 2024 meta-analysis of 18 studies involving over 12,103 adult patients from 15 countries concluded that a no-biopsy approach using tTG-IgA antibody levels ≥ 10 times the upper limit of normal was highly specific and predictive of celiac disease.
 

 

 

3. Celiac Disease Is Associated With Several Life-Threatening Conditions

Emerging data indicate that gastroenterologists should be vigilant in screening patients with celiac disease for several other GI conditions.

Inflammatory bowel disease and celiac disease have a strong bidirectional association, suggesting a possible genetic link between the conditions and indicating that physicians should consider the alternate diagnosis when symptoms persist after treatment.

Given the hypervigilance around food and diet inherent to celiac disease, patients are at an increased risk of developing avoidant/restrictive food intake disorder, according to a 2022 retrospective study.

In 2023, Italian investigators showed that children with celiac disease have an elevated prevalence of functional GI disorders even after adopting a GFD for a year, regardless of whether they consumed processed or natural foods. It was unclear whether this was due to a chronic inflammatory process or to nutritional factors.

Complications resulting from celiac disease are not limited to GI disorders. For a variety of underlying pathophysiological reasons, including intestinal permeability, hyposplenism, and malabsorption of nutrients, patients with celiac disease may be at a higher risk for non-GI conditions, such as osteopeniawomen’s health disorders (eg, ovarian failure, endometriosis, or pregnancy loss), juvenile idiopathic arthritis in children and rheumatoid arthritis in adultscertain forms of cancerinfectious diseases, and cardiomyopathy.
 

4. GFD Is the Only Treatment, but It’s Imperfect and Frustrating for Patients

GFD is the only treatment for celiac disease and must be adhered to without deviation throughout a patient’s life.

Maintaining unwavering adherence reaps considerable benefits: Improved clinical symptoms, robust mucosal healing, and normalization of serological markers. Yet it also takes a considerable toll on patients. Patients with celiac disease struggle with a host of negative physical, psychological, and social impacts. They also report a higher treatment burden than those with gastroesophageal reflux disease or hypertension, and comparable with end-stage renal disease.

GFD also poses financial challenges. Although the price of gluten-free products has decreased in recent years, they still cost significantly more than items with gluten.

Adherence to GFD does not always equate to complete mucosal recovery. While mucosal recovery is achieved in 95% of children within 2 years of the diet’s adoption, only 34% and 66% of adults obtain it within 2 and 5 years, respectively.

GFD may lead to nutrient imbalances because gluten-free foods are typically low in alimentary fiber, micronutrients (eg, vitamin D, vitamin B12, or folate), and minerals (eg, iron, zinc, magnesium, or calcium). With higher sugar and fat content, GFD may leave patients susceptible to unwanted weight gain.

The pervasiveness of gluten in the food production system makes the risk for cross-contamination high. Gluten is often found in both naturally gluten-free foods and products labeled as such. Gluten-sensing technologies, some of which can be used via smartphone apps, have been developed to help patients identify possible cross-contamination. However, the ACG guidelines recommend against the use of these technologies until there is sufficient evidence supporting their ability to improve adherence and clinical outcomes.
 

5. Novel Therapies for Celiac Disease Are in the Pipeline

The limitations of GFD as the standard treatment for celiac disease have led to an increased focus on developing novel therapeutic interventions. They can be sorted into five key categories: Modulation of the immunostimulatory effects of toxic gluten peptides, elimination of toxic gluten peptides before they reach the intestine, induction of gluten tolerance, modulation of intestinal permeability, and restoration of gut microbiota balance.

Three therapies designed to block antigen presentation by HLA-DQ2/8, the gene alleles that predispose people to celiac disease, show promise: TPM502, an agent that contains three gluten-specific antigenic peptides with overlapping T-cell epitopes for the HLA-DQ2.5 gene; KAN-101, designed to induce gluten tolerance by targeting receptors on the liver; and DONQ52, a multi-specific antibody that targets HLA-DQ2. The KAN-101 therapy received Fast Track designation by the US Food and Drug Administration in 2022.

These and several other agents in clinical and preclinical development are discussed in detail in a 2024 review article. Although no therapies have reached phase 3 testing, when they do, it will undoubtedly be welcomed by those with celiac disease.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>168334</fileName> <TBEID>0C05073F.SIG</TBEID> <TBUniqueIdentifier>MD_0C05073F</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240607T142521</QCDate> <firstPublished>20240607T163103</firstPublished> <LastPublished>20240607T163103</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240607T163103</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>John Watson</byline> <bylineText>JOHN WATSON</bylineText> <bylineFull>JOHN WATSON</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>Celiac disease is a chronic, immune-mediated, systemic disorder caused by intolerance to gluten — a protein present in rye, barley, and wheat grains — that affe</metaDescription> <articlePDF/> <teaserImage/> <teaser>Although avoidance of gluten is key to preventing villous atrophy, it is challenging and may not resolve all clinical issues.</teaser> <title>Celiac Disease: Five Things to Know</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>GIHOLD</publicationCode> <pubIssueName>January 2014</pubIssueName> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle/> <journalFullTitle/> <copyrightStatement/> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>pn</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term>15</term> <term>13</term> <term canonical="true">21</term> <term>25</term> </publications> <sections> <term>27980</term> <term canonical="true">39313</term> </sections> <topics> <term>204</term> <term canonical="true">213</term> <term>49620</term> <term>271</term> <term>280</term> <term>231</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Celiac Disease: Five Things to Know</title> <deck/> </itemMeta> <itemContent> <p>Celiac disease is a chronic, immune-mediated, <a href="https://journals.lww.com/ajg/fulltext/2023/01000/american_college_of_gastroenterology_guidelines.17.aspx">systemic disorder</a> caused by intolerance to gluten — a protein present in rye, barley, and wheat grains — that affects genetically predisposed individuals.</p> <p>Due to its wide spectrum of clinical manifestations, celiac disease resembles a multisystemic disorder. Its <a href="https://doi.org/10.1056/NEJMcp1113994">most common</a> gastrointestinal (GI) symptoms include chronic diarrhea, weight loss, and abdominal distention. However, celiac disease can also manifest in myriad <a href="https://doi.org/10.1097/MPG.0000000000001634">extraintestinal symptoms</a>, ranging from headache and fatigue to delayed puberty and psychiatric disorders, with differing presentations in children and adults.<br/><br/>To date, the only treatment is adopting a gluten-free diet (GFD). Although key to <a href="https://doi.org/10.1097/MOG.0000000000001009">preventing persistent villous atrophy</a>, the main cause of complications in celiac disease, lifelong adherence to GFD is challenging and may not resolve all clinical issues. These shortcomings have driven recent efforts to develop novel therapeutic options for patients with this disease.<br/><br/>Here are five things to know about celiac disease.<br/><br/></p> <h2>1. Rising Prevalence of Celiac Disease and Other Autoimmune Disorders Suggests Environmental Factors May Be at Play</h2> <p>Gluten was first identified as the cause of celiac disease <a href="https://doi.org/10.1177/0884533611399775">in the 1950s</a>. At that time, the condition was <a href="https://doi.org/10.1056/NEJMcp1113994">thought</a> to be a relatively rare GI disease of childhood that primarily affected people of European descent, but it is now known to be a common disease affecting those of various ages, races, and ethnicities.</p> <p>A <a href="https://doi.org/10.1016/j.cgh.2017.06.037">2018 meta-analysis</a> found the pooled global prevalence of celiac disease was 1.4%. Incidence has increased by as much as <a href="https://doi.org/10.14309/ajg.0000000000000523">7.5% annually</a> over the past several decades.<br/><br/><a href="https://doi.org/10.1053/jcgh.2003.50004">Increased awareness</a> among clinicians and improved detection likely play a role in the trend. However, the growth in celiac disease is consistent with that seen for other autoimmune disorders, according to <a href="https://doi.org/10.1016/j.intimp.2024.112020">a 2024 update</a> of evidence surrounding celiac disease. Shared environmental factors have been proposed as triggers for celiac disease and other autoimmune diseases and appear to be influencing their rise, the authors noted. These factors include migration and population growth, changing dietary patterns and food processing practices, and altered wheat consumption.<br/><br/></p> <h2>2. No-Biopsy Diagnosis Is Accepted for Children and Shows Promise for Adults</h2> <p>It is estimated that <a href="https://www.gastrojournal.org/article/S0016-5085(23)05688-3/fulltext">almost 60 million people</a> worldwide have celiac disease, but most remain undiagnosed or misdiagnosed, or they experience significant diagnostic delays.</p> <p><a href="https://www.medscape.com/viewarticle/974500">Prospective data</a> indicate that children with first-degree relatives with celiac disease are at a significantly higher risk of developing the condition, which should prompt screening efforts in this population.<br/><br/>The <a href="https://doi.org/10.14309/ajg.0000000000002075">2023 updated guidelines</a> from the American College of Gastroenterology (ACG) state that serology testing plays a central role in screening. This commonly <a href="https://doi.org/10.1097/MOG.0000000000001009">involves</a> serological testing for positive serological markers of the disease, including immunoglobulin A (IgA), anti-tissue transglutaminase IgA (tTG-IgA), anti-deamidated gliadin peptide, or endomysial antibodies.<br/><br/>To confirm diagnosis, clinicians have relied on intestinal biopsy since the late 1950s. The ACG <a href="https://doi.org/10.14309/ajg.0000000000002075">still recommends</a> esophagogastroduodenoscopy with multiple duodenal biopsies for confirmation of diagnosis in both children and adults with suspicion of celiac disease. However, recent years have seen a shift toward a no-biopsy approach.<br/><br/>For <a href="https://doi.org/10.1097/MPG.0000000000002497">more than a decade</a> in Europe, a no-biopsy approach has been established practice in pediatric patients, for whom the burden of obtaining a histological confirmation is understandably greater. <a href="http://dx.doi.org/10.3748/wjg.v28.i1.154">Most guidelines</a> now permit children to be diagnosed with celiac disease in the absence of a biopsy under specific circumstances (eg, characteristic symptoms of celiac disease and tTG-IgA levels &gt; 10 times the upper limit of normal). The ACG guidelines state that “this approach is a reasonable alternative to the standard approach to a [celiac disease] diagnosis in selected children.”<br/><br/>The ACG does not recommend a no-biopsy approach in adults, noting that, in comparison with children, there is a relative lack of data indicating that serology is predictive in this population. However, it does recognize that physicians may encounter patients for whom a biopsy diagnosis may not be safe or practical. In such cases, an “after-the-fact” diagnosis of likely celiac disease can be given to symptomatic adult patients with a ≥ 10-fold elevation of tTG-IgA and a positive endomysial antibody in a second blood sample.<br/><br/><a href="https://doi.org/10.1053/j.gastro.2023.12.023">A 2024 meta-analysis</a> of 18 studies involving over 12,103 adult patients from 15 countries concluded that a no-biopsy approach using tTG-IgA antibody levels ≥ 10 times the upper limit of normal was highly specific and predictive of celiac disease.<br/><br/></p> <h2>3. Celiac Disease Is Associated With Several Life-Threatening Conditions</h2> <p>Emerging data indicate that gastroenterologists should be vigilant in screening patients with celiac disease for several other GI conditions.</p> <p>Inflammatory bowel disease and celiac disease have <a href="https://www.medscape.com/viewarticle/975276">a strong bidirectional association</a>, suggesting a possible genetic link between the conditions and indicating that physicians should consider the alternate diagnosis when symptoms persist after treatment.<br/><br/>Given the hypervigilance around food and diet inherent to celiac disease, patients are at an increased risk of developing avoidant/restrictive food intake disorder, according to <a href="https://www.medscape.com/viewarticle/969075">a 2022 retrospective study</a>.<br/><br/>In 2023, Italian investigators <a href="https://www.medscape.com/viewarticle/987364">showed that</a> children with celiac disease have an elevated prevalence of functional GI disorders even after adopting a GFD for a year, regardless of whether they consumed processed or natural foods. It was unclear whether this was due to a chronic inflammatory process or to nutritional factors.<br/><br/>Complications resulting from celiac disease are not limited to GI disorders. For a variety of underlying pathophysiological reasons, including intestinal permeability, hyposplenism, and malabsorption of nutrients, patients with celiac disease may be at a higher risk for non-GI conditions, such as <a href="https://doi.org/10.1016/0016-5085(95)90276-7">osteopenia</a>, <a href="https://www.medscape.com/s/viewarticle/998008">women’s health disorders</a> (eg, ovarian failure, endometriosis, or pregnancy loss), <a href="https://www.medscape.com/viewarticle/984209">juvenile idiopathic arthritis in children and rheumatoid arthritis in adults</a>, <a href="https://doi.org/10.1136/gut.30.3.333">certain forms of cancer</a>, <a href="https://doi.org/10.1017/S0950268816003204">infectious diseases</a>, and <a href="https://doi.org/10.3390/jcm13041045">cardiomyopathy</a>.<br/><br/></p> <h2>4. GFD Is the Only Treatment, but It’s Imperfect and Frustrating for Patients</h2> <p>GFD is the only treatment for celiac disease and must be adhered to without deviation throughout a patient’s life.</p> <p>Maintaining unwavering adherence reaps considerable <a href="https://www.mdpi.com/2072-6643/16/7/1006">benefits</a>: Improved clinical symptoms, robust mucosal healing, and normalization of serological markers. Yet it also takes a considerable toll on patients. Patients with celiac disease struggle with <a href="https://doi.org/10.2174/1871530318666180723100003">a host of</a> negative physical, psychological, and social impacts. They also report <a href="https://doi.org/10.1038/ajg.2014.29">a higher treatment burden</a> than those with gastroesophageal reflux disease or hypertension, and comparable with end-stage renal disease.<br/><br/>GFD also poses financial challenges. Although the price of gluten-free products <a href="https://doi.org/10.3390/nu11020399">has decreased</a> in recent years, they still cost significantly more than items with gluten.<br/><br/>Adherence to GFD does not always equate to complete <a href="https://www.mdpi.com/2072-6643/16/7/1006">mucosal recovery</a>. While mucosal recovery is achieved in 95% of children within 2 years of the diet’s adoption, only 34% and 66% of adults obtain it within 2 and 5 years, respectively.<br/><br/>GFD may lead to <a href="https://doi.org/10.1016/j.clnu.2016.05.002">nutrient imbalances</a> because gluten-free foods are typically low in alimentary fiber, micronutrients (eg, vitamin D, vitamin B12, or folate), and minerals (eg, iron, zinc, magnesium, or calcium). With higher sugar and fat content, GFD may leave patients <a href="https://doi.org/10.1097/MCG.0000000000001998">susceptible to</a> unwanted weight gain.<br/><br/>The pervasiveness of gluten in the food production system makes the risk for cross-contamination high. Gluten is <a href="https://doi.org/10.3390/nu16071006">often found</a> in both naturally gluten-free foods and products labeled as such. Gluten-sensing technologies, some of which can be used <a href="https://www.medscape.com/s/viewarticle/974842">via smartphone apps</a>, have been developed to help patients identify possible cross-contamination. However, the ACG guidelines <a href="https://doi.org/10.14309/ajg.0000000000002075">recommend</a> against the use of these technologies until there is sufficient evidence supporting their ability to improve adherence and clinical outcomes.<br/><br/></p> <h2>5. Novel Therapies for Celiac Disease Are in the Pipeline</h2> <p>The limitations of GFD as the standard treatment for celiac disease have led to an increased focus on developing novel therapeutic interventions. They can be sorted into <a href="https://www.sciencedirect.com/science/article/abs/pii/S1567576924005381?via%3Dihub">five key categories</a>: Modulation of the immunostimulatory effects of toxic gluten peptides, elimination of toxic gluten peptides before they reach the intestine, induction of gluten tolerance, modulation of intestinal permeability, and restoration of gut microbiota balance.</p> <p><a href="https://www.medscape.com/s/viewarticle/992697">Three therapies</a> designed to block antigen presentation by <em>HLA-DQ2/8</em>, the gene alleles that predispose people to celiac disease, show promise: <a href="https://www.clinicaltrials.gov/study/NCT05660109">TPM502</a>, an agent that contains three gluten-specific antigenic peptides with overlapping T-cell epitopes for the <em>HLA-DQ2.5</em> gene; <a href="https://clinicaltrials.gov/study/NCT05574010">KAN-101</a>, designed to induce gluten tolerance by targeting receptors on the liver; and <a href="https://clinicaltrials.gov/study/NCT05425446">DONQ52</a>, a multi-specific antibody that targets <em>HLA-DQ2</em>. The KAN-101 therapy received Fast Track designation by the US Food and Drug Administration in 2022.<br/><br/>These and several other agents in clinical and preclinical development are discussed in detail in <a href="https://doi.org/10.1016/j.intimp.2024.112020">a 2024 review article</a>. Although no therapies have reached phase 3 testing, <a href="https://doi.org/10.1038/s41575-024-00895-3">when they do</a>, it will undoubtedly be welcomed by those with celiac disease. <span class="end"/></p> <p> <em>A version of this article first appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/celiac-disease-five-things-know-2024a1000ami">Medscape.com</a></span>.</em> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
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

Could a Fungal Infection Cause a Future Pandemic?

Article Type
Changed
Thu, 05/30/2024 - 15:51

The principle of resilience and survival is crucial for medically significant fungi. These microorganisms are far from creating the postapocalyptic scenario depicted in TV series like The Last of Us, and much work is necessary to learn more about them. Accurate statistics on fungal infections, accompanied by clinical histories, simple laboratory tests, new antifungals, and a necessary One Health approach are lacking. 

The entomopathogenic fungus Ophiocordyceps unilateralis was made notorious by the TV series, but for now, it only manages to control the brains of some ants at will. Luckily, there are no signs that fungi affecting humans are inclined to create zombies.

What is clear is that the world belongs to the kingdom of fungi and that fungi are everywhere. There are already close to 150,000 described species, but millions remain to be discovered. They abound in decomposing organic matter, soil, or animal excrement, including that of bats and pigeons. Some fungi have even managed to find a home in hospitals. Lastly, we must not forget those that establish themselves in the human microbiome.

Given such diversity, it is legitimate to ask whether any of them could be capable of generating new pandemics. Could the forgotten Cryptococcus neoformansAspergillus fumigatus, or Histoplasma species, among others, trigger new health emergencies on the scale of the one generated by SARS-CoV-2?

We cannot forget that a coronavirus has already confirmed that reality can surpass fiction. However, Edith Sánchez Paredes, a biologist, doctor in biomedical sciences, and specialist in medical mycology, provided a reassuring response to Medscape Spanish Edition on this point.

“That would be very difficult to see because the way fungal infections are acquired is not from person to person, in most cases,” said Dr. Sánchez Paredes, from the Mycology Unit of the Faculty of Medicine at the National Autonomous University of Mexico.

Close to 300 species have already been classified as pathogenic in humans. Although the numbers are not precise and are increasing, it is estimated that around 1,500,000 people worldwide die each year of systemic fungal infections.

“However, it is important to emphasize that establishment of an infection depends not only on the causal agent. A crucial factor is the host, in this case, the human. Generally, these types of infections will develop in individuals with some deficiency in their immune system. The more deficient the immune response, the more likely a fungal infection may occur,” stated Dr. Sánchez Paredes.

The possibility of a pandemic like the one experienced with SARS-CoV-2 in the short term is remote, but the threat posed by fungal infections persists.

In 2022, the World Health Organization (WHO) defined a priority list of pathogenic fungi, with the aim of guiding actions to control them. It is mentioned there that invasive fungal diseases are on the rise worldwide, particularly in immunocompromised populations.

“Despite the growing concern, fungal infections receive very little attention and resources, leading to a paucity of quality data on fungal disease distribution and antifungal resistance patterns. Consequently, it is impossible to estimate their exact burden,” as stated in the document.

In line with this, an article published in Mycoses in 2022 concluded that fungal infections are neglected diseases in Latin America. Among other difficulties, deficiencies in access to tests such as polymerase chain reaction or serum detection of beta-1,3-D-glucan have been reported there.

In terms of treatments, most countries encounter problems with access to liposomal amphotericin B and new azoles, such as posaconazole and isavuconazole.

“Unfortunately, in Latin America, we suffer from a poor infrastructure for diagnosing fungal infections; likewise, we have limited access to antifungals available in the global market. What’s more, we lack reliable data on the epidemiology of fungal infections in the region, so many times governments are unaware of the true extent of the problem,” said Rogelio de Jesús Treviño Rangel, PhD, a medical microbiologist and expert in clinical mycology, professor, and researcher at the Faculty of Medicine of the Autonomous University of Nuevo León in Mexico.
 

 

 

Need for More Medical Mycology Training

Dr. Fernando Messina is a medical mycologist with the Mycology Unit of the Francisco Javier Muñiz Infectious Diseases Hospital in Buenos Aires, Argentina. He has noted an increase in the number of cases of cryptococcosishistoplasmosis, and aspergillosis in his daily practice.

“Particularly, pulmonary aspergillosis is steadily increasing. This is because many patients have structural lung alterations that favor the appearance of this mycosis. This is related to the increase in cases of tuberculosis and the rise in life expectancy of patients with chronic obstructive pulmonary disease or other pulmonary or systemic diseases,” Dr. Messina stated.

For Dr. Messina, the main obstacle in current clinical practice is the low level of awareness among nonspecialist physicians regarding the presence of systemic fungal infections, and because these infections are more common than realized, it is vital to consider fungal etiology before starting empirical antibiotic therapy.

“Health professionals usually do not think about mycoses because mycology occupies a very small space in medical education at universities. As the Venezuelan mycologist Gioconda Cunto de San Blas once said, ‘Mycology is the Cinderella of microbiology.’ To change this, we need to give more space to mycoses in undergraduate and postgraduate studies,” Dr. Messina asserted.

He added, “The main challenge is to train professionals with an emphasis on the clinical interpretation of cases. Current medicine has a strong trend toward molecular biology and the use of rapid diagnostic methods, without considering the clinical symptoms or the patient’s history. Determinations are very useful, but it is necessary to interpret the results.”

Dr. Messina sees it as unlikely in the short term for a pandemic to be caused by fungi, but if it were to occur, he believes it would happen in healthcare systems in regions that are not prepared in terms of infrastructure. However, as seen in the health emergency resulting from SARS-CoV-2, he thinks the impact would be mitigated by the performance of healthcare professionals.

“In general, we have the ability to adapt to any adverse situation or change — although it is clear that we need more doctors, biochemists, and microbiologists trained in mycology,” emphasized Dr. Messina.

More than 40 interns pass through Muñiz Hospital each year. They are doctors and biochemists from Argentina, other countries in the region, or even Europe, seeking to enhance their training in mycology. Regarding fungal infection laboratory work, the interest lies in learning to use traditional techniques and innovative molecular methods.

“Rapid diagnostic methods, especially the detection of circulating antigens, have marked a change in the prognosis of deep mycosis in immunocompromised hosts. The possibility of screening and monitoring in this group of patients is very important and has a great benefit,” said Gabriela Santiso, PhD, a biochemist and head of the Mycology Unit of the Francisco Javier Muñiz Infectious Diseases Hospital.

According to Dr. Santiso, the current landscape includes the ability to identify genus and species, which can help in understanding resistance to antifungals. Furthermore, conducting sensitivity tests to these drugs, using standardized commercial methods, also provides timely information for treatment.

But Dr. Santiso warns that Latin America is a vast region with great disparity in human and technological resources. Although most countries in the region have networks facilitating access to timely diagnosis, resources are generally more available in major urban centers.

This often clashes with the epidemiology of most fungal infections. “Let’s not forget that many fungal pathologies affect low-income people who have difficulties accessing health centers, which sometimes turns them into chronic diseases that are hard to treat,” Dr. Santiso pointed out.

In mycology laboratories, the biggest cost is incurred by new diagnostic tests, such as those allowing molecular identification. Conventional methods are not usually expensive, but they require time and effort to train human resources to handle them.

Because new methodologies are not always available or easily accessible throughout the region, Dr. Santiso recommended not neglecting traditional mycological techniques. “Molecular methods, rapid diagnostic methods, and conventional mycology techniques are complementary and not mutually exclusive tests. Continuous training and updating are needed in this area,” she emphasized.
 

 

 

Why Are Resistant Fungal Infections Becoming Increasingly Common?

The first barrier for fungi to cause infection in humans is body temperature; most of them cannot withstand 37 °C. However, they also struggle to evade the immune response that is activated when they try to enter the body. 

“We are normally exposed to many of these fungi, almost all the time, but if our immune system is adequate, it may not go beyond a mild infection, in most cases subclinical, which will resolve quickly,” Dr. Sánchez Paredes stated.

However, according to Dr. Sánchez Paredes, if the immune response is weak, “the fungus will have no trouble establishing itself in our organs. Some are even part of our microbiota, such as Candida albicans, which in the face of an imbalance or immunocompromise, can lead to serious infections.”

It is clear that the population at risk for immunosuppression has increased. According to the WHO, this is due to the high prevalence of such diseases as tuberculosis, cancer, and HIV infection, among others.

But the WHO also believes that the increase in fungal infections is related to greater population access to critical care units, invasive procedures, chemotherapy, or immunotherapy treatments.

Furthermore, factors related to the fungus itself and the environment play a role. “These organisms have enzymes, proteins, and other molecules that allow them to survive in the environment in which they normally inhabit. When they face a new and stressful one, they must express other molecules that will allow them to survive. All of this helps them evade elements of the immune system, antifungals, and, of course, body temperature,” according to Dr. Sánchez Paredes.

It is possible that climate change is also behind the noticeable increase in fungal infections and that this crisis may have an even greater impact in the future. The temperature of the environment has increased, and fungi will have to adapt to the planet’s temperature, to the point where body temperature may no longer be a significant barrier for them.

Environmental changes would also be responsible for modifications in the distribution of endemic mycoses, and it is believed that fungi will more frequently find new ecological niches, be able to survive in other environments, and alter distribution zones.

This is what is happening between Mexico and the United States with coccidioidomycosis, or valley fever. “We will begin to see cases of some mycoses where they were not normally seen, so we will have to conduct more studies to confirm that the fungus is inhabiting these new areas or is simply appearing in new sites owing to migration and the great mobility of populations,” Dr. Sánchez Paredes said.

Finally, exposure to environmental factors would partly be responsible for the increasing resistance to first-line antifungals observed in these microorganisms. This seems to be the case with A. fumigatus when exposed to azoles used as fungicides in agriculture.
 

One Health in Fungal Infections

The increasing resistance to antifungals is a clear testament that human, animal, and environmental health are interconnected. This is why a multidisciplinary approach that adopts the perspective of One Health is necessary for its management.

“The use of fungicides in agriculture, structurally similar to the azoles used in clinics, generates resistance in Aspergillus fumigatus found in the environment. These fungi in humans can be associated with infections that do not respond to first-line treatment,” explained Carlos Arturo Álvarez, an infectious diseases physician and professor at the Faculty of Medicine at the National University of Colombia.

According to Dr. Álvarez, the approach to control them should not only focus on the search for diagnostic methods that allow early detection of antifungal resistance or research on new antifungal treatments. He believes that progress must also be made with strategies that allow for the proper use of antifungals in agriculture.

“Unfortunately, the One Health approach is not yet well implemented in the region, and in my view, there is a lack of articulation in the different sectors. That is, there is a need for true coordination between government offices of agriculture, animal and human health, academia, and international organizations. This is not happening yet, and I believe we are in the initial stage of visibility,” Dr. Álvarez opined.

Veterinary public health is another pillar of the aforementioned approach. For various reasons, animals experience a higher frequency of fungal infections. A few carry and transmit true zoonoses that affect human health, but most often, animals act only as sentinels indicating a potential source of transmission.

Carolina Segundo Zaragoza, PhD, has worked in veterinary mycology for 30 years. She currently heads the veterinary mycology laboratory at the Animal Production Teaching, Research, and Extension Center in Altiplano, under the Faculty of Veterinary Medicine and Animal Husbandry at the National Autonomous University of Mexico. Because she has frequent contact with specialists in human mycology, during her professional career she has received several patient consultations, most of which were for cutaneous mycoses.

“They detect some dermatomycosis and realize that the common factor is owning a companion animal or a production animal with which the patient has contact. Both animals and humans present the same type of lesions, and then comes the question: Who infected whom? I remind them that the main source of infection is the soil and that animals should not be blamed in the first instance,” Dr. Segundo Zaragoza clarified.

She is currently collaborating on a research project analyzing the presence of Coccidioides immitis in the soil. This pathogen is responsible for coccidioidomycosis in dogs and humans, and she sees with satisfaction how these types of initiatives, which include some components of the One Health vision, are becoming more common in Mexico.

“Fortunately, human mycologists are increasingly providing more space for the dissemination of veterinary mycology. So I have had the opportunity to be invited to different forums on medical mycology to present the clinical cases we can have in animals and talk about the research projects we carry out. I have more and more opportunities to conduct joint research with human mycologists and veterinary doctors,” she said.

Dr. Segundo Zaragoza believes that to better implement the One Health vision, standardizing the criteria for detecting, diagnosing, and treating mycoses is necessary. She considers that teamwork will be key to achieving the common goal of safeguarding the well-being and health of humans and animals.
 

 

 

Alarms Sound for Candida auris

The WHO included the yeast Candida auris in its group of pathogens with critical priority, and since 2009, it has raised alarm owing to the ease with which it grows in hospitals. In that setting, C auris is known for its high transmissibility, its ability to cause outbreaks, and the high mortality rate from disseminated infections.

“It has been a concern for the mycological community because it shows resistance to most antifungals used clinically, mainly azoles, but also for causing epidemic outbreaks,” emphasized Dr. Sánchez Paredes.

Its mode of transmission is not very clear, but it has been documented to be present on the skin and persist in hospital materials and furniture. It causes nosocomial infections in critically ill patients, such as those in intensive care, and those with cancer or who have received a transplant.

Risk factors for its development include renal insufficiency, hospital stays of more than 15 days, mechanical ventilation, central lines, use of parenteral nutrition, and presence of sepsis.

As for other mycoses, there are no precise studies reporting global incidence rates, but the trend indicates an increase in the detection of outbreaks in various countries lately — something that began to be visible during the COVID-19 pandemic.

In Mexico, Dr. Treviño Rangel and colleagues from Nuevo León reported the first case of candidemia caused by this agent. It occurred in May 2020 and involved a 58-year-old woman with a history of severe endometriosis and multiple complications in the gastrointestinal tract. The patient’s condition improved favorably thanks to antifungal therapy with caspofungin and liposomal amphotericin B.

However, 3 months after that episode, the group reported an outbreak of C. auris at the same hospital in 12 critically ill patients co-infected with SARS-CoV-2. All were on mechanical ventilation, had peripherally inserted central catheters and urinary catheters, and had a prolonged hospital stay (20-70 days). The mortality in patients with candidemia in this cohort was 83.3%.
 

Open Ending

As seen in some science fiction series, fungal infections in the region still have an open ending, and Global Action For Fungal Infections (GAFFI) has estimated that with better diagnostics and treatments, deaths caused by fungi could decrease to less than 750,000 per year worldwide.

But if everything continues as is, some aspects of what is to come may resemble the dystopia depicted in The Last of Us. No zombies, but emerging and reemerging fungi in a chaotic distribution, and resistant to all established treatments.

“The risk factors of patients and their immune status, combined with the behavior of mycoses, bring a complicated scenario. But therapeutic failure resulting from multidrug resistance to antifungals could make it catastrophic,” Dr. Sánchez Paredes summarized.

At the moment, there are only four families of drugs capable of counteracting fungal infections — and as mentioned, some are already scarce in Latin America’s hospital pharmacies.

“Historically, fungal infections have been given less importance than those caused by viruses or bacteria. Even in some developed countries, the true extent of morbidity and mortality they present is unknown. This results in less investment in the development of new antifungal molecules because knowledge is lacking about the incidence and prevalence of these diseases,” Dr. Treviño Rangel pointed out.

He added that the main limitation for the development of new drugs is economic. “Unfortunately, not many pharmaceutical companies are willing to invest in the development of new antifungals, and there are no government programs specifically promoting and supporting research into new therapeutic options against these neglected diseases,” he asserted.

Development of vaccines to prevent fungal infections faces the same barriers. Although, according to Dr. Treviño Rangel, the difficulties are compounded by the great similarity between fungal cells and human cells. This makes it possible for harmful cross-reactivity to occur. In addition, because most severe fungal infections occur in individuals with immunosuppression, a vaccine would need to trigger an adequate immune response despite this issue.

Meanwhile, fungi quietly continue to do what they do best: resist and survive. For millions of years, they have mutated and adapted to new environments. Some theories even blame them for the extinction of dinosaurs and the subsequent rise of mammals. They exist on the edge of life and death, decomposing and creating. There is consensus that at the moment, it does not seem feasible for them to generate a pandemic like the one due to SARS-CoV-2, given their transmission mechanism. But who is willing to rule out that this may not happen in the long or medium term?

Dr. Sánchez Paredes, Dr. Treviño Rangel, Dr. Messina, Dr. Santiso, Dr. Álvarez, and Dr. Segundo Zaragoza have declared no relevant financial conflicts of interest. 
 

This story was translated from Medscape Spanish Edition using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication. A version of this article appeared on Medscape.com.

Publications
Topics
Sections

The principle of resilience and survival is crucial for medically significant fungi. These microorganisms are far from creating the postapocalyptic scenario depicted in TV series like The Last of Us, and much work is necessary to learn more about them. Accurate statistics on fungal infections, accompanied by clinical histories, simple laboratory tests, new antifungals, and a necessary One Health approach are lacking. 

The entomopathogenic fungus Ophiocordyceps unilateralis was made notorious by the TV series, but for now, it only manages to control the brains of some ants at will. Luckily, there are no signs that fungi affecting humans are inclined to create zombies.

What is clear is that the world belongs to the kingdom of fungi and that fungi are everywhere. There are already close to 150,000 described species, but millions remain to be discovered. They abound in decomposing organic matter, soil, or animal excrement, including that of bats and pigeons. Some fungi have even managed to find a home in hospitals. Lastly, we must not forget those that establish themselves in the human microbiome.

Given such diversity, it is legitimate to ask whether any of them could be capable of generating new pandemics. Could the forgotten Cryptococcus neoformansAspergillus fumigatus, or Histoplasma species, among others, trigger new health emergencies on the scale of the one generated by SARS-CoV-2?

We cannot forget that a coronavirus has already confirmed that reality can surpass fiction. However, Edith Sánchez Paredes, a biologist, doctor in biomedical sciences, and specialist in medical mycology, provided a reassuring response to Medscape Spanish Edition on this point.

“That would be very difficult to see because the way fungal infections are acquired is not from person to person, in most cases,” said Dr. Sánchez Paredes, from the Mycology Unit of the Faculty of Medicine at the National Autonomous University of Mexico.

Close to 300 species have already been classified as pathogenic in humans. Although the numbers are not precise and are increasing, it is estimated that around 1,500,000 people worldwide die each year of systemic fungal infections.

“However, it is important to emphasize that establishment of an infection depends not only on the causal agent. A crucial factor is the host, in this case, the human. Generally, these types of infections will develop in individuals with some deficiency in their immune system. The more deficient the immune response, the more likely a fungal infection may occur,” stated Dr. Sánchez Paredes.

The possibility of a pandemic like the one experienced with SARS-CoV-2 in the short term is remote, but the threat posed by fungal infections persists.

In 2022, the World Health Organization (WHO) defined a priority list of pathogenic fungi, with the aim of guiding actions to control them. It is mentioned there that invasive fungal diseases are on the rise worldwide, particularly in immunocompromised populations.

“Despite the growing concern, fungal infections receive very little attention and resources, leading to a paucity of quality data on fungal disease distribution and antifungal resistance patterns. Consequently, it is impossible to estimate their exact burden,” as stated in the document.

In line with this, an article published in Mycoses in 2022 concluded that fungal infections are neglected diseases in Latin America. Among other difficulties, deficiencies in access to tests such as polymerase chain reaction or serum detection of beta-1,3-D-glucan have been reported there.

In terms of treatments, most countries encounter problems with access to liposomal amphotericin B and new azoles, such as posaconazole and isavuconazole.

“Unfortunately, in Latin America, we suffer from a poor infrastructure for diagnosing fungal infections; likewise, we have limited access to antifungals available in the global market. What’s more, we lack reliable data on the epidemiology of fungal infections in the region, so many times governments are unaware of the true extent of the problem,” said Rogelio de Jesús Treviño Rangel, PhD, a medical microbiologist and expert in clinical mycology, professor, and researcher at the Faculty of Medicine of the Autonomous University of Nuevo León in Mexico.
 

 

 

Need for More Medical Mycology Training

Dr. Fernando Messina is a medical mycologist with the Mycology Unit of the Francisco Javier Muñiz Infectious Diseases Hospital in Buenos Aires, Argentina. He has noted an increase in the number of cases of cryptococcosishistoplasmosis, and aspergillosis in his daily practice.

“Particularly, pulmonary aspergillosis is steadily increasing. This is because many patients have structural lung alterations that favor the appearance of this mycosis. This is related to the increase in cases of tuberculosis and the rise in life expectancy of patients with chronic obstructive pulmonary disease or other pulmonary or systemic diseases,” Dr. Messina stated.

For Dr. Messina, the main obstacle in current clinical practice is the low level of awareness among nonspecialist physicians regarding the presence of systemic fungal infections, and because these infections are more common than realized, it is vital to consider fungal etiology before starting empirical antibiotic therapy.

“Health professionals usually do not think about mycoses because mycology occupies a very small space in medical education at universities. As the Venezuelan mycologist Gioconda Cunto de San Blas once said, ‘Mycology is the Cinderella of microbiology.’ To change this, we need to give more space to mycoses in undergraduate and postgraduate studies,” Dr. Messina asserted.

He added, “The main challenge is to train professionals with an emphasis on the clinical interpretation of cases. Current medicine has a strong trend toward molecular biology and the use of rapid diagnostic methods, without considering the clinical symptoms or the patient’s history. Determinations are very useful, but it is necessary to interpret the results.”

Dr. Messina sees it as unlikely in the short term for a pandemic to be caused by fungi, but if it were to occur, he believes it would happen in healthcare systems in regions that are not prepared in terms of infrastructure. However, as seen in the health emergency resulting from SARS-CoV-2, he thinks the impact would be mitigated by the performance of healthcare professionals.

“In general, we have the ability to adapt to any adverse situation or change — although it is clear that we need more doctors, biochemists, and microbiologists trained in mycology,” emphasized Dr. Messina.

More than 40 interns pass through Muñiz Hospital each year. They are doctors and biochemists from Argentina, other countries in the region, or even Europe, seeking to enhance their training in mycology. Regarding fungal infection laboratory work, the interest lies in learning to use traditional techniques and innovative molecular methods.

“Rapid diagnostic methods, especially the detection of circulating antigens, have marked a change in the prognosis of deep mycosis in immunocompromised hosts. The possibility of screening and monitoring in this group of patients is very important and has a great benefit,” said Gabriela Santiso, PhD, a biochemist and head of the Mycology Unit of the Francisco Javier Muñiz Infectious Diseases Hospital.

According to Dr. Santiso, the current landscape includes the ability to identify genus and species, which can help in understanding resistance to antifungals. Furthermore, conducting sensitivity tests to these drugs, using standardized commercial methods, also provides timely information for treatment.

But Dr. Santiso warns that Latin America is a vast region with great disparity in human and technological resources. Although most countries in the region have networks facilitating access to timely diagnosis, resources are generally more available in major urban centers.

This often clashes with the epidemiology of most fungal infections. “Let’s not forget that many fungal pathologies affect low-income people who have difficulties accessing health centers, which sometimes turns them into chronic diseases that are hard to treat,” Dr. Santiso pointed out.

In mycology laboratories, the biggest cost is incurred by new diagnostic tests, such as those allowing molecular identification. Conventional methods are not usually expensive, but they require time and effort to train human resources to handle them.

Because new methodologies are not always available or easily accessible throughout the region, Dr. Santiso recommended not neglecting traditional mycological techniques. “Molecular methods, rapid diagnostic methods, and conventional mycology techniques are complementary and not mutually exclusive tests. Continuous training and updating are needed in this area,” she emphasized.
 

 

 

Why Are Resistant Fungal Infections Becoming Increasingly Common?

The first barrier for fungi to cause infection in humans is body temperature; most of them cannot withstand 37 °C. However, they also struggle to evade the immune response that is activated when they try to enter the body. 

“We are normally exposed to many of these fungi, almost all the time, but if our immune system is adequate, it may not go beyond a mild infection, in most cases subclinical, which will resolve quickly,” Dr. Sánchez Paredes stated.

However, according to Dr. Sánchez Paredes, if the immune response is weak, “the fungus will have no trouble establishing itself in our organs. Some are even part of our microbiota, such as Candida albicans, which in the face of an imbalance or immunocompromise, can lead to serious infections.”

It is clear that the population at risk for immunosuppression has increased. According to the WHO, this is due to the high prevalence of such diseases as tuberculosis, cancer, and HIV infection, among others.

But the WHO also believes that the increase in fungal infections is related to greater population access to critical care units, invasive procedures, chemotherapy, or immunotherapy treatments.

Furthermore, factors related to the fungus itself and the environment play a role. “These organisms have enzymes, proteins, and other molecules that allow them to survive in the environment in which they normally inhabit. When they face a new and stressful one, they must express other molecules that will allow them to survive. All of this helps them evade elements of the immune system, antifungals, and, of course, body temperature,” according to Dr. Sánchez Paredes.

It is possible that climate change is also behind the noticeable increase in fungal infections and that this crisis may have an even greater impact in the future. The temperature of the environment has increased, and fungi will have to adapt to the planet’s temperature, to the point where body temperature may no longer be a significant barrier for them.

Environmental changes would also be responsible for modifications in the distribution of endemic mycoses, and it is believed that fungi will more frequently find new ecological niches, be able to survive in other environments, and alter distribution zones.

This is what is happening between Mexico and the United States with coccidioidomycosis, or valley fever. “We will begin to see cases of some mycoses where they were not normally seen, so we will have to conduct more studies to confirm that the fungus is inhabiting these new areas or is simply appearing in new sites owing to migration and the great mobility of populations,” Dr. Sánchez Paredes said.

Finally, exposure to environmental factors would partly be responsible for the increasing resistance to first-line antifungals observed in these microorganisms. This seems to be the case with A. fumigatus when exposed to azoles used as fungicides in agriculture.
 

One Health in Fungal Infections

The increasing resistance to antifungals is a clear testament that human, animal, and environmental health are interconnected. This is why a multidisciplinary approach that adopts the perspective of One Health is necessary for its management.

“The use of fungicides in agriculture, structurally similar to the azoles used in clinics, generates resistance in Aspergillus fumigatus found in the environment. These fungi in humans can be associated with infections that do not respond to first-line treatment,” explained Carlos Arturo Álvarez, an infectious diseases physician and professor at the Faculty of Medicine at the National University of Colombia.

According to Dr. Álvarez, the approach to control them should not only focus on the search for diagnostic methods that allow early detection of antifungal resistance or research on new antifungal treatments. He believes that progress must also be made with strategies that allow for the proper use of antifungals in agriculture.

“Unfortunately, the One Health approach is not yet well implemented in the region, and in my view, there is a lack of articulation in the different sectors. That is, there is a need for true coordination between government offices of agriculture, animal and human health, academia, and international organizations. This is not happening yet, and I believe we are in the initial stage of visibility,” Dr. Álvarez opined.

Veterinary public health is another pillar of the aforementioned approach. For various reasons, animals experience a higher frequency of fungal infections. A few carry and transmit true zoonoses that affect human health, but most often, animals act only as sentinels indicating a potential source of transmission.

Carolina Segundo Zaragoza, PhD, has worked in veterinary mycology for 30 years. She currently heads the veterinary mycology laboratory at the Animal Production Teaching, Research, and Extension Center in Altiplano, under the Faculty of Veterinary Medicine and Animal Husbandry at the National Autonomous University of Mexico. Because she has frequent contact with specialists in human mycology, during her professional career she has received several patient consultations, most of which were for cutaneous mycoses.

“They detect some dermatomycosis and realize that the common factor is owning a companion animal or a production animal with which the patient has contact. Both animals and humans present the same type of lesions, and then comes the question: Who infected whom? I remind them that the main source of infection is the soil and that animals should not be blamed in the first instance,” Dr. Segundo Zaragoza clarified.

She is currently collaborating on a research project analyzing the presence of Coccidioides immitis in the soil. This pathogen is responsible for coccidioidomycosis in dogs and humans, and she sees with satisfaction how these types of initiatives, which include some components of the One Health vision, are becoming more common in Mexico.

“Fortunately, human mycologists are increasingly providing more space for the dissemination of veterinary mycology. So I have had the opportunity to be invited to different forums on medical mycology to present the clinical cases we can have in animals and talk about the research projects we carry out. I have more and more opportunities to conduct joint research with human mycologists and veterinary doctors,” she said.

Dr. Segundo Zaragoza believes that to better implement the One Health vision, standardizing the criteria for detecting, diagnosing, and treating mycoses is necessary. She considers that teamwork will be key to achieving the common goal of safeguarding the well-being and health of humans and animals.
 

 

 

Alarms Sound for Candida auris

The WHO included the yeast Candida auris in its group of pathogens with critical priority, and since 2009, it has raised alarm owing to the ease with which it grows in hospitals. In that setting, C auris is known for its high transmissibility, its ability to cause outbreaks, and the high mortality rate from disseminated infections.

“It has been a concern for the mycological community because it shows resistance to most antifungals used clinically, mainly azoles, but also for causing epidemic outbreaks,” emphasized Dr. Sánchez Paredes.

Its mode of transmission is not very clear, but it has been documented to be present on the skin and persist in hospital materials and furniture. It causes nosocomial infections in critically ill patients, such as those in intensive care, and those with cancer or who have received a transplant.

Risk factors for its development include renal insufficiency, hospital stays of more than 15 days, mechanical ventilation, central lines, use of parenteral nutrition, and presence of sepsis.

As for other mycoses, there are no precise studies reporting global incidence rates, but the trend indicates an increase in the detection of outbreaks in various countries lately — something that began to be visible during the COVID-19 pandemic.

In Mexico, Dr. Treviño Rangel and colleagues from Nuevo León reported the first case of candidemia caused by this agent. It occurred in May 2020 and involved a 58-year-old woman with a history of severe endometriosis and multiple complications in the gastrointestinal tract. The patient’s condition improved favorably thanks to antifungal therapy with caspofungin and liposomal amphotericin B.

However, 3 months after that episode, the group reported an outbreak of C. auris at the same hospital in 12 critically ill patients co-infected with SARS-CoV-2. All were on mechanical ventilation, had peripherally inserted central catheters and urinary catheters, and had a prolonged hospital stay (20-70 days). The mortality in patients with candidemia in this cohort was 83.3%.
 

Open Ending

As seen in some science fiction series, fungal infections in the region still have an open ending, and Global Action For Fungal Infections (GAFFI) has estimated that with better diagnostics and treatments, deaths caused by fungi could decrease to less than 750,000 per year worldwide.

But if everything continues as is, some aspects of what is to come may resemble the dystopia depicted in The Last of Us. No zombies, but emerging and reemerging fungi in a chaotic distribution, and resistant to all established treatments.

“The risk factors of patients and their immune status, combined with the behavior of mycoses, bring a complicated scenario. But therapeutic failure resulting from multidrug resistance to antifungals could make it catastrophic,” Dr. Sánchez Paredes summarized.

At the moment, there are only four families of drugs capable of counteracting fungal infections — and as mentioned, some are already scarce in Latin America’s hospital pharmacies.

“Historically, fungal infections have been given less importance than those caused by viruses or bacteria. Even in some developed countries, the true extent of morbidity and mortality they present is unknown. This results in less investment in the development of new antifungal molecules because knowledge is lacking about the incidence and prevalence of these diseases,” Dr. Treviño Rangel pointed out.

He added that the main limitation for the development of new drugs is economic. “Unfortunately, not many pharmaceutical companies are willing to invest in the development of new antifungals, and there are no government programs specifically promoting and supporting research into new therapeutic options against these neglected diseases,” he asserted.

Development of vaccines to prevent fungal infections faces the same barriers. Although, according to Dr. Treviño Rangel, the difficulties are compounded by the great similarity between fungal cells and human cells. This makes it possible for harmful cross-reactivity to occur. In addition, because most severe fungal infections occur in individuals with immunosuppression, a vaccine would need to trigger an adequate immune response despite this issue.

Meanwhile, fungi quietly continue to do what they do best: resist and survive. For millions of years, they have mutated and adapted to new environments. Some theories even blame them for the extinction of dinosaurs and the subsequent rise of mammals. They exist on the edge of life and death, decomposing and creating. There is consensus that at the moment, it does not seem feasible for them to generate a pandemic like the one due to SARS-CoV-2, given their transmission mechanism. But who is willing to rule out that this may not happen in the long or medium term?

Dr. Sánchez Paredes, Dr. Treviño Rangel, Dr. Messina, Dr. Santiso, Dr. Álvarez, and Dr. Segundo Zaragoza have declared no relevant financial conflicts of interest. 
 

This story was translated from Medscape Spanish Edition using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication. A version of this article appeared on Medscape.com.

The principle of resilience and survival is crucial for medically significant fungi. These microorganisms are far from creating the postapocalyptic scenario depicted in TV series like The Last of Us, and much work is necessary to learn more about them. Accurate statistics on fungal infections, accompanied by clinical histories, simple laboratory tests, new antifungals, and a necessary One Health approach are lacking. 

The entomopathogenic fungus Ophiocordyceps unilateralis was made notorious by the TV series, but for now, it only manages to control the brains of some ants at will. Luckily, there are no signs that fungi affecting humans are inclined to create zombies.

What is clear is that the world belongs to the kingdom of fungi and that fungi are everywhere. There are already close to 150,000 described species, but millions remain to be discovered. They abound in decomposing organic matter, soil, or animal excrement, including that of bats and pigeons. Some fungi have even managed to find a home in hospitals. Lastly, we must not forget those that establish themselves in the human microbiome.

Given such diversity, it is legitimate to ask whether any of them could be capable of generating new pandemics. Could the forgotten Cryptococcus neoformansAspergillus fumigatus, or Histoplasma species, among others, trigger new health emergencies on the scale of the one generated by SARS-CoV-2?

We cannot forget that a coronavirus has already confirmed that reality can surpass fiction. However, Edith Sánchez Paredes, a biologist, doctor in biomedical sciences, and specialist in medical mycology, provided a reassuring response to Medscape Spanish Edition on this point.

“That would be very difficult to see because the way fungal infections are acquired is not from person to person, in most cases,” said Dr. Sánchez Paredes, from the Mycology Unit of the Faculty of Medicine at the National Autonomous University of Mexico.

Close to 300 species have already been classified as pathogenic in humans. Although the numbers are not precise and are increasing, it is estimated that around 1,500,000 people worldwide die each year of systemic fungal infections.

“However, it is important to emphasize that establishment of an infection depends not only on the causal agent. A crucial factor is the host, in this case, the human. Generally, these types of infections will develop in individuals with some deficiency in their immune system. The more deficient the immune response, the more likely a fungal infection may occur,” stated Dr. Sánchez Paredes.

The possibility of a pandemic like the one experienced with SARS-CoV-2 in the short term is remote, but the threat posed by fungal infections persists.

In 2022, the World Health Organization (WHO) defined a priority list of pathogenic fungi, with the aim of guiding actions to control them. It is mentioned there that invasive fungal diseases are on the rise worldwide, particularly in immunocompromised populations.

“Despite the growing concern, fungal infections receive very little attention and resources, leading to a paucity of quality data on fungal disease distribution and antifungal resistance patterns. Consequently, it is impossible to estimate their exact burden,” as stated in the document.

In line with this, an article published in Mycoses in 2022 concluded that fungal infections are neglected diseases in Latin America. Among other difficulties, deficiencies in access to tests such as polymerase chain reaction or serum detection of beta-1,3-D-glucan have been reported there.

In terms of treatments, most countries encounter problems with access to liposomal amphotericin B and new azoles, such as posaconazole and isavuconazole.

“Unfortunately, in Latin America, we suffer from a poor infrastructure for diagnosing fungal infections; likewise, we have limited access to antifungals available in the global market. What’s more, we lack reliable data on the epidemiology of fungal infections in the region, so many times governments are unaware of the true extent of the problem,” said Rogelio de Jesús Treviño Rangel, PhD, a medical microbiologist and expert in clinical mycology, professor, and researcher at the Faculty of Medicine of the Autonomous University of Nuevo León in Mexico.
 

 

 

Need for More Medical Mycology Training

Dr. Fernando Messina is a medical mycologist with the Mycology Unit of the Francisco Javier Muñiz Infectious Diseases Hospital in Buenos Aires, Argentina. He has noted an increase in the number of cases of cryptococcosishistoplasmosis, and aspergillosis in his daily practice.

“Particularly, pulmonary aspergillosis is steadily increasing. This is because many patients have structural lung alterations that favor the appearance of this mycosis. This is related to the increase in cases of tuberculosis and the rise in life expectancy of patients with chronic obstructive pulmonary disease or other pulmonary or systemic diseases,” Dr. Messina stated.

For Dr. Messina, the main obstacle in current clinical practice is the low level of awareness among nonspecialist physicians regarding the presence of systemic fungal infections, and because these infections are more common than realized, it is vital to consider fungal etiology before starting empirical antibiotic therapy.

“Health professionals usually do not think about mycoses because mycology occupies a very small space in medical education at universities. As the Venezuelan mycologist Gioconda Cunto de San Blas once said, ‘Mycology is the Cinderella of microbiology.’ To change this, we need to give more space to mycoses in undergraduate and postgraduate studies,” Dr. Messina asserted.

He added, “The main challenge is to train professionals with an emphasis on the clinical interpretation of cases. Current medicine has a strong trend toward molecular biology and the use of rapid diagnostic methods, without considering the clinical symptoms or the patient’s history. Determinations are very useful, but it is necessary to interpret the results.”

Dr. Messina sees it as unlikely in the short term for a pandemic to be caused by fungi, but if it were to occur, he believes it would happen in healthcare systems in regions that are not prepared in terms of infrastructure. However, as seen in the health emergency resulting from SARS-CoV-2, he thinks the impact would be mitigated by the performance of healthcare professionals.

“In general, we have the ability to adapt to any adverse situation or change — although it is clear that we need more doctors, biochemists, and microbiologists trained in mycology,” emphasized Dr. Messina.

More than 40 interns pass through Muñiz Hospital each year. They are doctors and biochemists from Argentina, other countries in the region, or even Europe, seeking to enhance their training in mycology. Regarding fungal infection laboratory work, the interest lies in learning to use traditional techniques and innovative molecular methods.

“Rapid diagnostic methods, especially the detection of circulating antigens, have marked a change in the prognosis of deep mycosis in immunocompromised hosts. The possibility of screening and monitoring in this group of patients is very important and has a great benefit,” said Gabriela Santiso, PhD, a biochemist and head of the Mycology Unit of the Francisco Javier Muñiz Infectious Diseases Hospital.

According to Dr. Santiso, the current landscape includes the ability to identify genus and species, which can help in understanding resistance to antifungals. Furthermore, conducting sensitivity tests to these drugs, using standardized commercial methods, also provides timely information for treatment.

But Dr. Santiso warns that Latin America is a vast region with great disparity in human and technological resources. Although most countries in the region have networks facilitating access to timely diagnosis, resources are generally more available in major urban centers.

This often clashes with the epidemiology of most fungal infections. “Let’s not forget that many fungal pathologies affect low-income people who have difficulties accessing health centers, which sometimes turns them into chronic diseases that are hard to treat,” Dr. Santiso pointed out.

In mycology laboratories, the biggest cost is incurred by new diagnostic tests, such as those allowing molecular identification. Conventional methods are not usually expensive, but they require time and effort to train human resources to handle them.

Because new methodologies are not always available or easily accessible throughout the region, Dr. Santiso recommended not neglecting traditional mycological techniques. “Molecular methods, rapid diagnostic methods, and conventional mycology techniques are complementary and not mutually exclusive tests. Continuous training and updating are needed in this area,” she emphasized.
 

 

 

Why Are Resistant Fungal Infections Becoming Increasingly Common?

The first barrier for fungi to cause infection in humans is body temperature; most of them cannot withstand 37 °C. However, they also struggle to evade the immune response that is activated when they try to enter the body. 

“We are normally exposed to many of these fungi, almost all the time, but if our immune system is adequate, it may not go beyond a mild infection, in most cases subclinical, which will resolve quickly,” Dr. Sánchez Paredes stated.

However, according to Dr. Sánchez Paredes, if the immune response is weak, “the fungus will have no trouble establishing itself in our organs. Some are even part of our microbiota, such as Candida albicans, which in the face of an imbalance or immunocompromise, can lead to serious infections.”

It is clear that the population at risk for immunosuppression has increased. According to the WHO, this is due to the high prevalence of such diseases as tuberculosis, cancer, and HIV infection, among others.

But the WHO also believes that the increase in fungal infections is related to greater population access to critical care units, invasive procedures, chemotherapy, or immunotherapy treatments.

Furthermore, factors related to the fungus itself and the environment play a role. “These organisms have enzymes, proteins, and other molecules that allow them to survive in the environment in which they normally inhabit. When they face a new and stressful one, they must express other molecules that will allow them to survive. All of this helps them evade elements of the immune system, antifungals, and, of course, body temperature,” according to Dr. Sánchez Paredes.

It is possible that climate change is also behind the noticeable increase in fungal infections and that this crisis may have an even greater impact in the future. The temperature of the environment has increased, and fungi will have to adapt to the planet’s temperature, to the point where body temperature may no longer be a significant barrier for them.

Environmental changes would also be responsible for modifications in the distribution of endemic mycoses, and it is believed that fungi will more frequently find new ecological niches, be able to survive in other environments, and alter distribution zones.

This is what is happening between Mexico and the United States with coccidioidomycosis, or valley fever. “We will begin to see cases of some mycoses where they were not normally seen, so we will have to conduct more studies to confirm that the fungus is inhabiting these new areas or is simply appearing in new sites owing to migration and the great mobility of populations,” Dr. Sánchez Paredes said.

Finally, exposure to environmental factors would partly be responsible for the increasing resistance to first-line antifungals observed in these microorganisms. This seems to be the case with A. fumigatus when exposed to azoles used as fungicides in agriculture.
 

One Health in Fungal Infections

The increasing resistance to antifungals is a clear testament that human, animal, and environmental health are interconnected. This is why a multidisciplinary approach that adopts the perspective of One Health is necessary for its management.

“The use of fungicides in agriculture, structurally similar to the azoles used in clinics, generates resistance in Aspergillus fumigatus found in the environment. These fungi in humans can be associated with infections that do not respond to first-line treatment,” explained Carlos Arturo Álvarez, an infectious diseases physician and professor at the Faculty of Medicine at the National University of Colombia.

According to Dr. Álvarez, the approach to control them should not only focus on the search for diagnostic methods that allow early detection of antifungal resistance or research on new antifungal treatments. He believes that progress must also be made with strategies that allow for the proper use of antifungals in agriculture.

“Unfortunately, the One Health approach is not yet well implemented in the region, and in my view, there is a lack of articulation in the different sectors. That is, there is a need for true coordination between government offices of agriculture, animal and human health, academia, and international organizations. This is not happening yet, and I believe we are in the initial stage of visibility,” Dr. Álvarez opined.

Veterinary public health is another pillar of the aforementioned approach. For various reasons, animals experience a higher frequency of fungal infections. A few carry and transmit true zoonoses that affect human health, but most often, animals act only as sentinels indicating a potential source of transmission.

Carolina Segundo Zaragoza, PhD, has worked in veterinary mycology for 30 years. She currently heads the veterinary mycology laboratory at the Animal Production Teaching, Research, and Extension Center in Altiplano, under the Faculty of Veterinary Medicine and Animal Husbandry at the National Autonomous University of Mexico. Because she has frequent contact with specialists in human mycology, during her professional career she has received several patient consultations, most of which were for cutaneous mycoses.

“They detect some dermatomycosis and realize that the common factor is owning a companion animal or a production animal with which the patient has contact. Both animals and humans present the same type of lesions, and then comes the question: Who infected whom? I remind them that the main source of infection is the soil and that animals should not be blamed in the first instance,” Dr. Segundo Zaragoza clarified.

She is currently collaborating on a research project analyzing the presence of Coccidioides immitis in the soil. This pathogen is responsible for coccidioidomycosis in dogs and humans, and she sees with satisfaction how these types of initiatives, which include some components of the One Health vision, are becoming more common in Mexico.

“Fortunately, human mycologists are increasingly providing more space for the dissemination of veterinary mycology. So I have had the opportunity to be invited to different forums on medical mycology to present the clinical cases we can have in animals and talk about the research projects we carry out. I have more and more opportunities to conduct joint research with human mycologists and veterinary doctors,” she said.

Dr. Segundo Zaragoza believes that to better implement the One Health vision, standardizing the criteria for detecting, diagnosing, and treating mycoses is necessary. She considers that teamwork will be key to achieving the common goal of safeguarding the well-being and health of humans and animals.
 

 

 

Alarms Sound for Candida auris

The WHO included the yeast Candida auris in its group of pathogens with critical priority, and since 2009, it has raised alarm owing to the ease with which it grows in hospitals. In that setting, C auris is known for its high transmissibility, its ability to cause outbreaks, and the high mortality rate from disseminated infections.

“It has been a concern for the mycological community because it shows resistance to most antifungals used clinically, mainly azoles, but also for causing epidemic outbreaks,” emphasized Dr. Sánchez Paredes.

Its mode of transmission is not very clear, but it has been documented to be present on the skin and persist in hospital materials and furniture. It causes nosocomial infections in critically ill patients, such as those in intensive care, and those with cancer or who have received a transplant.

Risk factors for its development include renal insufficiency, hospital stays of more than 15 days, mechanical ventilation, central lines, use of parenteral nutrition, and presence of sepsis.

As for other mycoses, there are no precise studies reporting global incidence rates, but the trend indicates an increase in the detection of outbreaks in various countries lately — something that began to be visible during the COVID-19 pandemic.

In Mexico, Dr. Treviño Rangel and colleagues from Nuevo León reported the first case of candidemia caused by this agent. It occurred in May 2020 and involved a 58-year-old woman with a history of severe endometriosis and multiple complications in the gastrointestinal tract. The patient’s condition improved favorably thanks to antifungal therapy with caspofungin and liposomal amphotericin B.

However, 3 months after that episode, the group reported an outbreak of C. auris at the same hospital in 12 critically ill patients co-infected with SARS-CoV-2. All were on mechanical ventilation, had peripherally inserted central catheters and urinary catheters, and had a prolonged hospital stay (20-70 days). The mortality in patients with candidemia in this cohort was 83.3%.
 

Open Ending

As seen in some science fiction series, fungal infections in the region still have an open ending, and Global Action For Fungal Infections (GAFFI) has estimated that with better diagnostics and treatments, deaths caused by fungi could decrease to less than 750,000 per year worldwide.

But if everything continues as is, some aspects of what is to come may resemble the dystopia depicted in The Last of Us. No zombies, but emerging and reemerging fungi in a chaotic distribution, and resistant to all established treatments.

“The risk factors of patients and their immune status, combined with the behavior of mycoses, bring a complicated scenario. But therapeutic failure resulting from multidrug resistance to antifungals could make it catastrophic,” Dr. Sánchez Paredes summarized.

At the moment, there are only four families of drugs capable of counteracting fungal infections — and as mentioned, some are already scarce in Latin America’s hospital pharmacies.

“Historically, fungal infections have been given less importance than those caused by viruses or bacteria. Even in some developed countries, the true extent of morbidity and mortality they present is unknown. This results in less investment in the development of new antifungal molecules because knowledge is lacking about the incidence and prevalence of these diseases,” Dr. Treviño Rangel pointed out.

He added that the main limitation for the development of new drugs is economic. “Unfortunately, not many pharmaceutical companies are willing to invest in the development of new antifungals, and there are no government programs specifically promoting and supporting research into new therapeutic options against these neglected diseases,” he asserted.

Development of vaccines to prevent fungal infections faces the same barriers. Although, according to Dr. Treviño Rangel, the difficulties are compounded by the great similarity between fungal cells and human cells. This makes it possible for harmful cross-reactivity to occur. In addition, because most severe fungal infections occur in individuals with immunosuppression, a vaccine would need to trigger an adequate immune response despite this issue.

Meanwhile, fungi quietly continue to do what they do best: resist and survive. For millions of years, they have mutated and adapted to new environments. Some theories even blame them for the extinction of dinosaurs and the subsequent rise of mammals. They exist on the edge of life and death, decomposing and creating. There is consensus that at the moment, it does not seem feasible for them to generate a pandemic like the one due to SARS-CoV-2, given their transmission mechanism. But who is willing to rule out that this may not happen in the long or medium term?

Dr. Sánchez Paredes, Dr. Treviño Rangel, Dr. Messina, Dr. Santiso, Dr. Álvarez, and Dr. Segundo Zaragoza have declared no relevant financial conflicts of interest. 
 

This story was translated from Medscape Spanish Edition using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication. A version of this article appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>168241</fileName> <TBEID>0C050576.SIG</TBEID> <TBUniqueIdentifier>MD_0C050576</TBUniqueIdentifier> <newsOrJournal>News</newsOrJournal> <publisherName>Frontline Medical Communications</publisherName> <storyname/> <articleType>2</articleType> <TBLocation>QC Done-All Pubs</TBLocation> <QCDate>20240530T151600</QCDate> <firstPublished>20240530T154542</firstPublished> <LastPublished>20240530T154542</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240530T154541</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Fernando Fuentes</byline> <bylineText>FERNANDO FUENTES, MD</bylineText> <bylineFull>FERNANDO FUENTES, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:imng"> <name>IMNG Medical Media</name> <rightsInfo> <copyrightHolder> <name>Frontline Medical News</name> </copyrightHolder> <copyrightNotice>Copyright (c) 2015 Frontline Medical News, a Frontline Medical Communications Inc. company. All rights reserved. This material may not be published, broadcast, copied, or otherwise reproduced or distributed without the prior written permission of Frontline Medical Communications Inc.</copyrightNotice> </rightsInfo> </provider> <abstract/> <metaDescription>The principle of resilience and survival is crucial for medically significant fungi. These microorganisms are far from creating the postapocalyptic scenario dep</metaDescription> <articlePDF/> <teaserImage/> <teaser>Fungal infections are generally not passed between individuals and immunocompetence makes most infections unlikely, but multidrug resistance could change the story.</teaser> <title>Could a Fungal Infection Cause a Future Pandemic?</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear/> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume/> <pubNumber/> <wireChannels/> <primaryCMSID/> <CMSIDs/> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>chph</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>skin</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>fp</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>idprac</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>im</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> </publications_g> <publications> <term>6</term> <term>13</term> <term>15</term> <term canonical="true">20</term> <term>21</term> </publications> <sections> <term>27980</term> <term canonical="true">39313</term> </sections> <topics> <term canonical="true">321</term> <term>204</term> <term>234</term> <term>284</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Could a Fungal Infection Cause a Future Pandemic?</title> <deck/> </itemMeta> <itemContent> <p>The principle of resilience and survival is crucial for medically significant fungi. These microorganisms are far from creating the <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/987442">postapocalyptic scenario</a></span> depicted in TV series like The Last of Us, and much work is necessary to learn more about them. Accurate statistics on fungal infections, accompanied by clinical histories, simple laboratory tests, new antifungals, and a necessary <span class="Hyperlink"><a href="https://www.who.int/health-topics/one-health#tab=tab_1">One Health</a> </span>approach are lacking. </p> <p>The entomopathogenic fungus <em>Ophiocordyceps unilaterali</em>s was made notorious by the TV series, but for now, it only manages to control the brains of some ants at will. Luckily, there are no signs that fungi affecting humans are inclined to create zombies.<br/><br/>What is clear is that the world belongs to the kingdom of fungi and that fungi are everywhere. There are already close to 150,000 described species, but millions remain to be discovered. They abound in decomposing organic matter, soil, or animal excrement, including that of bats and pigeons. Some fungi have even managed to find a home in hospitals. Lastly, we must not forget those that establish themselves in the human microbiome.<br/><br/>Given such diversity, it is legitimate to ask whether any of them could be capable of generating new pandemics. Could the forgotten <em>Cryptococcus neoformans</em>, <em>Aspergillus fumigatus</em>, or <em>Histoplasma species</em>, among others, trigger new health emergencies on the scale of the one generated by SARS-CoV-2?<br/><br/>We cannot forget that a coronavirus has already confirmed that reality can surpass fiction. However, Edith Sánchez Paredes, a biologist, doctor in biomedical sciences, and specialist in medical mycology, provided a reassuring response to Medscape Spanish Edition on this point.<br/><br/>“That would be very difficult to see because the way fungal infections are acquired is not from person to person, in most cases,” said Dr. Sánchez Paredes, from the Mycology Unit of the Faculty of Medicine at the National Autonomous University of Mexico.<br/><br/>Close to 300 species have already been classified as pathogenic in humans. Although the numbers are not precise and are increasing, it is estimated that around 1,500,000 people worldwide die each year of systemic fungal infections.<br/><br/>“However, it is important to emphasize that establishment of an infection depends not only on the causal agent. A crucial factor is the host, in this case, the human. Generally, these types of infections will develop in individuals with some deficiency in their immune system. The more deficient the immune response, the more likely a fungal infection may occur,” stated Dr. Sánchez Paredes.<br/><br/>The possibility of a pandemic like the one experienced with SARS-CoV-2 in the short term is remote, but the threat posed by fungal infections persists.<br/><br/>In 2022, the World Health Organization (WHO) defined a <span class="Hyperlink"><a href="https://www.who.int/publications/i/item/9789240060241">priority list</a></span> of <span class="Hyperlink"><a href="https://espanol.medscape.com/verarticulo/5910011">pathogenic fungi</a></span>, with the aim of guiding actions to control them. It is mentioned there that invasive fungal diseases are on the rise worldwide, particularly in <span class="Hyperlink">immunocompromised</span> populations.<br/><br/>“Despite the growing concern, fungal infections receive very little attention and resources, leading to a paucity of quality data on fungal disease distribution and antifungal resistance patterns. Consequently, it is impossible to estimate their exact burden,” as stated in the document.<br/><br/>In line with this, an <span class="Hyperlink"><a href="https://onlinelibrary.wiley.com/doi/10.1111/myc.13510">article published</a></span> in <em>Mycoses</em> in 2022 concluded that fungal infections are neglected diseases in Latin America. Among other difficulties, deficiencies in access to tests such as polymerase chain reaction or serum detection of beta-1,3-D-glucan have been reported there.<br/><br/>In terms of treatments, most countries encounter problems with access to <span class="Hyperlink">liposomal amphotericin B</span> and new azoles, such as <span class="Hyperlink"><a href="https://reference.medscape.com/drug/noxafil-posaconazole-342588">posaconazole</a></span> and <span class="Hyperlink"><a href="https://reference.medscape.com/drug/cresemba-isavuconazole-isavuconazonium-sulfate-999988">isavuconazole</a></span>.<br/><br/>“Unfortunately, in Latin America, we suffer from a poor infrastructure for diagnosing fungal infections; likewise, we have limited access to antifungals available in the global market. What’s more, we lack reliable data on the epidemiology of fungal infections in the region, so many times governments are unaware of the true extent of the problem,” said Rogelio de Jesús Treviño Rangel, PhD, a medical microbiologist and expert in clinical mycology, professor, and researcher at the Faculty of Medicine of the Autonomous University of Nuevo León in Mexico.<br/><br/></p> <h2>Need for More Medical Mycology Training</h2> <p>Dr. Fernando Messina is a medical mycologist with the Mycology Unit of the Francisco Javier Muñiz Infectious Diseases Hospital in Buenos Aires, Argentina. He has noted an increase in the number of cases of <span class="Hyperlink">cryptococcosis</span>, <span class="Hyperlink">histoplasmosis</span>, and <span class="Hyperlink">aspergillosis</span> in his daily practice.<br/><br/>“Particularly, pulmonary aspergillosis is steadily increasing. This is because many patients have structural lung alterations that favor the appearance of this mycosis. This is related to the increase in cases of <span class="Hyperlink">tuberculosis</span> and the rise in life expectancy of patients with <span class="Hyperlink">chronic obstructive pulmonary disease</span> or other pulmonary or systemic diseases,” Dr. Messina stated.<br/><br/>For Dr. Messina, the main obstacle in current clinical practice is the low level of awareness among nonspecialist physicians regarding the presence of systemic fungal infections, and because these infections are more common than realized, it is vital to consider fungal etiology before starting empirical antibiotic therapy.<br/><br/>“Health professionals usually do not think about mycoses because mycology occupies a very small space in medical education at universities. As the Venezuelan mycologist Gioconda Cunto de San Blas once said, ‘Mycology is the Cinderella of microbiology.’ To change this, we need to give more space to mycoses in undergraduate and postgraduate studies,” Dr. Messina asserted.<br/><br/>He added, “The main challenge is to train professionals with an emphasis on the clinical interpretation of cases. Current medicine has a strong trend toward molecular biology and the use of rapid diagnostic methods, without considering the clinical symptoms or the patient’s history. Determinations are very useful, but it is necessary to interpret the results.”<br/><br/>Dr. Messina sees it as unlikely in the short term for a pandemic to be caused by fungi, but if it were to occur, he believes it would happen in healthcare systems in regions that are not prepared in terms of infrastructure. However, as seen in the health emergency resulting from SARS-CoV-2, he thinks the impact would be mitigated by the performance of healthcare professionals.<br/><br/>“In general, we have the ability to adapt to any adverse situation or change — although it is clear that we need more doctors, biochemists, and microbiologists trained in mycology,” emphasized Dr. Messina.<br/><br/>More than 40 interns pass through Muñiz Hospital each year. They are doctors and biochemists from Argentina, other countries in the region, or even Europe, seeking to enhance their training in mycology. Regarding fungal infection laboratory work, the interest lies in learning to use traditional techniques and innovative molecular methods.<br/><br/>“Rapid diagnostic methods, especially the detection of circulating antigens, have marked a change in the prognosis of deep mycosis in immunocompromised hosts. The possibility of screening and monitoring in this group of patients is very important and has a great benefit,” said Gabriela Santiso, PhD, a biochemist and head of the Mycology Unit of the Francisco Javier Muñiz Infectious Diseases Hospital.<br/><br/>According to Dr. Santiso, the current landscape includes the ability to identify genus and species, which can help in understanding resistance to antifungals. Furthermore, conducting sensitivity tests to these drugs, using standardized commercial methods, also provides timely information for treatment.<br/><br/>But Dr. Santiso warns that Latin America is a vast region with great disparity in human and technological resources. Although most countries in the region have networks facilitating access to timely diagnosis, resources are generally more available in major urban centers.<br/><br/>This often clashes with the epidemiology of most fungal infections. “Let’s not forget that many fungal pathologies affect low-income people who have difficulties accessing health centers, which sometimes turns them into chronic diseases that are hard to treat,” Dr. Santiso pointed out.<br/><br/>In mycology laboratories, the biggest cost is incurred by new diagnostic tests, such as those allowing molecular identification. Conventional methods are not usually expensive, but they require time and effort to train human resources to handle them.<br/><br/>Because new methodologies are not always available or easily accessible throughout the region, Dr. Santiso recommended not neglecting traditional mycological techniques. “Molecular methods, rapid diagnostic methods, and conventional mycology techniques are complementary and not mutually exclusive tests. Continuous training and updating are needed in this area,” she emphasized.<br/><br/></p> <h2>Why Are Resistant Fungal Infections Becoming Increasingly Common?</h2> <p>The first barrier for fungi to cause infection in humans is body temperature; most of them cannot withstand 37 °C. However, they also struggle to evade the immune response that is activated when they try to enter the body. <br/><br/>“We are normally exposed to many of these fungi, almost all the time, but if our immune system is adequate, it may not go beyond a mild infection, in most cases subclinical, which will resolve quickly,” Dr. Sánchez Paredes stated.<br/><br/>However, according to Dr. Sánchez Paredes, if the immune response is weak, “the fungus will have no trouble establishing itself in our organs. Some are even part of our microbiota, such as <em>Candida albicans</em>, which in the face of an imbalance or immunocompromise, can lead to serious infections.”<br/><br/>It is clear that the population at risk for <span class="Hyperlink">immunosuppression</span> has increased. According to the WHO, this is due to the high prevalence of such diseases as tuberculosis, cancer, and <span class="Hyperlink">HIV infection</span>, among others.<br/><br/>But the WHO also believes that the increase in fungal infections is related to greater population access to critical care units, invasive procedures, chemotherapy, or immunotherapy treatments.<br/><br/>Furthermore, factors related to the fungus itself and the environment play a role. “These organisms have enzymes, proteins, and other molecules that allow them to survive in the environment in which they normally inhabit. When they face a new and stressful one, they must express other molecules that will allow them to survive. All of this helps them evade elements of the immune system, antifungals, and, of course, body temperature,” according to Dr. Sánchez Paredes.<br/><br/>It is possible that climate change is also behind the noticeable increase in fungal infections and that this crisis may have an even greater impact in the future. The temperature of the environment has increased, and fungi will have to adapt to the planet’s temperature, to the point where body temperature may no longer be a significant barrier for them.<br/><br/>Environmental changes would also be responsible for modifications in the distribution of endemic mycoses, and it is believed that fungi will more frequently find new ecological niches, be able to survive in other environments, and alter distribution zones.<br/><br/>This is what is happening between Mexico and the United States with <span class="Hyperlink">coccidioidomycosis</span>, or <span class="Hyperlink">valley fever</span>. “We will begin to see cases of some mycoses where they were not normally seen, so we will have to conduct more studies to confirm that the fungus is inhabiting these new areas or is simply appearing in new sites owing to migration and the great mobility of populations,” Dr. Sánchez Paredes said.<br/><br/>Finally, exposure to environmental factors would partly be responsible for the increasing resistance to first-line antifungals observed in these microorganisms. This seems to be the case with <em>A. fumigatus</em> when exposed to azoles used as fungicides in agriculture.<br/><br/></p> <h2>One Health in Fungal Infections</h2> <p>The increasing resistance to antifungals is a clear testament that human, animal, and environmental health are interconnected. This is why a <span class="Hyperlink"><a href="https://wires.onlinelibrary.wiley.com/doi/10.1002/wsbm.1610">multidisciplinary approach</a></span> that adopts the perspective of One Health is necessary for its management.<br/><br/>“The use of fungicides in agriculture, structurally similar to the azoles used in clinics, generates resistance in <em>Aspergillus fumigatus</em> found in the environment. These fungi in humans can be associated with infections that do not respond to first-line treatment,” explained Carlos Arturo Álvarez, an infectious diseases physician and professor at the Faculty of Medicine at the National University of Colombia.<br/><br/>According to Dr. Álvarez, the approach to control them should not only focus on the search for diagnostic methods that allow early detection of antifungal resistance or research on new antifungal treatments. He believes that progress must also be made with strategies that allow for the proper use of antifungals in agriculture.<br/><br/>“Unfortunately, the One Health approach is not yet well implemented in the region, and in my view, there is a lack of articulation in the different sectors. That is, there is a need for true coordination between government offices of agriculture, animal and human health, academia, and international organizations. This is not happening yet, and I believe we are in the initial stage of visibility,” Dr. Álvarez opined.<br/><br/>Veterinary public health is another pillar of the aforementioned approach. For various reasons, animals experience a higher frequency of fungal infections. A few carry and transmit true zoonoses that affect human health, but most often, animals act only as sentinels indicating a potential source of transmission.<br/><br/>Carolina Segundo Zaragoza, PhD, has worked in veterinary mycology for 30 years. She currently heads the veterinary mycology laboratory at the Animal Production Teaching, Research, and Extension Center in Altiplano, under the Faculty of Veterinary Medicine and Animal Husbandry at the National Autonomous University of Mexico. Because she has frequent contact with specialists in human mycology, during her professional career she has received several patient consultations, most of which were for cutaneous mycoses.<br/><br/>“They detect some dermatomycosis and realize that the common factor is owning a companion animal or a production animal with which the patient has contact. Both animals and humans present the same type of lesions, and then comes the question: Who infected whom? I remind them that the main source of infection is the soil and that animals should not be blamed in the first instance,” Dr. Segundo Zaragoza clarified.<br/><br/>She is currently collaborating on a research project analyzing the presence of Coccidioides immitis in the soil. This pathogen is responsible for coccidioidomycosis in dogs and humans, and she sees with satisfaction how these types of initiatives, which include some components of the One Health vision, are becoming more common in Mexico.<br/><br/>“Fortunately, human mycologists are increasingly providing more space for the dissemination of veterinary mycology. So I have had the opportunity to be invited to different forums on medical mycology to present the clinical cases we can have in animals and talk about the research projects we carry out. I have more and more opportunities to conduct joint research with human mycologists and veterinary doctors,” she said.<br/><br/>Dr. Segundo Zaragoza believes that to better implement the One Health vision, standardizing the criteria for detecting, diagnosing, and treating mycoses is necessary. She considers that teamwork will be key to achieving the common goal of safeguarding the well-being and health of humans and animals.<br/><br/></p> <h2>Alarms Sound for Candida auris</h2> <p>The WHO included the yeast <em>Candida auris</em> in its group of pathogens with critical priority, and since 2009, it has raised alarm owing to the ease with which it grows in hospitals. In that setting, <em>C auris</em> is known for its high transmissibility, its ability to cause outbreaks, and the high mortality rate from disseminated infections.<br/><br/>“It has been a concern for the mycological community because it shows resistance to most antifungals used clinically, mainly azoles, but also for causing epidemic outbreaks,” emphasized Dr. Sánchez Paredes.<br/><br/>Its mode of transmission is not very clear, but it has been documented to be present on the skin and persist in hospital materials and furniture. It causes nosocomial infections in critically ill patients, such as those in intensive care, and those with cancer or who have received a transplant.<br/><br/>Risk factors for its development include renal insufficiency, hospital stays of more than 15 days, <span class="Hyperlink">mechanical ventilation</span>, central lines, use of parenteral nutrition, and presence of <span class="Hyperlink">sepsis</span>.<br/><br/>As for other mycoses, there are no precise studies reporting global incidence rates, but the trend indicates an increase in the detection of outbreaks in various countries lately — something that began to be visible during the COVID-19 pandemic.<br/><br/>In Mexico, Dr. Treviño Rangel and colleagues from Nuevo León <span class="Hyperlink"><a href="https://link.springer.com/article/10.1007/s15010-020-01525-1">reported</a></span> the first case of candidemia caused by this agent. It occurred in May 2020 and involved a 58-year-old woman with a history of severe <span class="Hyperlink">endometriosis</span> and multiple complications in the gastrointestinal tract. The patient’s condition improved favorably thanks to antifungal therapy with <span class="Hyperlink"><a href="https://reference.medscape.com/drug/cancidas-caspofungin-342584">caspofungin</a></span> and liposomal amphotericin B.<br/><br/>However, 3 months after that episode, the group <span class="Hyperlink"><a href="https://www.clinicalmicrobiologyandinfection.com/article/S1198-743X(20)30790-4/fulltext">reported</a></span> an outbreak of <em>C. auris</em> at the same hospital in 12 critically ill patients co-infected with SARS-CoV-2. All were on mechanical ventilation, had peripherally inserted central catheters and urinary catheters, and had a prolonged hospital stay (20-70 days). The mortality in patients with candidemia in this cohort was 83.3%.<br/><br/></p> <h2>Open Ending</h2> <p>As seen in some science fiction series, fungal infections in the region still have an open ending, and Global Action For Fungal Infections (GAFFI) has estimated that with better diagnostics and treatments, deaths caused by fungi could decrease to less than 750,000 per year worldwide.<br/><br/>But if everything continues as is, some aspects of what is to come may resemble the dystopia depicted in The Last of Us. No zombies, but emerging and reemerging fungi in a chaotic distribution, and resistant to all established treatments.<br/><br/>“The risk factors of patients and their immune status, combined with the behavior of mycoses, bring a complicated scenario. But therapeutic failure resulting from multidrug resistance to antifungals could make it catastrophic,” Dr. Sánchez Paredes summarized.<br/><br/>At the moment, there are only four families of drugs capable of counteracting fungal infections — and as mentioned, some are already scarce in Latin America’s hospital pharmacies.<br/><br/>“Historically, fungal infections have been given less importance than those caused by viruses or bacteria. Even in some developed countries, the true extent of morbidity and mortality they present is unknown. This results in less investment in the development of new antifungal molecules because knowledge is lacking about the incidence and prevalence of these diseases,” Dr. Treviño Rangel pointed out.<br/><br/>He added that the main limitation for the development of new drugs is economic. “Unfortunately, not many pharmaceutical companies are willing to invest in the development of new antifungals, and there are no government programs specifically promoting and supporting research into new therapeutic options against these neglected diseases,” he asserted.<br/><br/>Development of vaccines to prevent fungal infections faces the same barriers. Although, according to Dr. Treviño Rangel, the difficulties are compounded by the great similarity between fungal cells and human cells. This makes it possible for harmful cross-reactivity to occur. In addition, because most severe fungal infections occur in individuals with immunosuppression, a vaccine would need to trigger an adequate immune response despite this issue.<br/><br/>Meanwhile, fungi quietly continue to do what they do best: resist and survive. For millions of years, they have mutated and adapted to new environments. Some theories even blame them for the extinction of dinosaurs and the subsequent rise of mammals. They exist on the edge of life and death, decomposing and creating. There is consensus that at the moment, it does not seem feasible for them to generate a pandemic like the one due to SARS-CoV-2, given their transmission mechanism. But who is willing to rule out that this may not happen in the long or medium term?<br/><br/>Dr. Sánchez Paredes, Dr. Treviño Rangel, Dr. Messina, Dr. Santiso, Dr. Álvarez, and Dr. Segundo Zaragoza have declared no relevant financial conflicts of interest. <br/><br/></p> <p> <em>This story was translated from <span class="Hyperlink"><a href="https://espanol.medscape.com/verarticulo/5912421">Medscape Spanish Edition</a></span> using several editorial tools, including AI, as part of the process. Human editors reviewed this content before publication. A version of this article appeared on <span class="Hyperlink"><a href="https://www.medscape.com/viewarticle/could-fungal-infection-cause-future-pandemic-2024a1000a39">Medscape.com</a></span>.</em> </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>teaser</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> </itemContent> </newsItem> </itemSet></root>
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

Multiple Asymptomatic Dome-Shaped Papules on the Scalp

Article Type
Changed
Tue, 05/07/2024 - 14:09
Display Headline
Multiple Asymptomatic Dome-Shaped Papules on the Scalp

The Diagnosis: Spiradenocylindroma

Shave biopsies of our patient’s lesions showed wellcircumscribed dermal nodules resembling a spiradenoma with 3 cell populations: those with lighter nuclei, darker nuclei, and scattered lymphocytes. However, the conspicuous globules of basement membrane material were reminiscent of a cylindroma. These overlapping features and the patient’s history of cylindroma were suggestive of a diagnosis of spiradenocylindroma.

Spiradenocylindroma is an uncommon dermal tumor with features that overlap with spiradenoma and cylindroma.1 It may manifest as a solitary lesion or multiple lesions and can occur sporadically or in the context of a family history. Histologically, it must be distinguished from other intradermal basaloid neoplasms including conventional cylindroma and spiradenoma, dermal duct tumor, hidradenoma, and trichoblastoma.

When patients present with multiple cylindromas, spiradenomas, or spiradenocylindromas, physicians should consider genetic testing and review of the family history to assess for cylindromatosis gene mutations or Brooke-Spiegler syndrome. Biopsy and histologic examination are important because malignant tumors can evolve from pre-existing spiradenocylindromas, cylindromas, and spiradenomas,2 with an increased risk in patients with Brooke-Spiegler syndrome.1 Our patient declined further genetic workup but continues to follow up with dermatology for monitoring of lesions.

Dermal duct tumors are morphologic variants of poromas that are derived from sweat gland lineage and usually manifest as solitary dome-shaped papules, plaques, or nodules most often seen on acral surfaces as well as the head and neck.3 Clinically, they may be indistinguishable from spiradenocylindromas and require biopsy for histologic evaluation. They can be distinguished from spiradenocylindroma by the presence of small dermal nodules composed of cuboidal cells with ample pink cytoplasm and cuticle-lined ducts (Figure 1).

Charest_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Dermal%20duct%20tumors%20have%20a%20relatively%20eosinophilic%20appearance%20due%20to%20their%20cuboidal%20poroid%20cells%20that%20resemble%20epidermal%20keratinocytes%20punctuated%20by%20cuticle-lined%20ducts%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

Trichoblastomas typically are deep-seated basaloid follicular neoplasms on the scalp with papillary mesenchyme resembling the normal fibrous sheath of the hair follicle, often replete with papillary mesenchymal bodies (Figure 2). There generally are no retraction spaces between its basaloid nests and the surrounding stroma, which is unlikely to contain mucin relative to basal cell carcinoma (BCC).4,5

Charest_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Trichoblastomas%20have%20germinative%20follicular%20basaloid%20cells%20that%20closely%20resemble%20those%20of%20basal%20cell%20carcinoma%20but%20are%20deepseated%20tumors%20with%20papillary%20mesenchyme%20and%20less%20stromal%20mucin%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%2C%20original%20magnification%20%C3%9740).%3C%2Fp%3E

Adenoid cystic carcinoma is a rare salivary gland tumor that can metastasize to the skin and rarely arises as a primary skin adnexal tumor. It manifests as a slowgrowing mass that can be tender to palpation.6 Histologic examination shows dermal islands with cribriform blue and pink spaces. Compared to BCC, adenoid cystic carcinoma cells are enlarged and epithelioid with relatively scarce cytoplasm (Figure 3).6,7 Adenoid cystic carcinoma can show variable growth patterns including infiltrative nests and trabeculae. Perineural invasion is common, and there is a high risk for local recurrence.7 First-line therapy usually is surgical, and postoperative radiotherapy may be required.6,7

Charest_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Adenoid%20cystic%20carcinoma%20is%20composed%20of%20cribriform%20nests%20and%20irregular%20infiltrative%20trabeculae.%20Its%20two%20constituent%20cell%20populations%20have%20large%20ovoid%20nuclei%20with%20variable%20hyperchromasia.%20No%20papillary%20mesenchyme%2C%20mucinous%20stroma%2C%20palisading%2C%20or%20clefting%20is%20present%20at%20the%20perimeter%20of%20infiltrative%20nests%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

Nodular BCC commonly manifests as an enlarging nonhealing lesion on sun-exposed skin and has many subtypes, typically with arborizing telangiectases on dermoscopy. Histopathologic examination of nodular BCC reveals a nest of basaloid follicular germinative cells in the dermis with peripheral palisading and a fibromyxoid stroma (Figure 4).8 Patients with Brooke-Spiegler syndrome are at increased risk for nodular BCC, which may be clinically indistinguishable from spiradenoma, cylindroma, and spiradenocylindroma, necessitating histologic assessment.

Charest_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Basal%20cell%20carcinoma%20with%20nodular%20morphology%20is%20composed%20of%20germinative%20follicular%20basaloid%20cells%20with%20peripheral%20palisading%20and%20clefts%20filled%20with%20mucin%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

References
  1. Facchini V, Colangeli W, Bozza F, et al. A rare histopathological spiradenocylindroma: a case report. Clin Ter. 2022;173:292-294. doi:10.7417/ CT.2022.2433
  2. Kazakov DV. Brooke-Spiegler syndrome and phenotypic variants: an update [published online March 14, 2016]. Head Neck Pathol. 2016;10:125-30. doi:10.1007/s12105-016-0705-x
  3. Miller AC, Adjei S, Temiz LA, et al. Dermal duct tumor: a diagnostic dilemma. Dermatopathology (Basel). 2022;9:36-47. doi:10.3390/dermatopathology9010007
  4. Elston DM. Pilar and sebaceous neoplasms. In: Elston DM, Ferringer T, Ko C, et al. Dermatopathology. 3rd ed. Elsevier; 2018:71-85.
  5. McCalmont TH, Pincus LB. Adnexal neoplasms. In: Bolognia J, Schaffer J, Cerroni, L. Dermatology. 4th ed. Elsevier; 2017:1930-1953.
  6. Coca-Pelaz A, Rodrigo JP, Bradley PJ, et al. Adenoid cystic carcinoma of the head and neck—an update [published online May 2, 2015]. Oral Oncol. 2015;51:652-661. doi:10.1016/j.oraloncology.2015.04.005
  7. Tonev ID, Pirgova YS, Conev NV. Primary adenoid cystic carcinoma of the skin with multiple local recurrences. Case Rep Oncol. 2015;8:251- 255. doi:10.1159/000431082
  8. Cameron MC, Lee E, Hibler BP, et al. Basal cell carcinoma: epidemiology; pathophysiology; clinical and histological subtypes; and disease associations [published online May 18, 2018]. J Am Acad Dermatol. 2019;80:303-317. doi:10.1016/j.jaad.2018.03.060
Article PDF
Author and Disclosure Information

Drs. Charest, Chandra, and Hamann are from HonorHealth Dermatology Residency, Scottsdale, Arizona. Dr. Hamann also is from The Contact Dermatitis Institute, Phoenix, Arizona. Dr. Hamann also is from and Dr. Salem is from the Department of Dermatology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire. Dr. LeBlanc is from the Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center.

The authors report no conflict of interest.

Correspondence: Guy Charest, DO, HonorHealth Dermatology Residency, 7400 E Thompson Peak Pkwy, Scottsdale, AZ 85255 (gucharest@honorhealth.com).

Issue
Cutis - 113(5)
Publications
Topics
Page Number
210,215
Sections
Author and Disclosure Information

Drs. Charest, Chandra, and Hamann are from HonorHealth Dermatology Residency, Scottsdale, Arizona. Dr. Hamann also is from The Contact Dermatitis Institute, Phoenix, Arizona. Dr. Hamann also is from and Dr. Salem is from the Department of Dermatology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire. Dr. LeBlanc is from the Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center.

The authors report no conflict of interest.

Correspondence: Guy Charest, DO, HonorHealth Dermatology Residency, 7400 E Thompson Peak Pkwy, Scottsdale, AZ 85255 (gucharest@honorhealth.com).

Author and Disclosure Information

Drs. Charest, Chandra, and Hamann are from HonorHealth Dermatology Residency, Scottsdale, Arizona. Dr. Hamann also is from The Contact Dermatitis Institute, Phoenix, Arizona. Dr. Hamann also is from and Dr. Salem is from the Department of Dermatology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire. Dr. LeBlanc is from the Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center.

The authors report no conflict of interest.

Correspondence: Guy Charest, DO, HonorHealth Dermatology Residency, 7400 E Thompson Peak Pkwy, Scottsdale, AZ 85255 (gucharest@honorhealth.com).

Article PDF
Article PDF
Related Articles

The Diagnosis: Spiradenocylindroma

Shave biopsies of our patient’s lesions showed wellcircumscribed dermal nodules resembling a spiradenoma with 3 cell populations: those with lighter nuclei, darker nuclei, and scattered lymphocytes. However, the conspicuous globules of basement membrane material were reminiscent of a cylindroma. These overlapping features and the patient’s history of cylindroma were suggestive of a diagnosis of spiradenocylindroma.

Spiradenocylindroma is an uncommon dermal tumor with features that overlap with spiradenoma and cylindroma.1 It may manifest as a solitary lesion or multiple lesions and can occur sporadically or in the context of a family history. Histologically, it must be distinguished from other intradermal basaloid neoplasms including conventional cylindroma and spiradenoma, dermal duct tumor, hidradenoma, and trichoblastoma.

When patients present with multiple cylindromas, spiradenomas, or spiradenocylindromas, physicians should consider genetic testing and review of the family history to assess for cylindromatosis gene mutations or Brooke-Spiegler syndrome. Biopsy and histologic examination are important because malignant tumors can evolve from pre-existing spiradenocylindromas, cylindromas, and spiradenomas,2 with an increased risk in patients with Brooke-Spiegler syndrome.1 Our patient declined further genetic workup but continues to follow up with dermatology for monitoring of lesions.

Dermal duct tumors are morphologic variants of poromas that are derived from sweat gland lineage and usually manifest as solitary dome-shaped papules, plaques, or nodules most often seen on acral surfaces as well as the head and neck.3 Clinically, they may be indistinguishable from spiradenocylindromas and require biopsy for histologic evaluation. They can be distinguished from spiradenocylindroma by the presence of small dermal nodules composed of cuboidal cells with ample pink cytoplasm and cuticle-lined ducts (Figure 1).

Charest_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Dermal%20duct%20tumors%20have%20a%20relatively%20eosinophilic%20appearance%20due%20to%20their%20cuboidal%20poroid%20cells%20that%20resemble%20epidermal%20keratinocytes%20punctuated%20by%20cuticle-lined%20ducts%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

Trichoblastomas typically are deep-seated basaloid follicular neoplasms on the scalp with papillary mesenchyme resembling the normal fibrous sheath of the hair follicle, often replete with papillary mesenchymal bodies (Figure 2). There generally are no retraction spaces between its basaloid nests and the surrounding stroma, which is unlikely to contain mucin relative to basal cell carcinoma (BCC).4,5

Charest_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Trichoblastomas%20have%20germinative%20follicular%20basaloid%20cells%20that%20closely%20resemble%20those%20of%20basal%20cell%20carcinoma%20but%20are%20deepseated%20tumors%20with%20papillary%20mesenchyme%20and%20less%20stromal%20mucin%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%2C%20original%20magnification%20%C3%9740).%3C%2Fp%3E

Adenoid cystic carcinoma is a rare salivary gland tumor that can metastasize to the skin and rarely arises as a primary skin adnexal tumor. It manifests as a slowgrowing mass that can be tender to palpation.6 Histologic examination shows dermal islands with cribriform blue and pink spaces. Compared to BCC, adenoid cystic carcinoma cells are enlarged and epithelioid with relatively scarce cytoplasm (Figure 3).6,7 Adenoid cystic carcinoma can show variable growth patterns including infiltrative nests and trabeculae. Perineural invasion is common, and there is a high risk for local recurrence.7 First-line therapy usually is surgical, and postoperative radiotherapy may be required.6,7

Charest_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Adenoid%20cystic%20carcinoma%20is%20composed%20of%20cribriform%20nests%20and%20irregular%20infiltrative%20trabeculae.%20Its%20two%20constituent%20cell%20populations%20have%20large%20ovoid%20nuclei%20with%20variable%20hyperchromasia.%20No%20papillary%20mesenchyme%2C%20mucinous%20stroma%2C%20palisading%2C%20or%20clefting%20is%20present%20at%20the%20perimeter%20of%20infiltrative%20nests%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

Nodular BCC commonly manifests as an enlarging nonhealing lesion on sun-exposed skin and has many subtypes, typically with arborizing telangiectases on dermoscopy. Histopathologic examination of nodular BCC reveals a nest of basaloid follicular germinative cells in the dermis with peripheral palisading and a fibromyxoid stroma (Figure 4).8 Patients with Brooke-Spiegler syndrome are at increased risk for nodular BCC, which may be clinically indistinguishable from spiradenoma, cylindroma, and spiradenocylindroma, necessitating histologic assessment.

Charest_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Basal%20cell%20carcinoma%20with%20nodular%20morphology%20is%20composed%20of%20germinative%20follicular%20basaloid%20cells%20with%20peripheral%20palisading%20and%20clefts%20filled%20with%20mucin%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

The Diagnosis: Spiradenocylindroma

Shave biopsies of our patient’s lesions showed wellcircumscribed dermal nodules resembling a spiradenoma with 3 cell populations: those with lighter nuclei, darker nuclei, and scattered lymphocytes. However, the conspicuous globules of basement membrane material were reminiscent of a cylindroma. These overlapping features and the patient’s history of cylindroma were suggestive of a diagnosis of spiradenocylindroma.

Spiradenocylindroma is an uncommon dermal tumor with features that overlap with spiradenoma and cylindroma.1 It may manifest as a solitary lesion or multiple lesions and can occur sporadically or in the context of a family history. Histologically, it must be distinguished from other intradermal basaloid neoplasms including conventional cylindroma and spiradenoma, dermal duct tumor, hidradenoma, and trichoblastoma.

When patients present with multiple cylindromas, spiradenomas, or spiradenocylindromas, physicians should consider genetic testing and review of the family history to assess for cylindromatosis gene mutations or Brooke-Spiegler syndrome. Biopsy and histologic examination are important because malignant tumors can evolve from pre-existing spiradenocylindromas, cylindromas, and spiradenomas,2 with an increased risk in patients with Brooke-Spiegler syndrome.1 Our patient declined further genetic workup but continues to follow up with dermatology for monitoring of lesions.

Dermal duct tumors are morphologic variants of poromas that are derived from sweat gland lineage and usually manifest as solitary dome-shaped papules, plaques, or nodules most often seen on acral surfaces as well as the head and neck.3 Clinically, they may be indistinguishable from spiradenocylindromas and require biopsy for histologic evaluation. They can be distinguished from spiradenocylindroma by the presence of small dermal nodules composed of cuboidal cells with ample pink cytoplasm and cuticle-lined ducts (Figure 1).

Charest_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Dermal%20duct%20tumors%20have%20a%20relatively%20eosinophilic%20appearance%20due%20to%20their%20cuboidal%20poroid%20cells%20that%20resemble%20epidermal%20keratinocytes%20punctuated%20by%20cuticle-lined%20ducts%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

Trichoblastomas typically are deep-seated basaloid follicular neoplasms on the scalp with papillary mesenchyme resembling the normal fibrous sheath of the hair follicle, often replete with papillary mesenchymal bodies (Figure 2). There generally are no retraction spaces between its basaloid nests and the surrounding stroma, which is unlikely to contain mucin relative to basal cell carcinoma (BCC).4,5

Charest_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Trichoblastomas%20have%20germinative%20follicular%20basaloid%20cells%20that%20closely%20resemble%20those%20of%20basal%20cell%20carcinoma%20but%20are%20deepseated%20tumors%20with%20papillary%20mesenchyme%20and%20less%20stromal%20mucin%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%2C%20original%20magnification%20%C3%9740).%3C%2Fp%3E

Adenoid cystic carcinoma is a rare salivary gland tumor that can metastasize to the skin and rarely arises as a primary skin adnexal tumor. It manifests as a slowgrowing mass that can be tender to palpation.6 Histologic examination shows dermal islands with cribriform blue and pink spaces. Compared to BCC, adenoid cystic carcinoma cells are enlarged and epithelioid with relatively scarce cytoplasm (Figure 3).6,7 Adenoid cystic carcinoma can show variable growth patterns including infiltrative nests and trabeculae. Perineural invasion is common, and there is a high risk for local recurrence.7 First-line therapy usually is surgical, and postoperative radiotherapy may be required.6,7

Charest_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Adenoid%20cystic%20carcinoma%20is%20composed%20of%20cribriform%20nests%20and%20irregular%20infiltrative%20trabeculae.%20Its%20two%20constituent%20cell%20populations%20have%20large%20ovoid%20nuclei%20with%20variable%20hyperchromasia.%20No%20papillary%20mesenchyme%2C%20mucinous%20stroma%2C%20palisading%2C%20or%20clefting%20is%20present%20at%20the%20perimeter%20of%20infiltrative%20nests%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

Nodular BCC commonly manifests as an enlarging nonhealing lesion on sun-exposed skin and has many subtypes, typically with arborizing telangiectases on dermoscopy. Histopathologic examination of nodular BCC reveals a nest of basaloid follicular germinative cells in the dermis with peripheral palisading and a fibromyxoid stroma (Figure 4).8 Patients with Brooke-Spiegler syndrome are at increased risk for nodular BCC, which may be clinically indistinguishable from spiradenoma, cylindroma, and spiradenocylindroma, necessitating histologic assessment.

Charest_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Basal%20cell%20carcinoma%20with%20nodular%20morphology%20is%20composed%20of%20germinative%20follicular%20basaloid%20cells%20with%20peripheral%20palisading%20and%20clefts%20filled%20with%20mucin%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

References
  1. Facchini V, Colangeli W, Bozza F, et al. A rare histopathological spiradenocylindroma: a case report. Clin Ter. 2022;173:292-294. doi:10.7417/ CT.2022.2433
  2. Kazakov DV. Brooke-Spiegler syndrome and phenotypic variants: an update [published online March 14, 2016]. Head Neck Pathol. 2016;10:125-30. doi:10.1007/s12105-016-0705-x
  3. Miller AC, Adjei S, Temiz LA, et al. Dermal duct tumor: a diagnostic dilemma. Dermatopathology (Basel). 2022;9:36-47. doi:10.3390/dermatopathology9010007
  4. Elston DM. Pilar and sebaceous neoplasms. In: Elston DM, Ferringer T, Ko C, et al. Dermatopathology. 3rd ed. Elsevier; 2018:71-85.
  5. McCalmont TH, Pincus LB. Adnexal neoplasms. In: Bolognia J, Schaffer J, Cerroni, L. Dermatology. 4th ed. Elsevier; 2017:1930-1953.
  6. Coca-Pelaz A, Rodrigo JP, Bradley PJ, et al. Adenoid cystic carcinoma of the head and neck—an update [published online May 2, 2015]. Oral Oncol. 2015;51:652-661. doi:10.1016/j.oraloncology.2015.04.005
  7. Tonev ID, Pirgova YS, Conev NV. Primary adenoid cystic carcinoma of the skin with multiple local recurrences. Case Rep Oncol. 2015;8:251- 255. doi:10.1159/000431082
  8. Cameron MC, Lee E, Hibler BP, et al. Basal cell carcinoma: epidemiology; pathophysiology; clinical and histological subtypes; and disease associations [published online May 18, 2018]. J Am Acad Dermatol. 2019;80:303-317. doi:10.1016/j.jaad.2018.03.060
References
  1. Facchini V, Colangeli W, Bozza F, et al. A rare histopathological spiradenocylindroma: a case report. Clin Ter. 2022;173:292-294. doi:10.7417/ CT.2022.2433
  2. Kazakov DV. Brooke-Spiegler syndrome and phenotypic variants: an update [published online March 14, 2016]. Head Neck Pathol. 2016;10:125-30. doi:10.1007/s12105-016-0705-x
  3. Miller AC, Adjei S, Temiz LA, et al. Dermal duct tumor: a diagnostic dilemma. Dermatopathology (Basel). 2022;9:36-47. doi:10.3390/dermatopathology9010007
  4. Elston DM. Pilar and sebaceous neoplasms. In: Elston DM, Ferringer T, Ko C, et al. Dermatopathology. 3rd ed. Elsevier; 2018:71-85.
  5. McCalmont TH, Pincus LB. Adnexal neoplasms. In: Bolognia J, Schaffer J, Cerroni, L. Dermatology. 4th ed. Elsevier; 2017:1930-1953.
  6. Coca-Pelaz A, Rodrigo JP, Bradley PJ, et al. Adenoid cystic carcinoma of the head and neck—an update [published online May 2, 2015]. Oral Oncol. 2015;51:652-661. doi:10.1016/j.oraloncology.2015.04.005
  7. Tonev ID, Pirgova YS, Conev NV. Primary adenoid cystic carcinoma of the skin with multiple local recurrences. Case Rep Oncol. 2015;8:251- 255. doi:10.1159/000431082
  8. Cameron MC, Lee E, Hibler BP, et al. Basal cell carcinoma: epidemiology; pathophysiology; clinical and histological subtypes; and disease associations [published online May 18, 2018]. J Am Acad Dermatol. 2019;80:303-317. doi:10.1016/j.jaad.2018.03.060
Issue
Cutis - 113(5)
Issue
Cutis - 113(5)
Page Number
210,215
Page Number
210,215
Publications
Publications
Topics
Article Type
Display Headline
Multiple Asymptomatic Dome-Shaped Papules on the Scalp
Display Headline
Multiple Asymptomatic Dome-Shaped Papules on the Scalp
Sections
Questionnaire Body

A 62-year-old man with a history of cylindromas presented to our clinic with multiple asymptomatic, 3- to 4-mm, nonmobile, dome-shaped, telangiectatic, pink papules over the parietal and vertex scalp that had been present for more than 10 years without change. Several family members had similar lesions that had not been evaluated by a physician, and there had been no genetic evaluation. Shave biopsies of several lesions were performed.

CT113005210_Quiz_top_bottom.jpg
%3Cp%3EH%26amp%3BE%2C%20original%20magnifications%20%C3%9740%20and%20%C3%97100%2C%20respectively.%3C%2Fp%3E

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 05/07/2024 - 09:15
Un-Gate On Date
Tue, 05/07/2024 - 09:15
Use ProPublica
CFC Schedule Remove Status
Tue, 05/07/2024 - 09:15
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
Image
Disable zoom
Off

Clinical Manifestation of Degos Disease: Painful Penile Ulcers

Article Type
Changed
Thu, 05/09/2024 - 14:12
Display Headline
Clinical Manifestation of Degos Disease: Painful Penile Ulcers

To the Editor:

A 56-year-old man was referred to our Grand Rounds by another dermatologist in our health system for evaluation of a red scaly rash on the trunk that had been present for more than a year. More recently, over the course of approximately 9 months he experienced recurrent painful penile ulcers that lasted for approximately 4 weeks and then self-resolved. He had a medical history of central retinal vein occlusion, primary hyperparathyroidism, and nonspecific colitis. A family history was notable for lung cancer in the patient’s father and myelodysplastic syndrome and breast cancer in his mother; however, there was no family history of a similar rash. A bacterial culture of the penile ulcer was negative. Testing for antibodies against HIV and herpes simplex virus (HSV) types 1 and 2 was negative. Results of a serum VDRL test were nonreactive, which ruled out syphilis. The patient was treated by the referring dermatologist with azithromycin for possible chancroid without relief.

The patient was being followed by the referring dermatologist who initially was concerned for Degos disease based on clinical examination findings, prompting biopsy of a lesion on the back, which revealed vacuolar interface dermatitis, a sparse superficial perivascular lymphocytic infiltrate, and increased mucin—all highly suspicious for connective tissue disease (Figure 1). An antinuclear antibody test was positive, with a titer of 1:640. The patient was started on prednisone and referred to rheumatology; however, further evaluation by rheumatology for an autoimmune process—including anticardiolipin antibodies—was unremarkable. A few months prior to the current presentation, he also had mildly elevated liver function test results. A colonoscopy was performed, and a biopsy revealed nonspecific colitis. A biopsy of the penile ulcer also was nonspecific, showing only ulceration and acute and chronic inflammation. No epidermal interface change was seen. Results from a Grocott-Gomori methenamine-silver stain, Treponema pallidum immunostain, and HSV polymerase chain reaction were negative for fungal organisms, spirochetes, and HSV, respectively. The differential diagnosis included trauma, aphthous ulceration, and Behçet disease. Behçet disease was suspected by the referring dermatologist, and the patient was treated with colchicine, prednisone, pimecrolimus cream, and topical lidocaine; however, the lesions persisted, and he was subsequently referred to our Grand Rounds for further evaluation.

CT113004033_e_Fig1_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%2C%20Initial%20biopsy%20of%20the%20back%20showed%20vacuolar%20interface%20dermatitis%20and%20a%20sparse%20superficial%20perivascular%20lymphocytic%20infiltrate%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%20B%2C%20Alcian%20blue%20stain%20highlighted%20increased%20dermal%20mucin%20(original%20magnification%20%C3%97100).%20Photographs%20courtesy%20of%20Jennifer%20Leininger%2C%20MD%20(Cedar%20Park%2C%20Texas).%3C%2Fp%3E

At the current presentation, physical examination revealed several small papules with white atrophic centers and erythematous rims on the trunk and extremities (Figure 2A). An ulceration was noted on the penile shaft (Figure 2B). Further evaluation for Behçet disease, including testing for pathergy and HLA-B51, was negative. Degos disease was strongly suspected clinically, and a repeat biopsy was performed of a lesion on the abdomen, which revealed central epidermal necrosis, atrophy, and parakeratosis with an underlying wedge-shaped dermal infarct surrounded by multiple small occluded dermal vessels, perivascular inflammation, and dermal edema (Figure 3). Direct immunofluorescence was performed using antibodies against IgG, IgA, IgM, fibrinogen, albumin, and C3, which was negative. These findings from direct immunofluorescence and histopathology as well as the clinical presentation were considered compatible with Degos disease. The patient was started on aspirin and pentoxifylline. Pentoxifylline 400 mg twice daily appeared to lessen some of the pain. Pain management specialists started the patient on gabapentin.

CT113004033_e_Fig2_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Degos%20disease.%20A%2C%20Papules%20with%20white%20atrophic%20centers%20and%20erythematous%20rims%20on%20the%20anterior%20trunk.%20B%2C%20An%20ulceration%20on%20the%20penile%20shaft.%3C%2Fp%3E

Approximately 4 months after the Grand Rounds evaluation, during which time he continued treatment with pentoxifylline, he was admitted to the hospital for intractable nausea and vomiting. His condition acutely declined due to bowel perforation, and he was started on eculizumab 1200 mg every 14 days. Because of an increased risk for meningococcal meningitis while on this medication, he also was given erythromycin 500 mg twice daily prophylactically. He was being followed by hematology for the vasculopathy, and they were planning to monitor for any disease changes with computed tomography of the chest, abdomen, and pelvis every 3 months, as well as echocardiogram every 6 months for any development of pericardial or pleural fibrosis. Approximately 1 month later, the patient was admitted to the hospital again but died after 1 week from gastrointestinal complications (approximately 22 months after the onset of the rash).

CT113004033_e_Fig3_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20A%2C%20A%20biopsy%20of%20a%20lesion%20on%20the%20abdomen%20showed%20central%20epidermal%20necrosis%20and%20parakeratosis%20with%20an%20underlying%20wedgeshaped%20dermal%20infarct%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%20B%2C%20The%20infarct%20was%20surrounded%20by%20small%20occluded%20dermal%20vessels%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

Degos disease (atrophic papulosis) is a rare small vessel vasculopathy of unknown etiology, but complement-mediated endothelial injury plays a role.1,2 It typically occurs in the fourth decade of life, with a slight female predominance.3,4 The skin lesions are characteristic and described as 5- to 10-mm papules with atrophic white centers and erythematous telangiectatic rims, most commonly on the upper body and typically sparing the head, palms, and soles.1 Penile ulceration is an uncommon cutaneous feature, with only a few cases reported in the literature.5,6 Approximately one-third of patients will have only skin lesions, but two-thirds will develop systemic involvement 1 to 2 years after onset, with the gastrointestinal tract and central nervous system most commonly involved. For those with systemic involvement, the 5-year survival rate is approximately 55%, and the most common causes of death are bowel perforation, peritonitis, and stroke.3,4 Because some patients appear to never develop systemic complications, Theodoridis et al4 proposed that the disease be classified as either malignant atrophic papulosis or benign atrophic papulosis to indicate the malignant systemic form and the benign cutaneous form, respectively.

The histopathology of Degos disease changes as the lesions evolve.7 Early lesions show a superficial and deep perivascular and periadnexal lymphocytic infiltrate, possible interface dermatitis, and dermal mucin resembling lupus. The more fully developed lesions show a greater degree of inflammation and interface change as well as lymphocytic vasculitis. This stage also may have epidermal atrophy and early papillary dermal sclerosis resembling lichen sclerosus. The late-stage lesions, clinically observed as papules with atrophic white centers and surrounding erythema, show the classic pathology of wedge-shaped dermal sclerosis and central epidermal atrophy with surrounding hyperkeratosis. Interface dermatitis and dermal mucin can be seen in all stages, though mucin is diminished in the later stage.

Effective treatment options are limited; however, antithrombotics or compounds that facilitate blood perfusion, such as aspirin or pentoxifylline, initially can be used.1 Eculizumab, a humanized monoclonal antibody that prevents the cleavage of C5, has been used for salvage therapy,8 as in our case. Treprostinil, a prostacyclin analog that causes arterial vasodilation and inhibition of platelet aggregation, has been reported to improve bowel and cutaneous lesions, functional status, and neurologic symptoms.9

Our case highlights important features of Degos disease. First, it is important for both the clinician and the pathologist to recognize that the histopathology of Degos disease changes as the lesions evolve. In our case, although the lesions were characteristic of Degos disease clinically, the initial biopsy was suspicious for connective tissue disease, which led to an autoimmune evaluation that ultimately was unremarkable. Recognizing that early lesions of Degos disease can resemble connective tissue disease histologically could have prevented this delay in diagnosis. However, Degos disease has been reported in association with autoimmune diseases.10 Second, although penile ulceration is uncommon, it can be a prominent cutaneous manifestation of the disease. Finally, eculizumab and treprostinil are therapeutic options that have shown some efficacy in improving symptoms and cutaneous lesions.8,9

References
  1. Theodoridis A, Makrantonaki E, Zouboulis CC. Malignant atrophic papulosis (Köhlmeier-Degos disease)—a review. Orphanet J Rare Dis. 2013;8:10. doi:10.1186/1750-1172-8-10
  2. Magro CM, Poe JC, Kim C, et al. Degos disease: a C5b-9/interferon-α-mediated endotheliopathy syndrome. Am J Clin Pathol. 2011;135:599-610. doi:10.1309/AJCP66QIMFARLZKI
  3. Hu P, Mao Z, Liu C, et al. Malignant atrophic papulosis with motor aphasia and intestinal perforation: a case report and review of published works. J Dermatol. 2018;45:723-726. doi:10.1111/1346-8138.14280
  4. Theodoridis A, Konstantinidou A, Makrantonaki E, et al. Malignant and benign forms of atrophic papulosis (Köhlmeier-Degos disease): systemic involvement determines the prognosis. Br J Dermatol. 2014;170:110-115. doi:10.1111/bjd.12642
  5. Thomson KF, Highet AS. Penile ulceration in fatal malignant atrophic papulosis (Degos’ disease). Br J Dermatol. 2000;143:1320-1322. doi:10.1046/j.1365-2133.2000.03911.x
  6. Aydogan K, Alkan G, Karadogan Koran S, et al. Painful penile ulceration in a patient with malignant atrophic papulosis. J Eur Acad Dermatol Venereol. 2005;19:612-616. doi:10.1111/j.1468-3083.2005.01227.x
  7. Harvell JD, Williford PL, White WL. Benign cutaneous Degos’ disease: a case report with emphasis on histopathology as papules chronologically evolve. Am J Dermatopathol. 2001;23:116-123. doi:10.1097/00000372-200104000-00006
  8. Oliver B, Boehm M, Rosing DR, et al. Diffuse atrophic papules and plaques, intermittent abdominal pain, paresthesias, and cardiac abnormalities in a 55-year-old woman. J Am Acad Dermatol. 2016;75:1274-1277. doi:10.1016/j.jaad.2016.09.015
  9. Shapiro LS, Toledo-Garcia AE, Farrell JF. Effective treatment of malignant atrophic papulosis (Köhlmeier-Degos disease) with treprostinil—early experience. Orphanet J Rare Dis. 2013;8:52. doi:10.1186/1750-1172-8-52
  10. Burgin S, Stone JH, Shenoy-Bhangle AS, et al. Case records of the Massachusetts General Hospital. Case 18-2014. A 32-year-old man with a rash, myalgia, and weakness. N Engl J Med. 2014;370:2327-2337. doi:10.1056/NEJMcpc1304161
Article PDF
Author and Disclosure Information

From Baylor Scott & White Health Central Texas Foundation, Temple.

The authors report no conflict of interest.

Correspondence: Andrew Neal Minzenmayer, MD, Department of Dermatology, Baylor Scott & White Health Central Texas Foundation, 2401 S 31st St, Temple, TX 76508 (Andrew.minzenmayer@bswhealth.org).

Issue
Cutis - 113(4)
Publications
Topics
Page Number
E33-E35
Sections
Author and Disclosure Information

From Baylor Scott & White Health Central Texas Foundation, Temple.

The authors report no conflict of interest.

Correspondence: Andrew Neal Minzenmayer, MD, Department of Dermatology, Baylor Scott & White Health Central Texas Foundation, 2401 S 31st St, Temple, TX 76508 (Andrew.minzenmayer@bswhealth.org).

Author and Disclosure Information

From Baylor Scott & White Health Central Texas Foundation, Temple.

The authors report no conflict of interest.

Correspondence: Andrew Neal Minzenmayer, MD, Department of Dermatology, Baylor Scott & White Health Central Texas Foundation, 2401 S 31st St, Temple, TX 76508 (Andrew.minzenmayer@bswhealth.org).

Article PDF
Article PDF

To the Editor:

A 56-year-old man was referred to our Grand Rounds by another dermatologist in our health system for evaluation of a red scaly rash on the trunk that had been present for more than a year. More recently, over the course of approximately 9 months he experienced recurrent painful penile ulcers that lasted for approximately 4 weeks and then self-resolved. He had a medical history of central retinal vein occlusion, primary hyperparathyroidism, and nonspecific colitis. A family history was notable for lung cancer in the patient’s father and myelodysplastic syndrome and breast cancer in his mother; however, there was no family history of a similar rash. A bacterial culture of the penile ulcer was negative. Testing for antibodies against HIV and herpes simplex virus (HSV) types 1 and 2 was negative. Results of a serum VDRL test were nonreactive, which ruled out syphilis. The patient was treated by the referring dermatologist with azithromycin for possible chancroid without relief.

The patient was being followed by the referring dermatologist who initially was concerned for Degos disease based on clinical examination findings, prompting biopsy of a lesion on the back, which revealed vacuolar interface dermatitis, a sparse superficial perivascular lymphocytic infiltrate, and increased mucin—all highly suspicious for connective tissue disease (Figure 1). An antinuclear antibody test was positive, with a titer of 1:640. The patient was started on prednisone and referred to rheumatology; however, further evaluation by rheumatology for an autoimmune process—including anticardiolipin antibodies—was unremarkable. A few months prior to the current presentation, he also had mildly elevated liver function test results. A colonoscopy was performed, and a biopsy revealed nonspecific colitis. A biopsy of the penile ulcer also was nonspecific, showing only ulceration and acute and chronic inflammation. No epidermal interface change was seen. Results from a Grocott-Gomori methenamine-silver stain, Treponema pallidum immunostain, and HSV polymerase chain reaction were negative for fungal organisms, spirochetes, and HSV, respectively. The differential diagnosis included trauma, aphthous ulceration, and Behçet disease. Behçet disease was suspected by the referring dermatologist, and the patient was treated with colchicine, prednisone, pimecrolimus cream, and topical lidocaine; however, the lesions persisted, and he was subsequently referred to our Grand Rounds for further evaluation.

CT113004033_e_Fig1_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%2C%20Initial%20biopsy%20of%20the%20back%20showed%20vacuolar%20interface%20dermatitis%20and%20a%20sparse%20superficial%20perivascular%20lymphocytic%20infiltrate%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%20B%2C%20Alcian%20blue%20stain%20highlighted%20increased%20dermal%20mucin%20(original%20magnification%20%C3%97100).%20Photographs%20courtesy%20of%20Jennifer%20Leininger%2C%20MD%20(Cedar%20Park%2C%20Texas).%3C%2Fp%3E

At the current presentation, physical examination revealed several small papules with white atrophic centers and erythematous rims on the trunk and extremities (Figure 2A). An ulceration was noted on the penile shaft (Figure 2B). Further evaluation for Behçet disease, including testing for pathergy and HLA-B51, was negative. Degos disease was strongly suspected clinically, and a repeat biopsy was performed of a lesion on the abdomen, which revealed central epidermal necrosis, atrophy, and parakeratosis with an underlying wedge-shaped dermal infarct surrounded by multiple small occluded dermal vessels, perivascular inflammation, and dermal edema (Figure 3). Direct immunofluorescence was performed using antibodies against IgG, IgA, IgM, fibrinogen, albumin, and C3, which was negative. These findings from direct immunofluorescence and histopathology as well as the clinical presentation were considered compatible with Degos disease. The patient was started on aspirin and pentoxifylline. Pentoxifylline 400 mg twice daily appeared to lessen some of the pain. Pain management specialists started the patient on gabapentin.

CT113004033_e_Fig2_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Degos%20disease.%20A%2C%20Papules%20with%20white%20atrophic%20centers%20and%20erythematous%20rims%20on%20the%20anterior%20trunk.%20B%2C%20An%20ulceration%20on%20the%20penile%20shaft.%3C%2Fp%3E

Approximately 4 months after the Grand Rounds evaluation, during which time he continued treatment with pentoxifylline, he was admitted to the hospital for intractable nausea and vomiting. His condition acutely declined due to bowel perforation, and he was started on eculizumab 1200 mg every 14 days. Because of an increased risk for meningococcal meningitis while on this medication, he also was given erythromycin 500 mg twice daily prophylactically. He was being followed by hematology for the vasculopathy, and they were planning to monitor for any disease changes with computed tomography of the chest, abdomen, and pelvis every 3 months, as well as echocardiogram every 6 months for any development of pericardial or pleural fibrosis. Approximately 1 month later, the patient was admitted to the hospital again but died after 1 week from gastrointestinal complications (approximately 22 months after the onset of the rash).

CT113004033_e_Fig3_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20A%2C%20A%20biopsy%20of%20a%20lesion%20on%20the%20abdomen%20showed%20central%20epidermal%20necrosis%20and%20parakeratosis%20with%20an%20underlying%20wedgeshaped%20dermal%20infarct%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%20B%2C%20The%20infarct%20was%20surrounded%20by%20small%20occluded%20dermal%20vessels%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

Degos disease (atrophic papulosis) is a rare small vessel vasculopathy of unknown etiology, but complement-mediated endothelial injury plays a role.1,2 It typically occurs in the fourth decade of life, with a slight female predominance.3,4 The skin lesions are characteristic and described as 5- to 10-mm papules with atrophic white centers and erythematous telangiectatic rims, most commonly on the upper body and typically sparing the head, palms, and soles.1 Penile ulceration is an uncommon cutaneous feature, with only a few cases reported in the literature.5,6 Approximately one-third of patients will have only skin lesions, but two-thirds will develop systemic involvement 1 to 2 years after onset, with the gastrointestinal tract and central nervous system most commonly involved. For those with systemic involvement, the 5-year survival rate is approximately 55%, and the most common causes of death are bowel perforation, peritonitis, and stroke.3,4 Because some patients appear to never develop systemic complications, Theodoridis et al4 proposed that the disease be classified as either malignant atrophic papulosis or benign atrophic papulosis to indicate the malignant systemic form and the benign cutaneous form, respectively.

The histopathology of Degos disease changes as the lesions evolve.7 Early lesions show a superficial and deep perivascular and periadnexal lymphocytic infiltrate, possible interface dermatitis, and dermal mucin resembling lupus. The more fully developed lesions show a greater degree of inflammation and interface change as well as lymphocytic vasculitis. This stage also may have epidermal atrophy and early papillary dermal sclerosis resembling lichen sclerosus. The late-stage lesions, clinically observed as papules with atrophic white centers and surrounding erythema, show the classic pathology of wedge-shaped dermal sclerosis and central epidermal atrophy with surrounding hyperkeratosis. Interface dermatitis and dermal mucin can be seen in all stages, though mucin is diminished in the later stage.

Effective treatment options are limited; however, antithrombotics or compounds that facilitate blood perfusion, such as aspirin or pentoxifylline, initially can be used.1 Eculizumab, a humanized monoclonal antibody that prevents the cleavage of C5, has been used for salvage therapy,8 as in our case. Treprostinil, a prostacyclin analog that causes arterial vasodilation and inhibition of platelet aggregation, has been reported to improve bowel and cutaneous lesions, functional status, and neurologic symptoms.9

Our case highlights important features of Degos disease. First, it is important for both the clinician and the pathologist to recognize that the histopathology of Degos disease changes as the lesions evolve. In our case, although the lesions were characteristic of Degos disease clinically, the initial biopsy was suspicious for connective tissue disease, which led to an autoimmune evaluation that ultimately was unremarkable. Recognizing that early lesions of Degos disease can resemble connective tissue disease histologically could have prevented this delay in diagnosis. However, Degos disease has been reported in association with autoimmune diseases.10 Second, although penile ulceration is uncommon, it can be a prominent cutaneous manifestation of the disease. Finally, eculizumab and treprostinil are therapeutic options that have shown some efficacy in improving symptoms and cutaneous lesions.8,9

To the Editor:

A 56-year-old man was referred to our Grand Rounds by another dermatologist in our health system for evaluation of a red scaly rash on the trunk that had been present for more than a year. More recently, over the course of approximately 9 months he experienced recurrent painful penile ulcers that lasted for approximately 4 weeks and then self-resolved. He had a medical history of central retinal vein occlusion, primary hyperparathyroidism, and nonspecific colitis. A family history was notable for lung cancer in the patient’s father and myelodysplastic syndrome and breast cancer in his mother; however, there was no family history of a similar rash. A bacterial culture of the penile ulcer was negative. Testing for antibodies against HIV and herpes simplex virus (HSV) types 1 and 2 was negative. Results of a serum VDRL test were nonreactive, which ruled out syphilis. The patient was treated by the referring dermatologist with azithromycin for possible chancroid without relief.

The patient was being followed by the referring dermatologist who initially was concerned for Degos disease based on clinical examination findings, prompting biopsy of a lesion on the back, which revealed vacuolar interface dermatitis, a sparse superficial perivascular lymphocytic infiltrate, and increased mucin—all highly suspicious for connective tissue disease (Figure 1). An antinuclear antibody test was positive, with a titer of 1:640. The patient was started on prednisone and referred to rheumatology; however, further evaluation by rheumatology for an autoimmune process—including anticardiolipin antibodies—was unremarkable. A few months prior to the current presentation, he also had mildly elevated liver function test results. A colonoscopy was performed, and a biopsy revealed nonspecific colitis. A biopsy of the penile ulcer also was nonspecific, showing only ulceration and acute and chronic inflammation. No epidermal interface change was seen. Results from a Grocott-Gomori methenamine-silver stain, Treponema pallidum immunostain, and HSV polymerase chain reaction were negative for fungal organisms, spirochetes, and HSV, respectively. The differential diagnosis included trauma, aphthous ulceration, and Behçet disease. Behçet disease was suspected by the referring dermatologist, and the patient was treated with colchicine, prednisone, pimecrolimus cream, and topical lidocaine; however, the lesions persisted, and he was subsequently referred to our Grand Rounds for further evaluation.

CT113004033_e_Fig1_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%2C%20Initial%20biopsy%20of%20the%20back%20showed%20vacuolar%20interface%20dermatitis%20and%20a%20sparse%20superficial%20perivascular%20lymphocytic%20infiltrate%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%20B%2C%20Alcian%20blue%20stain%20highlighted%20increased%20dermal%20mucin%20(original%20magnification%20%C3%97100).%20Photographs%20courtesy%20of%20Jennifer%20Leininger%2C%20MD%20(Cedar%20Park%2C%20Texas).%3C%2Fp%3E

At the current presentation, physical examination revealed several small papules with white atrophic centers and erythematous rims on the trunk and extremities (Figure 2A). An ulceration was noted on the penile shaft (Figure 2B). Further evaluation for Behçet disease, including testing for pathergy and HLA-B51, was negative. Degos disease was strongly suspected clinically, and a repeat biopsy was performed of a lesion on the abdomen, which revealed central epidermal necrosis, atrophy, and parakeratosis with an underlying wedge-shaped dermal infarct surrounded by multiple small occluded dermal vessels, perivascular inflammation, and dermal edema (Figure 3). Direct immunofluorescence was performed using antibodies against IgG, IgA, IgM, fibrinogen, albumin, and C3, which was negative. These findings from direct immunofluorescence and histopathology as well as the clinical presentation were considered compatible with Degos disease. The patient was started on aspirin and pentoxifylline. Pentoxifylline 400 mg twice daily appeared to lessen some of the pain. Pain management specialists started the patient on gabapentin.

CT113004033_e_Fig2_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Degos%20disease.%20A%2C%20Papules%20with%20white%20atrophic%20centers%20and%20erythematous%20rims%20on%20the%20anterior%20trunk.%20B%2C%20An%20ulceration%20on%20the%20penile%20shaft.%3C%2Fp%3E

Approximately 4 months after the Grand Rounds evaluation, during which time he continued treatment with pentoxifylline, he was admitted to the hospital for intractable nausea and vomiting. His condition acutely declined due to bowel perforation, and he was started on eculizumab 1200 mg every 14 days. Because of an increased risk for meningococcal meningitis while on this medication, he also was given erythromycin 500 mg twice daily prophylactically. He was being followed by hematology for the vasculopathy, and they were planning to monitor for any disease changes with computed tomography of the chest, abdomen, and pelvis every 3 months, as well as echocardiogram every 6 months for any development of pericardial or pleural fibrosis. Approximately 1 month later, the patient was admitted to the hospital again but died after 1 week from gastrointestinal complications (approximately 22 months after the onset of the rash).

CT113004033_e_Fig3_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20A%2C%20A%20biopsy%20of%20a%20lesion%20on%20the%20abdomen%20showed%20central%20epidermal%20necrosis%20and%20parakeratosis%20with%20an%20underlying%20wedgeshaped%20dermal%20infarct%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%20B%2C%20The%20infarct%20was%20surrounded%20by%20small%20occluded%20dermal%20vessels%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100).%3C%2Fp%3E

Degos disease (atrophic papulosis) is a rare small vessel vasculopathy of unknown etiology, but complement-mediated endothelial injury plays a role.1,2 It typically occurs in the fourth decade of life, with a slight female predominance.3,4 The skin lesions are characteristic and described as 5- to 10-mm papules with atrophic white centers and erythematous telangiectatic rims, most commonly on the upper body and typically sparing the head, palms, and soles.1 Penile ulceration is an uncommon cutaneous feature, with only a few cases reported in the literature.5,6 Approximately one-third of patients will have only skin lesions, but two-thirds will develop systemic involvement 1 to 2 years after onset, with the gastrointestinal tract and central nervous system most commonly involved. For those with systemic involvement, the 5-year survival rate is approximately 55%, and the most common causes of death are bowel perforation, peritonitis, and stroke.3,4 Because some patients appear to never develop systemic complications, Theodoridis et al4 proposed that the disease be classified as either malignant atrophic papulosis or benign atrophic papulosis to indicate the malignant systemic form and the benign cutaneous form, respectively.

The histopathology of Degos disease changes as the lesions evolve.7 Early lesions show a superficial and deep perivascular and periadnexal lymphocytic infiltrate, possible interface dermatitis, and dermal mucin resembling lupus. The more fully developed lesions show a greater degree of inflammation and interface change as well as lymphocytic vasculitis. This stage also may have epidermal atrophy and early papillary dermal sclerosis resembling lichen sclerosus. The late-stage lesions, clinically observed as papules with atrophic white centers and surrounding erythema, show the classic pathology of wedge-shaped dermal sclerosis and central epidermal atrophy with surrounding hyperkeratosis. Interface dermatitis and dermal mucin can be seen in all stages, though mucin is diminished in the later stage.

Effective treatment options are limited; however, antithrombotics or compounds that facilitate blood perfusion, such as aspirin or pentoxifylline, initially can be used.1 Eculizumab, a humanized monoclonal antibody that prevents the cleavage of C5, has been used for salvage therapy,8 as in our case. Treprostinil, a prostacyclin analog that causes arterial vasodilation and inhibition of platelet aggregation, has been reported to improve bowel and cutaneous lesions, functional status, and neurologic symptoms.9

Our case highlights important features of Degos disease. First, it is important for both the clinician and the pathologist to recognize that the histopathology of Degos disease changes as the lesions evolve. In our case, although the lesions were characteristic of Degos disease clinically, the initial biopsy was suspicious for connective tissue disease, which led to an autoimmune evaluation that ultimately was unremarkable. Recognizing that early lesions of Degos disease can resemble connective tissue disease histologically could have prevented this delay in diagnosis. However, Degos disease has been reported in association with autoimmune diseases.10 Second, although penile ulceration is uncommon, it can be a prominent cutaneous manifestation of the disease. Finally, eculizumab and treprostinil are therapeutic options that have shown some efficacy in improving symptoms and cutaneous lesions.8,9

References
  1. Theodoridis A, Makrantonaki E, Zouboulis CC. Malignant atrophic papulosis (Köhlmeier-Degos disease)—a review. Orphanet J Rare Dis. 2013;8:10. doi:10.1186/1750-1172-8-10
  2. Magro CM, Poe JC, Kim C, et al. Degos disease: a C5b-9/interferon-α-mediated endotheliopathy syndrome. Am J Clin Pathol. 2011;135:599-610. doi:10.1309/AJCP66QIMFARLZKI
  3. Hu P, Mao Z, Liu C, et al. Malignant atrophic papulosis with motor aphasia and intestinal perforation: a case report and review of published works. J Dermatol. 2018;45:723-726. doi:10.1111/1346-8138.14280
  4. Theodoridis A, Konstantinidou A, Makrantonaki E, et al. Malignant and benign forms of atrophic papulosis (Köhlmeier-Degos disease): systemic involvement determines the prognosis. Br J Dermatol. 2014;170:110-115. doi:10.1111/bjd.12642
  5. Thomson KF, Highet AS. Penile ulceration in fatal malignant atrophic papulosis (Degos’ disease). Br J Dermatol. 2000;143:1320-1322. doi:10.1046/j.1365-2133.2000.03911.x
  6. Aydogan K, Alkan G, Karadogan Koran S, et al. Painful penile ulceration in a patient with malignant atrophic papulosis. J Eur Acad Dermatol Venereol. 2005;19:612-616. doi:10.1111/j.1468-3083.2005.01227.x
  7. Harvell JD, Williford PL, White WL. Benign cutaneous Degos’ disease: a case report with emphasis on histopathology as papules chronologically evolve. Am J Dermatopathol. 2001;23:116-123. doi:10.1097/00000372-200104000-00006
  8. Oliver B, Boehm M, Rosing DR, et al. Diffuse atrophic papules and plaques, intermittent abdominal pain, paresthesias, and cardiac abnormalities in a 55-year-old woman. J Am Acad Dermatol. 2016;75:1274-1277. doi:10.1016/j.jaad.2016.09.015
  9. Shapiro LS, Toledo-Garcia AE, Farrell JF. Effective treatment of malignant atrophic papulosis (Köhlmeier-Degos disease) with treprostinil—early experience. Orphanet J Rare Dis. 2013;8:52. doi:10.1186/1750-1172-8-52
  10. Burgin S, Stone JH, Shenoy-Bhangle AS, et al. Case records of the Massachusetts General Hospital. Case 18-2014. A 32-year-old man with a rash, myalgia, and weakness. N Engl J Med. 2014;370:2327-2337. doi:10.1056/NEJMcpc1304161
References
  1. Theodoridis A, Makrantonaki E, Zouboulis CC. Malignant atrophic papulosis (Köhlmeier-Degos disease)—a review. Orphanet J Rare Dis. 2013;8:10. doi:10.1186/1750-1172-8-10
  2. Magro CM, Poe JC, Kim C, et al. Degos disease: a C5b-9/interferon-α-mediated endotheliopathy syndrome. Am J Clin Pathol. 2011;135:599-610. doi:10.1309/AJCP66QIMFARLZKI
  3. Hu P, Mao Z, Liu C, et al. Malignant atrophic papulosis with motor aphasia and intestinal perforation: a case report and review of published works. J Dermatol. 2018;45:723-726. doi:10.1111/1346-8138.14280
  4. Theodoridis A, Konstantinidou A, Makrantonaki E, et al. Malignant and benign forms of atrophic papulosis (Köhlmeier-Degos disease): systemic involvement determines the prognosis. Br J Dermatol. 2014;170:110-115. doi:10.1111/bjd.12642
  5. Thomson KF, Highet AS. Penile ulceration in fatal malignant atrophic papulosis (Degos’ disease). Br J Dermatol. 2000;143:1320-1322. doi:10.1046/j.1365-2133.2000.03911.x
  6. Aydogan K, Alkan G, Karadogan Koran S, et al. Painful penile ulceration in a patient with malignant atrophic papulosis. J Eur Acad Dermatol Venereol. 2005;19:612-616. doi:10.1111/j.1468-3083.2005.01227.x
  7. Harvell JD, Williford PL, White WL. Benign cutaneous Degos’ disease: a case report with emphasis on histopathology as papules chronologically evolve. Am J Dermatopathol. 2001;23:116-123. doi:10.1097/00000372-200104000-00006
  8. Oliver B, Boehm M, Rosing DR, et al. Diffuse atrophic papules and plaques, intermittent abdominal pain, paresthesias, and cardiac abnormalities in a 55-year-old woman. J Am Acad Dermatol. 2016;75:1274-1277. doi:10.1016/j.jaad.2016.09.015
  9. Shapiro LS, Toledo-Garcia AE, Farrell JF. Effective treatment of malignant atrophic papulosis (Köhlmeier-Degos disease) with treprostinil—early experience. Orphanet J Rare Dis. 2013;8:52. doi:10.1186/1750-1172-8-52
  10. Burgin S, Stone JH, Shenoy-Bhangle AS, et al. Case records of the Massachusetts General Hospital. Case 18-2014. A 32-year-old man with a rash, myalgia, and weakness. N Engl J Med. 2014;370:2327-2337. doi:10.1056/NEJMcpc1304161
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
E33-E35
Page Number
E33-E35
Publications
Publications
Topics
Article Type
Display Headline
Clinical Manifestation of Degos Disease: Painful Penile Ulcers
Display Headline
Clinical Manifestation of Degos Disease: Painful Penile Ulcers
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Mizenmayer</fileName> <TBEID>0C02F63F.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F63F</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Mizenmayer</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240503T111205</firstPublished> <LastPublished>20240503T111205</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240503T111204</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Andrew Neal Minzenmayer, MD; Katherine Fiala, MD</byline> <bylineText>Andrew Neal Minzenmayer, MD; Katherine Fiala, MD</bylineText> <bylineFull>Andrew Neal Minzenmayer, MD; Katherine Fiala, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E33-E35</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>To the Editor:A 56-year-old man was referred to our Grand Rounds by another dermatologist in our health system for evaluation of a red scaly rash on the trunk t</metaDescription> <articlePDF>301309</articlePDF> <teaserImage/> <title>Clinical Manifestation of Degos Disease: Painful Penile Ulcers</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>April</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>4</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2163</CMSID> </CMSIDs> <keywords> <keyword>dermatopathology</keyword> <keyword> penile ulcers</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>April 2024</pubIssueName> <pubArticleType>Online Exclusive | 2163</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">44</term> </sections> <topics> <term canonical="true">204</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002725.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Clinical Manifestation of Degos Disease: Painful Penile Ulcers</title> <deck/> </itemMeta> <itemContent> <p>To the Editor:<br/><br/>A 56-year-old man was referred to our Grand Rounds by another dermatologist in our health system for evaluation of a red scaly rash on the trunk that had been present for more than a year. More recently, over the course of approximately 9 months he experienced recurrent painful penile ulcers that lasted for approximately 4 weeks and then self-resolved. He had a medical history of central retinal vein occlusion, primary hyperparathyroidism, and nonspecific colitis. A family history was notable for lung cancer in the patient’s father and myelodysplastic syndrome and breast cancer in his mother; however, there was no family history of a similar rash. A bacterial culture of the penile ulcer was negative. Testing for antibodies against HIV and herpes simplex virus (HSV) types 1 and 2 was negative. Results of a serum VDRL test were nonreactive, which ruled out syphilis. The patient was treated by the referring dermatologist with azithromycin for possible chancroid without relief.</p> <p>The patient was being followed by the referring dermatologist who initially was concerned for Degos disease based on clinical examination findings, prompting biopsy of a lesion on the back, which revealed vacuolar interface dermatitis, a sparse superficial perivascular lymphocytic infiltrate, and increased mucin—all highly suspicious for connective tissue disease (Figure 1). An antinuclear antibody test was positive, with a titer of 1:640. The patient was started on prednisone and referred to rheumatology; however, further evaluation by rheumatology for an autoimmune process—including anticardiolipin antibodies—was unremarkable. A few months prior to the current presentation, he also had mildly elevated liver function test results. A colonoscopy was performed, and a biopsy revealed nonspecific colitis. A biopsy of the penile ulcer also was nonspecific, showing only ulceration and acute and chronic inflammation. No epidermal interface change was seen. Results from a Grocott-Gomori methenamine-silver stain, <i>Treponema pallidum</i> immunostain, and HSV polymerase chain reaction were negative for fungal organisms, spirochetes, and HSV, respectively. The differential diagnosis included trauma, aphthous ulceration, and Behçet disease. Behçet disease was suspected by the referring dermatologist, and the patient was treated with colchicine, prednisone, pimecrolimus cream, and topical lidocaine; however, the lesions persisted, and he was subsequently referred to our Grand Rounds for further evaluation.<br/><br/>At the current presentation, physical examination revealed several small papules with white atrophic centers and erythematous rims on the trunk and extremities (Figure 2A). An ulceration was noted on the penile shaft (Figure 2B). Further evaluation for Behçet disease, including testing for pathergy and HLA-B51, was negative. Degos disease was strongly suspected clinically, and a repeat biopsy was performed of a lesion on the abdomen, which revealed central epidermal necrosis, atrophy, and parakeratosis with an underlying wedge-shaped dermal infarct surrounded by multiple small occluded dermal vessels, perivascular inflammation, and dermal edema (Figure 3). Direct immunofluorescence was performed using antibodies against IgG, IgA, IgM, fibrinogen, albumin, and C3, which was negative. These findings from direct immunofluorescence and histopathology as well as the clinical presentation were considered compatible with Degos disease. The patient was started on aspirin and pentoxifylline. Pentoxifylline 400 mg twice daily appeared to lessen some of the pain. Pain management specialists started the patient on gabapentin. <br/><br/>Approximately 4 months after the Grand Rounds evaluation, during which time he continued treatment with pentoxifylline, he was admitted to the hospital for intractable nausea and vomiting. His condition acutely declined due to bowel perforation, and he was started on eculizumab 1200 mg every 14 days. Because of an increased risk for meningococcal meningitis while on this medication, he also was given erythromycin 500 mg twice daily prophylactically. He was being followed by hematology for the vasculopathy, and they were planning to monitor for any disease changes with computed tomography of the chest, abdomen, and pelvis every 3 months, as well as echocardiogram every 6 months for any development of pericardial or pleural fibrosis. Approximately 1 month later, the patient was admitted to the hospital again but died after 1 week from gastrointestinal complications (approximately 22 months after the onset of the rash).<br/><br/>Degos disease (atrophic papulosis) is a rare small vessel vasculopathy of unknown etiology, but complement-mediated endothelial injury plays a role.<sup>1,2</sup> It typically occurs in the fourth decade of life, with a slight female predominance.<sup>3,4</sup> The skin lesions are characteristic and described as 5- to 10-mm papules with atrophic white centers and erythematous telangiectatic rims, most commonly on the upper body and typically sparing the head, palms, and soles.<sup>1</sup> Penile ulceration is an uncommon cutaneous feature, with only a few cases reported in the literature.<sup>5,6</sup> Approximately one-third of patients will have only skin lesions, but two-thirds will develop systemic involvement 1 to 2 years after onset, with the gastrointestinal tract and central nervous system most commonly involved. For those with systemic involvement, the 5-year survival rate is approximately 55%, and the most common causes of death are bowel perforation, peritonitis, and stroke.<sup>3,4</sup> Because some patients appear to never develop systemic complications, Theodoridis et al<sup>4</sup> proposed that the disease be classified as either malignant atrophic papulosis or benign atrophic papulosis to indicate the malignant systemic form and the benign cutaneous form, respectively. <br/><br/>The histopathology of Degos disease changes as the lesions evolve.<sup>7</sup> Early lesions show a superficial and deep perivascular and periadnexal lymphocytic infiltrate, possible interface dermatitis, and dermal mucin resembling lupus. The more fully developed lesions show a greater degree of inflammation and interface change as well as lymphocytic vasculitis. This stage also may have epidermal atrophy and early papillary dermal sclerosis resembling lichen sclerosus. The late-stage lesions, clinically observed as papules with atrophic white centers and surrounding erythema, show the classic pathology of wedge-shaped dermal sclerosis and central epidermal atrophy with surrounding hyperkeratosis. Interface dermatitis and dermal mucin can be seen in all stages, though mucin is diminished in the later stage. <br/><br/>Effective treatment options are limited; however, antithrombotics or compounds that facilitate blood perfusion, such as aspirin or pentoxifylline, initially can be used.<sup>1</sup> Eculizumab, a humanized monoclonal antibody that prevents the cleavage of C5, has been used for salvage therapy,<sup>8</sup> as in our case. Treprostinil, a prostacyclin analog that causes arterial vasodilation and inhibition of platelet aggregation, has been reported to improve bowel and cutaneous lesions, functional status, and neurologic symptoms.<sup>9</sup> <br/><br/>Our case highlights important features of Degos disease. First, it is important for both the clinician and the pathologist to recognize that the histopathology of Degos disease changes as the lesions evolve. In our case, although the lesions were characteristic of Degos disease clinically, the initial biopsy was suspicious for connective tissue disease, which led to an autoimmune evaluation that ultimately was unremarkable. Recognizing that early lesions of Degos disease can resemble connective tissue disease histologically could have prevented this delay in diagnosis. However, Degos disease has been reported in association with autoimmune diseases.<sup>10</sup> Second, although penile ulceration is uncommon, it can be a prominent cutaneous manifestation of the disease. Finally, eculizumab and treprostinil are therapeutic options that have shown some efficacy in improving symptoms and cutaneous lesions.<sup>8,9</sup></p> <h2>References</h2> <p class="reference"> 1. Theodoridis A, Makrantonaki E, Zouboulis CC. Malignant atrophic papulosis (Köhlmeier-Degos disease)—a review. <i>Orphanet J Rare Dis</i>. 2013;8:10. doi:10.1186/1750-1172-8-10<br/><br/> 2. Magro CM, Poe JC, Kim C, et al. Degos disease: a C5b-9/interferon-α-mediated endotheliopathy syndrome. <i>Am J Clin Pathol</i>. 2011;135:599-610. doi:10.1309/AJCP66QIMFARLZKI<br/><br/> 3. Hu P, Mao Z, Liu C, et al. Malignant atrophic papulosis with motor aphasia and intestinal perforation: a case report and review of published works. <i>J Dermatol</i>. 2018;45:723-726. doi:10.1111/1346-8138.14280<br/><br/> 4. Theodoridis A, Konstantinidou A, Makrantonaki E, et al. Malignant and benign forms of atrophic papulosis (Köhlmeier-Degos disease): systemic involvement determines the prognosis. <i>Br J Dermatol</i>. 2014;170:110-115. doi:10.1111/bjd.12642</p> <p class="reference"> 5. Thomson KF, Highet AS. Penile ulceration in fatal malignant atrophic papulosis (Degos’ disease). <i>Br J Dermatol</i>. 2000;143:1320-1322. doi:10.1046/j.1365-2133.2000.03911.x<br/><br/> 6. Aydogan K, Alkan G, Karadogan Koran S, et al. Painful penile ulceration in a patient with malignant atrophic papulosis. <i>J Eur Acad Dermatol Venereol</i>. 2005;19:612-616. doi:10.1111/j.1468-3083.2005.01227.x<br/><br/> 7. Harvell JD, Williford PL, White WL. Benign cutaneous Degos’ disease: a case report with emphasis on histopathology as papules chronologically evolve. <i>Am J Dermatopathol</i>. 2001;23:116-123. doi:10.1097/00000372-200104000-00006<br/><br/> 8. Oliver B, Boehm M, Rosing DR, et al. Diffuse atrophic papules and plaques, intermittent abdominal pain, paresthesias, and cardiac abnormalities in a 55-year-old woman. <i>J Am Acad Dermatol</i>. 2016;75:1274-1277. doi:10.1016/j.jaad.2016.09.015<br/><br/> 9. Shapiro LS, Toledo-Garcia AE, Farrell JF. Effective treatment of malignant atrophic papulosis (Köhlmeier-Degos disease) with treprostinil—early experience. <i>Orphanet J Rare Dis</i>. 2013;8:52. doi:10.1186/1750-1172-8-52<br/><br/>10. Burgin S, Stone JH, Shenoy-Bhangle AS, et al. Case records of the Massachusetts General Hospital. Case 18-2014. A 32-year-old man with a rash, myalgia, and weakness. <i>N Engl J Med</i>. 2014;370:2327-2337. doi:10.1056/NEJMcpc1304161</p> </itemContent> </newsItem> </itemSet></root>
Inside the Article

PRACTICE POINTS

  • Papules with atrophic white centers and erythematous telangiectatic rims are the characteristic skin lesions found in Degos disease.
  • A painful penile ulceration also may occur in Degos disease, though it is uncommon.
  • The histopathology of skin lesions changes as the lesions evolve. Early lesions may resemble connective tissue disease. Late lesions show the classic pathology of wedge-shaped dermal sclerosis.
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
Image
Disable zoom
Off

Occipital Scalp Nodule in a Newborn

Article Type
Changed
Wed, 04/24/2024 - 11:18
Display Headline
Occipital Scalp Nodule in a Newborn

The Diagnosis: Subcutaneous Fat Necrosis

Histopathology revealed lobular panniculitis with lymphohistiocytic inflammation, lipid crystals, and calcifications in our patient (Figure). Subcutaneous fat necrosis (SCFN) was diagnosed based on these characteristic histopathologic findings. No further treatment was pursued.

CT113004027_figAB.jpg
%3Cp%3EA%20and%20B%2C%20Histopathology%20revealed%20lobular%20panniculitis%20with%20lymphohistiocytic%20inflammation%2C%20lipid%20crystals%2C%20and%20calcifications%20characteristic%20of%20subcutaneous%20fat%20necrosis%20(H%26amp%3BE%2C%20original%20magnifications%20%C3%97100%20and%20%C3%9720).%3C%2Fp%3E

Subcutaneous fat necrosis is a rare, self-limiting panniculitis that typically resolves within several weeks to months without scarring. It manifests as red or violaceous subcutaneous nodules or plaques most commonly on the buttocks, trunk, proximal arms and legs, and cheeks.1 Histopathology reveals lobular panniculitis with dense granulomatous infiltrates of histiocytes, eosinophils, and multinucleated giant cells with needle-shaped crystals. Focal areas of fat necrosis with calcification also can be seen.2

The epidemiology of SCFN is unknown. Most cases occur in healthy full-term to postterm neonates who experience hypoxia, other prenatal stressors, or therapeutic hypothermia for the treatment of hypoxic-ischemic encephalopathy.3 Although the etiology is unclear, certain inciting factors such as local tissue hypoxia, cold exposure, meconium aspiration, maternal diabetes, preeclampsia, and mechanical pressure have been proposed. Our patient underwent hypothermic cooling protocol, and it has been suggested that the increased saturated to unsaturated fat concentration in the skin of newborns increases the melting point, thus predisposing them to fat crystalization.4 Cases of SCFN involving the scalp are rare; therefore, any newborns receiving hypothermic therapy for hypoxic-ischemic encephalopathy should have a thorough skin examination with possible biopsy of lesions that are characteristic of SCFN, such as red or violaceous subcutaneous nodules or plaques, for specific disease identification.

The main complication of SCFN is hypercalcemia, which occurs in approximately 50% of cases. Other serum abnormalities include hyperglycemia, hypertriglyceridemia, and thrombocytopenia, though these findings are not as well associated.4 Patients with associated hypercalcemia may be asymptomatic, as in our patient, but other presentations include irritability, weakness, anorexia, vomiting, renal failure, failure to thrive, and encephalopathy. Nephrocalcinosis is a common complication of severe hypercalcemia; however, there is little evidence of associated major renal dysfunction.5 The exact mechanism of hypercalcemia is poorly understood. A widely accepted theory postulates that a granulomatous inflammatory infiltrate upregulates 1-α-hydroxylase activity, which enzymatically converts 25-hydroxyvitamin D to its active form, 1,25-dihydroxycholecalciferol, which increases bone resorption and calcium absorption through the gastrointestinal tract and renal systems. Treatments for hypercalcemia include hyperhydration, calcium-wasting diuretics, and low calcium intake.6 Furthermore, calcium levels should be obtained at the time of diagnosis and 30, 45, and 60 days after the lesions resolve.4

Subcutaneous fat necrosis needs to be differentiated from the more severe panniculitis, sclerema neonatorum (SN), which typically affects critically ill, preterm, and small-for-gestational-age newborns. It is associated with a high mortality rate and is characterized by skin and subadjacent tissue structures. The process typically begins in the thighs, buttocks, or trunk and spreads diffusely, sparing the fat-free palms, soles, and genitalia.7 Although our patient was born preterm, the physical characteristics of the nodule and the lack of severe illness placed SN lower on our differential. Histopathologic differences between SCFN and SN involve the extent of tissue fibrosis and presence of inflammatory cells. Sclerema neonatorum typically manifests with thickened connective tissue with a sparse inflammatory infiltrate, including lymphocytes, histiocytes, and multinucleated giant cells.7 Conversely, SCFN manifests with fat necrosis with an extensive inflammatory infiltrate. It is important to be able to distinguish between these 2 conditions, as both have vastly different prognoses.

Cold panniculitis, sometimes called “popsicle panniculitis,” is a phenomenon in which cold contact with the skin causes eruption of firm, erythematous, indurated plaques at the site of exposure. This self-limiting condition typically appears hours to days after cold exposure and spontaneously resolves in a few weeks.8 Therapeutic hypothermic protocol treatment involves using cooling devices to lower the body temperature for a short duration. The temperature typically is lowered to approximately 32 °C to 36 °C. These temperatures are not low enough to induce cold panniculitis, which is more commonly seen in facial ice applications when managing supraventricular tachycardia in neonates.

Cephalohematoma is a birthing injury that causes blood accumulation within the subperiosteal space. During parturition, the compressive and sheering forces on the calvarium rupture the vessels passing through the periosteum, causing blood to pool slowly into the subperiostium; thus, a cephalohematoma usually manifests later at 1 to 3 days of life as localized head swelling.9 The bleeding typically does not cross suture lines and is primarily found in the occipital or parietal regions. The incidence has been reported to be 0.4% to 2.5% of all live births.10 Although the location of the nodule in our patient was in the occipital region, imaging and biopsy results did not show hemorrhagic findings consistent with cephalohematoma. Management of cephalohematoma mainly is observational, as the mass slowly regresses and the accumulated blood gradually is reabsorbed.

Fungal scalp infections (tinea capitis) are common in the pediatric population. The peak incidence of this infection has been reported in children aged 3 to 7 years, with Trichophyton tonsurans and Microsporum canis as the usual causative organisms.11 Clinical features of tinea capitis include scaly patches with hair loss, hair loss with black pigmented dots at the follicular openings, diffuse scalp scaling with subtle hair loss, and cervical lymphadenopathy.12 Although less common, tinea capitis can progress to a more severe form known as a kerion, which is characterized by a tender plaque with pustules and crusting. A kerion can result in permanent scarring and alopecia if left untreated.12 In our patient, a nodule with scaling and faint erythema was observed, but no black pigmented dots at the follicular orifices were present. Therefore, a potassium hydroxide wet mount preparation used to diagnose tinea capitis was unnecessary. Systemic oral antifungal therapy such as fluconazole or terbinafine is the standard treatment for tinea capitis.

References
  1. Coondoo A, Lahiry R, Choudhury A, et al. Tender skin nodules in a newborn. Indian J Dermatol. 2013;58:328. doi:10.4103/0019-5154.113983
  2. Mitra S, Dove J, Somisetty SK. Subcutaneous fat necrosis in newbornan unusual case and review of literature. Eur J Pediatr. 2011;170:1107- 1110. doi:10.1007/s00431-011-1405-x
  3. Velasquez JH, Mendez MD. Newborn subcutaneous fat necrosis. In: StatPearls. StatPearls Publishing; 2022.
  4. Stefanko NS, Drolet BA. Subcutaneous fat necrosis of the newborn and associated hypercalcemia: a systematic review of the literature. Pediatr Dermatol. 2019;36:24-30. doi:10.1111/pde.13640
  5. Shumer DE, Thaker V, Taylor GA, et al. Severe hypercalcaemia due to subcutaneous fat necrosis: presentation, management and complications. Arch Dis Child Fetal Neonatal Ed. 2014;99:F419-F421. doi:10.1136/ archdischild-2014-306069
  6. Farooque A, Moss C, Zehnder D, et al. Expression of 25-hydroxyvitamin D3-1alpha-hydroxylase in subcutaneous fat necrosis. Br J Dermatol. 2009;160:423-425. doi:10.1111/j.1365-2133.2008.08844.x
  7. Zeb A, Darmstadt GL. Sclerema neonatorum: a review of nomenclature, clinical presentation, histological features, differential diagnoses and management. J Perinatol. 2008;28:453-460. doi:10.1038/jp.2008.33
  8. Quesada-Cortés A, Campos-Muñoz L, Díaz-Díaz RM, et al. Cold panniculitis. Dermatol Clin. 2008;26:485-489, vii. doi:10.1016 /j.det.2008.05.015
  9. Raines DA, Krawiec C, Jain S. Cephalohematoma. In: StatPearls. StatPearls Publishing; 2023.
  10. Chung HY, Chung JY, Lee DG, et al. Surgical treatment of ossified cephalhematoma. J Craniofac Surg. 2004;15:774-779. doi:10.1097/00001665- 200409000-00015
  11. Leung AKC, Hon KL, Leong KF, et al. Tinea capitis: an updated review. Recent Pat Inflamm Allergy Drug Discov. 2020;14:58-68. doi:10.2174/1872 213x14666200106145624
  12. Kovitwanichkanont T, Chong A. Superficial fungal infections. Aust J Gen Pract. 2019;48:706-711. doi:10.31128/ajgp-05-19-4930
Article PDF
Author and Disclosure Information

From the University of Texas Health Science Center at San Antonio. Tue F. Nguyen and Moses A. Alfaro are from the Long School of Medicine. Dr. Fletcher, Dr. Messer, and Dr. Osswald are from the Division of Dermatology. Dr. Bode-Omoleye is from the Division of Pathology and Laboratory Medicine.

The authors report no conflict of interest.

Correspondence: Sandra Osswald, MD, 7979 Wurzbach Rd, 3rd Fl, Department of Dermatology, San Antonio, TX 78229 (osswald@uthscsa.edu).

Issue
Cutis - 113(4)
Publications
Topics
Page Number
E27-E29
Sections
Author and Disclosure Information

From the University of Texas Health Science Center at San Antonio. Tue F. Nguyen and Moses A. Alfaro are from the Long School of Medicine. Dr. Fletcher, Dr. Messer, and Dr. Osswald are from the Division of Dermatology. Dr. Bode-Omoleye is from the Division of Pathology and Laboratory Medicine.

The authors report no conflict of interest.

Correspondence: Sandra Osswald, MD, 7979 Wurzbach Rd, 3rd Fl, Department of Dermatology, San Antonio, TX 78229 (osswald@uthscsa.edu).

Author and Disclosure Information

From the University of Texas Health Science Center at San Antonio. Tue F. Nguyen and Moses A. Alfaro are from the Long School of Medicine. Dr. Fletcher, Dr. Messer, and Dr. Osswald are from the Division of Dermatology. Dr. Bode-Omoleye is from the Division of Pathology and Laboratory Medicine.

The authors report no conflict of interest.

Correspondence: Sandra Osswald, MD, 7979 Wurzbach Rd, 3rd Fl, Department of Dermatology, San Antonio, TX 78229 (osswald@uthscsa.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Subcutaneous Fat Necrosis

Histopathology revealed lobular panniculitis with lymphohistiocytic inflammation, lipid crystals, and calcifications in our patient (Figure). Subcutaneous fat necrosis (SCFN) was diagnosed based on these characteristic histopathologic findings. No further treatment was pursued.

CT113004027_figAB.jpg
%3Cp%3EA%20and%20B%2C%20Histopathology%20revealed%20lobular%20panniculitis%20with%20lymphohistiocytic%20inflammation%2C%20lipid%20crystals%2C%20and%20calcifications%20characteristic%20of%20subcutaneous%20fat%20necrosis%20(H%26amp%3BE%2C%20original%20magnifications%20%C3%97100%20and%20%C3%9720).%3C%2Fp%3E

Subcutaneous fat necrosis is a rare, self-limiting panniculitis that typically resolves within several weeks to months without scarring. It manifests as red or violaceous subcutaneous nodules or plaques most commonly on the buttocks, trunk, proximal arms and legs, and cheeks.1 Histopathology reveals lobular panniculitis with dense granulomatous infiltrates of histiocytes, eosinophils, and multinucleated giant cells with needle-shaped crystals. Focal areas of fat necrosis with calcification also can be seen.2

The epidemiology of SCFN is unknown. Most cases occur in healthy full-term to postterm neonates who experience hypoxia, other prenatal stressors, or therapeutic hypothermia for the treatment of hypoxic-ischemic encephalopathy.3 Although the etiology is unclear, certain inciting factors such as local tissue hypoxia, cold exposure, meconium aspiration, maternal diabetes, preeclampsia, and mechanical pressure have been proposed. Our patient underwent hypothermic cooling protocol, and it has been suggested that the increased saturated to unsaturated fat concentration in the skin of newborns increases the melting point, thus predisposing them to fat crystalization.4 Cases of SCFN involving the scalp are rare; therefore, any newborns receiving hypothermic therapy for hypoxic-ischemic encephalopathy should have a thorough skin examination with possible biopsy of lesions that are characteristic of SCFN, such as red or violaceous subcutaneous nodules or plaques, for specific disease identification.

The main complication of SCFN is hypercalcemia, which occurs in approximately 50% of cases. Other serum abnormalities include hyperglycemia, hypertriglyceridemia, and thrombocytopenia, though these findings are not as well associated.4 Patients with associated hypercalcemia may be asymptomatic, as in our patient, but other presentations include irritability, weakness, anorexia, vomiting, renal failure, failure to thrive, and encephalopathy. Nephrocalcinosis is a common complication of severe hypercalcemia; however, there is little evidence of associated major renal dysfunction.5 The exact mechanism of hypercalcemia is poorly understood. A widely accepted theory postulates that a granulomatous inflammatory infiltrate upregulates 1-α-hydroxylase activity, which enzymatically converts 25-hydroxyvitamin D to its active form, 1,25-dihydroxycholecalciferol, which increases bone resorption and calcium absorption through the gastrointestinal tract and renal systems. Treatments for hypercalcemia include hyperhydration, calcium-wasting diuretics, and low calcium intake.6 Furthermore, calcium levels should be obtained at the time of diagnosis and 30, 45, and 60 days after the lesions resolve.4

Subcutaneous fat necrosis needs to be differentiated from the more severe panniculitis, sclerema neonatorum (SN), which typically affects critically ill, preterm, and small-for-gestational-age newborns. It is associated with a high mortality rate and is characterized by skin and subadjacent tissue structures. The process typically begins in the thighs, buttocks, or trunk and spreads diffusely, sparing the fat-free palms, soles, and genitalia.7 Although our patient was born preterm, the physical characteristics of the nodule and the lack of severe illness placed SN lower on our differential. Histopathologic differences between SCFN and SN involve the extent of tissue fibrosis and presence of inflammatory cells. Sclerema neonatorum typically manifests with thickened connective tissue with a sparse inflammatory infiltrate, including lymphocytes, histiocytes, and multinucleated giant cells.7 Conversely, SCFN manifests with fat necrosis with an extensive inflammatory infiltrate. It is important to be able to distinguish between these 2 conditions, as both have vastly different prognoses.

Cold panniculitis, sometimes called “popsicle panniculitis,” is a phenomenon in which cold contact with the skin causes eruption of firm, erythematous, indurated plaques at the site of exposure. This self-limiting condition typically appears hours to days after cold exposure and spontaneously resolves in a few weeks.8 Therapeutic hypothermic protocol treatment involves using cooling devices to lower the body temperature for a short duration. The temperature typically is lowered to approximately 32 °C to 36 °C. These temperatures are not low enough to induce cold panniculitis, which is more commonly seen in facial ice applications when managing supraventricular tachycardia in neonates.

Cephalohematoma is a birthing injury that causes blood accumulation within the subperiosteal space. During parturition, the compressive and sheering forces on the calvarium rupture the vessels passing through the periosteum, causing blood to pool slowly into the subperiostium; thus, a cephalohematoma usually manifests later at 1 to 3 days of life as localized head swelling.9 The bleeding typically does not cross suture lines and is primarily found in the occipital or parietal regions. The incidence has been reported to be 0.4% to 2.5% of all live births.10 Although the location of the nodule in our patient was in the occipital region, imaging and biopsy results did not show hemorrhagic findings consistent with cephalohematoma. Management of cephalohematoma mainly is observational, as the mass slowly regresses and the accumulated blood gradually is reabsorbed.

Fungal scalp infections (tinea capitis) are common in the pediatric population. The peak incidence of this infection has been reported in children aged 3 to 7 years, with Trichophyton tonsurans and Microsporum canis as the usual causative organisms.11 Clinical features of tinea capitis include scaly patches with hair loss, hair loss with black pigmented dots at the follicular openings, diffuse scalp scaling with subtle hair loss, and cervical lymphadenopathy.12 Although less common, tinea capitis can progress to a more severe form known as a kerion, which is characterized by a tender plaque with pustules and crusting. A kerion can result in permanent scarring and alopecia if left untreated.12 In our patient, a nodule with scaling and faint erythema was observed, but no black pigmented dots at the follicular orifices were present. Therefore, a potassium hydroxide wet mount preparation used to diagnose tinea capitis was unnecessary. Systemic oral antifungal therapy such as fluconazole or terbinafine is the standard treatment for tinea capitis.

The Diagnosis: Subcutaneous Fat Necrosis

Histopathology revealed lobular panniculitis with lymphohistiocytic inflammation, lipid crystals, and calcifications in our patient (Figure). Subcutaneous fat necrosis (SCFN) was diagnosed based on these characteristic histopathologic findings. No further treatment was pursued.

CT113004027_figAB.jpg
%3Cp%3EA%20and%20B%2C%20Histopathology%20revealed%20lobular%20panniculitis%20with%20lymphohistiocytic%20inflammation%2C%20lipid%20crystals%2C%20and%20calcifications%20characteristic%20of%20subcutaneous%20fat%20necrosis%20(H%26amp%3BE%2C%20original%20magnifications%20%C3%97100%20and%20%C3%9720).%3C%2Fp%3E

Subcutaneous fat necrosis is a rare, self-limiting panniculitis that typically resolves within several weeks to months without scarring. It manifests as red or violaceous subcutaneous nodules or plaques most commonly on the buttocks, trunk, proximal arms and legs, and cheeks.1 Histopathology reveals lobular panniculitis with dense granulomatous infiltrates of histiocytes, eosinophils, and multinucleated giant cells with needle-shaped crystals. Focal areas of fat necrosis with calcification also can be seen.2

The epidemiology of SCFN is unknown. Most cases occur in healthy full-term to postterm neonates who experience hypoxia, other prenatal stressors, or therapeutic hypothermia for the treatment of hypoxic-ischemic encephalopathy.3 Although the etiology is unclear, certain inciting factors such as local tissue hypoxia, cold exposure, meconium aspiration, maternal diabetes, preeclampsia, and mechanical pressure have been proposed. Our patient underwent hypothermic cooling protocol, and it has been suggested that the increased saturated to unsaturated fat concentration in the skin of newborns increases the melting point, thus predisposing them to fat crystalization.4 Cases of SCFN involving the scalp are rare; therefore, any newborns receiving hypothermic therapy for hypoxic-ischemic encephalopathy should have a thorough skin examination with possible biopsy of lesions that are characteristic of SCFN, such as red or violaceous subcutaneous nodules or plaques, for specific disease identification.

The main complication of SCFN is hypercalcemia, which occurs in approximately 50% of cases. Other serum abnormalities include hyperglycemia, hypertriglyceridemia, and thrombocytopenia, though these findings are not as well associated.4 Patients with associated hypercalcemia may be asymptomatic, as in our patient, but other presentations include irritability, weakness, anorexia, vomiting, renal failure, failure to thrive, and encephalopathy. Nephrocalcinosis is a common complication of severe hypercalcemia; however, there is little evidence of associated major renal dysfunction.5 The exact mechanism of hypercalcemia is poorly understood. A widely accepted theory postulates that a granulomatous inflammatory infiltrate upregulates 1-α-hydroxylase activity, which enzymatically converts 25-hydroxyvitamin D to its active form, 1,25-dihydroxycholecalciferol, which increases bone resorption and calcium absorption through the gastrointestinal tract and renal systems. Treatments for hypercalcemia include hyperhydration, calcium-wasting diuretics, and low calcium intake.6 Furthermore, calcium levels should be obtained at the time of diagnosis and 30, 45, and 60 days after the lesions resolve.4

Subcutaneous fat necrosis needs to be differentiated from the more severe panniculitis, sclerema neonatorum (SN), which typically affects critically ill, preterm, and small-for-gestational-age newborns. It is associated with a high mortality rate and is characterized by skin and subadjacent tissue structures. The process typically begins in the thighs, buttocks, or trunk and spreads diffusely, sparing the fat-free palms, soles, and genitalia.7 Although our patient was born preterm, the physical characteristics of the nodule and the lack of severe illness placed SN lower on our differential. Histopathologic differences between SCFN and SN involve the extent of tissue fibrosis and presence of inflammatory cells. Sclerema neonatorum typically manifests with thickened connective tissue with a sparse inflammatory infiltrate, including lymphocytes, histiocytes, and multinucleated giant cells.7 Conversely, SCFN manifests with fat necrosis with an extensive inflammatory infiltrate. It is important to be able to distinguish between these 2 conditions, as both have vastly different prognoses.

Cold panniculitis, sometimes called “popsicle panniculitis,” is a phenomenon in which cold contact with the skin causes eruption of firm, erythematous, indurated plaques at the site of exposure. This self-limiting condition typically appears hours to days after cold exposure and spontaneously resolves in a few weeks.8 Therapeutic hypothermic protocol treatment involves using cooling devices to lower the body temperature for a short duration. The temperature typically is lowered to approximately 32 °C to 36 °C. These temperatures are not low enough to induce cold panniculitis, which is more commonly seen in facial ice applications when managing supraventricular tachycardia in neonates.

Cephalohematoma is a birthing injury that causes blood accumulation within the subperiosteal space. During parturition, the compressive and sheering forces on the calvarium rupture the vessels passing through the periosteum, causing blood to pool slowly into the subperiostium; thus, a cephalohematoma usually manifests later at 1 to 3 days of life as localized head swelling.9 The bleeding typically does not cross suture lines and is primarily found in the occipital or parietal regions. The incidence has been reported to be 0.4% to 2.5% of all live births.10 Although the location of the nodule in our patient was in the occipital region, imaging and biopsy results did not show hemorrhagic findings consistent with cephalohematoma. Management of cephalohematoma mainly is observational, as the mass slowly regresses and the accumulated blood gradually is reabsorbed.

Fungal scalp infections (tinea capitis) are common in the pediatric population. The peak incidence of this infection has been reported in children aged 3 to 7 years, with Trichophyton tonsurans and Microsporum canis as the usual causative organisms.11 Clinical features of tinea capitis include scaly patches with hair loss, hair loss with black pigmented dots at the follicular openings, diffuse scalp scaling with subtle hair loss, and cervical lymphadenopathy.12 Although less common, tinea capitis can progress to a more severe form known as a kerion, which is characterized by a tender plaque with pustules and crusting. A kerion can result in permanent scarring and alopecia if left untreated.12 In our patient, a nodule with scaling and faint erythema was observed, but no black pigmented dots at the follicular orifices were present. Therefore, a potassium hydroxide wet mount preparation used to diagnose tinea capitis was unnecessary. Systemic oral antifungal therapy such as fluconazole or terbinafine is the standard treatment for tinea capitis.

References
  1. Coondoo A, Lahiry R, Choudhury A, et al. Tender skin nodules in a newborn. Indian J Dermatol. 2013;58:328. doi:10.4103/0019-5154.113983
  2. Mitra S, Dove J, Somisetty SK. Subcutaneous fat necrosis in newbornan unusual case and review of literature. Eur J Pediatr. 2011;170:1107- 1110. doi:10.1007/s00431-011-1405-x
  3. Velasquez JH, Mendez MD. Newborn subcutaneous fat necrosis. In: StatPearls. StatPearls Publishing; 2022.
  4. Stefanko NS, Drolet BA. Subcutaneous fat necrosis of the newborn and associated hypercalcemia: a systematic review of the literature. Pediatr Dermatol. 2019;36:24-30. doi:10.1111/pde.13640
  5. Shumer DE, Thaker V, Taylor GA, et al. Severe hypercalcaemia due to subcutaneous fat necrosis: presentation, management and complications. Arch Dis Child Fetal Neonatal Ed. 2014;99:F419-F421. doi:10.1136/ archdischild-2014-306069
  6. Farooque A, Moss C, Zehnder D, et al. Expression of 25-hydroxyvitamin D3-1alpha-hydroxylase in subcutaneous fat necrosis. Br J Dermatol. 2009;160:423-425. doi:10.1111/j.1365-2133.2008.08844.x
  7. Zeb A, Darmstadt GL. Sclerema neonatorum: a review of nomenclature, clinical presentation, histological features, differential diagnoses and management. J Perinatol. 2008;28:453-460. doi:10.1038/jp.2008.33
  8. Quesada-Cortés A, Campos-Muñoz L, Díaz-Díaz RM, et al. Cold panniculitis. Dermatol Clin. 2008;26:485-489, vii. doi:10.1016 /j.det.2008.05.015
  9. Raines DA, Krawiec C, Jain S. Cephalohematoma. In: StatPearls. StatPearls Publishing; 2023.
  10. Chung HY, Chung JY, Lee DG, et al. Surgical treatment of ossified cephalhematoma. J Craniofac Surg. 2004;15:774-779. doi:10.1097/00001665- 200409000-00015
  11. Leung AKC, Hon KL, Leong KF, et al. Tinea capitis: an updated review. Recent Pat Inflamm Allergy Drug Discov. 2020;14:58-68. doi:10.2174/1872 213x14666200106145624
  12. Kovitwanichkanont T, Chong A. Superficial fungal infections. Aust J Gen Pract. 2019;48:706-711. doi:10.31128/ajgp-05-19-4930
References
  1. Coondoo A, Lahiry R, Choudhury A, et al. Tender skin nodules in a newborn. Indian J Dermatol. 2013;58:328. doi:10.4103/0019-5154.113983
  2. Mitra S, Dove J, Somisetty SK. Subcutaneous fat necrosis in newbornan unusual case and review of literature. Eur J Pediatr. 2011;170:1107- 1110. doi:10.1007/s00431-011-1405-x
  3. Velasquez JH, Mendez MD. Newborn subcutaneous fat necrosis. In: StatPearls. StatPearls Publishing; 2022.
  4. Stefanko NS, Drolet BA. Subcutaneous fat necrosis of the newborn and associated hypercalcemia: a systematic review of the literature. Pediatr Dermatol. 2019;36:24-30. doi:10.1111/pde.13640
  5. Shumer DE, Thaker V, Taylor GA, et al. Severe hypercalcaemia due to subcutaneous fat necrosis: presentation, management and complications. Arch Dis Child Fetal Neonatal Ed. 2014;99:F419-F421. doi:10.1136/ archdischild-2014-306069
  6. Farooque A, Moss C, Zehnder D, et al. Expression of 25-hydroxyvitamin D3-1alpha-hydroxylase in subcutaneous fat necrosis. Br J Dermatol. 2009;160:423-425. doi:10.1111/j.1365-2133.2008.08844.x
  7. Zeb A, Darmstadt GL. Sclerema neonatorum: a review of nomenclature, clinical presentation, histological features, differential diagnoses and management. J Perinatol. 2008;28:453-460. doi:10.1038/jp.2008.33
  8. Quesada-Cortés A, Campos-Muñoz L, Díaz-Díaz RM, et al. Cold panniculitis. Dermatol Clin. 2008;26:485-489, vii. doi:10.1016 /j.det.2008.05.015
  9. Raines DA, Krawiec C, Jain S. Cephalohematoma. In: StatPearls. StatPearls Publishing; 2023.
  10. Chung HY, Chung JY, Lee DG, et al. Surgical treatment of ossified cephalhematoma. J Craniofac Surg. 2004;15:774-779. doi:10.1097/00001665- 200409000-00015
  11. Leung AKC, Hon KL, Leong KF, et al. Tinea capitis: an updated review. Recent Pat Inflamm Allergy Drug Discov. 2020;14:58-68. doi:10.2174/1872 213x14666200106145624
  12. Kovitwanichkanont T, Chong A. Superficial fungal infections. Aust J Gen Pract. 2019;48:706-711. doi:10.31128/ajgp-05-19-4930
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
E27-E29
Page Number
E27-E29
Publications
Publications
Topics
Article Type
Display Headline
Occipital Scalp Nodule in a Newborn
Display Headline
Occipital Scalp Nodule in a Newborn
Sections
Questionnaire Body

A 4-week-old male infant was referred to dermatology for evaluation of a nodule on the occipital protuberance of 2 weeks’ duration. The patient was born at 36 weeks and 6 days’ gestation via an emergency cesarean delivery due to fetal distress. He later was found to have hypoxic-ischemic encephalopathy, pulmonary hypertension, and hypertrophic cardiomyopathy. He underwent therapeutic hypothermia protocol treatment starting at less than 6 hours after birth. At the current presentation, physical examination showed a 2.5-cm, erythematous, firm, mobile nodule on the occipital scalp with some overlying crusting and minimal surrounding erythema. No other cutaneous features or lesions were present. Initial laboratory findings were remarkable for hypercalcemia at 11 mg/dL (reference range, 8.5-10.5 mg/dL). Magnetic resonance imaging showed a faint abnormality in the subcutaneous tissue in this region without a noted connection to the underlying brain/meningeal matter. A punch biopsy was performed.

CT113004027_fig.jpg

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 04/24/2024 - 08:15
Un-Gate On Date
Wed, 04/24/2024 - 08:15
Use ProPublica
CFC Schedule Remove Status
Wed, 04/24/2024 - 08:15
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
Image
Disable zoom
Off

Tender Dermal Nodule on the Temple

Article Type
Changed
Mon, 05/06/2024 - 14:48
Display Headline
Tender Dermal Nodule on the Temple

The Diagnosis: Lymphoepithelioma-like Carcinoma

Lymphoepithelioma-like carcinoma (LELC) is a rare, poorly differentiated, primary cutaneous neoplasm that occurs on sun-exposed skin, particularly on the head and neck of elderly individuals. It often manifests as an asymptomatic, slow-growing, flesh-colored or erythematous dermal nodule, though ulceration and tenderness have been reported.1 Histopathologically, these neoplasms often are poorly circumscribed and can infiltrate surrounding subcutaneous and soft tissue. As a biphasic tumor, LELC is characterized by islands, nests, or trabeculae of epithelioid cells within the mid dermis surrounded by a dense lymphocytic infiltrate with plasma cells (Figure 1).1 The epithelial component rarely communicates with the overlying epidermis and is composed of atypical polygonal cells with eosinophilic cytoplasm, vesicular nuclei, prominent nucleoli, and frequent mitosis.2 These epithelial nests can be highlighted by pancytokeratin AE1/AE3 or other epithelial differentiation markers (eg, CAM 5.2, CK5/6, epithelial membrane antigen, high-molecular-weight cytokeratin), while the surrounding lymphocytic infiltrate consists of an admixture of T cells and B cells. Lymphoepithelioma-like carcinomas also can demonstrate sebaceous, eccrine, or follicular differentiations.3 The epithelial nests of LELC also are positive for p63 and epithelial membrane antigen.2

Chaiprasit_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Lymphoepithelial-like%20carcinoma.%20Dermal%20nests%20of%20epithelioid%20cells%20surrounded%20by%20a%20dense%20lymphocytic%20infiltrate%20are%20observed.%20The%20overlying%20epidermis%20is%20uninvolved%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%3C%2Fp%3E

The usual treatment of LELC is wide local excision or Mohs micrographic surgery.1 Despite the poorly differentiated morphology of the tumor, LELC has a generally good prognosis with low metastatic potential and few reports of local recurrence after incomplete excision.3 Patients who are not candidates for surgery as well as recalcitrant cases are managed with radiotherapy.1

Cutaneous lymphadenoma (CL) is a benign adnexal neoplasm that manifests as a small, solitary, fleshcolored nodule usually in the head and neck region.4 Histologically, CL consists of well-circumscribed epithelial nests within the dermis that are peripherally outlined by palisading basaloid cells and filled with clear to eosinophilic epithelioid cells (Figure 2).5 The fibrotic tumor stroma often is infiltrated by numerous intralobular dendritic cells and lymphocytes that occasionally can be arranged in germinal center–like nodules.4 The lymphoepithelial nature of CL can be challenging to distinguish morphologically from LELC, and immunohistochemistry stains may be required. In CL, both the basaloid and epithelioid cells stain positive for pancytokeratin AE1/ AE3, but the peripheral palisaded basaloid cells also stain positive for BerEP4. Additionally, the fibrotic stroma can be highlighted by CD34 and the intralobular dendritic cells by S-100.4

Chaiprasit_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Cutaneous%20lymphadenoma.%20Nests%20of%20clear%20epithelioid%20cells%20with%20peripheral%20basaloid%20cells%20are%20present%20within%20the%20dermis.%20The%20tumor%20stroma%20is%20fibrotic%20with%20lymphocytic%20infiltration%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

Nasopharyngeal carcinoma (NPC), formerly known as lymphoepithelioma, refers to carcinoma arising within the epithelium of the nasopharynx.6 Endemic to China, NPC manifests as an enlarging nasopharyngeal mass, causing clinical symptoms such as nasal obstruction and epistaxis.7 Histologically, nonkeratinizing NPC exhibits a biphasic morphology consisting of epithelioid neoplastic cells and background lymphocytic infiltrates (Figure 3). The epithelial component consists of round to oval neoplastic cells with amphophilic to eosinophilic cytoplasm, vesicular nuclei, and prominent nucleoli.6 Nasopharyngeal carcinoma is associated strongly with the Epstein-Barr virus while LELC is not; thus, Epstein- Barr encoding region in situ hybridization can reliably distinguish these entities. Metastatic NPC is rare but has been reported; therefore, it is highly recommended to perform an otolaryngologic examination in addition to testing for Epstein-Barr virus reactivity as part of a complete evaluation.8

Chaiprasit_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Nonkeratinizing%20nasopharyngeal%20carcinoma%20(formerly%20known%20as%20lymphoepithelioma).%20A%20dense%20lymphocytic%20infiltrate%20is%20present%20in%20the%20background%20of%20basaloid%20epithelial%20nests%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

Cutaneous squamous cell carcinoma (SCC) is a common epidermal malignancy with multiple subtypes and variable morphology. The clinical presentation of SCC is similar to LELC—an enlarging hyperkeratotic papule or nodule on sun-exposed skin that often is ulcerated and tender.9 Histologically, poorly differentiated nonkeratinizing SCC can form nests and trabeculae of epithelioid cells that are stained by epithelial differentiation markers, resembling the epithelioid nests of LELC. Distinguishing between LELC and poorly differentiated SCC with robust inflammatory infiltrate can be challenging (Figure 4). In fact, some experts support LELC as an SCC variant rather than a separate entity.9 However, in contrast to LELC, the dermal nests of SCC usually maintain an epidermal connection and often are associated with an overlying area of SCC in situ or welldifferentiated SCC.3

Chaiprasit_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Poorly%20differentiated%20squamous%20cell%20carcinoma.%20Robust%20inflammation%20is%20present%20as%20well%20as%20infiltrative%20nests%20and%20trabeculae%20of%20neoplastic%20epithelial%20cells%20with%20surrounding%20desmoplastic%20reaction%20and%20brisk%20mixed%20inflammatory%20infiltrate.%20The%20associated%20squamous%20cell%20carcinoma%20in%20situ%20also%20is%20present%20in%20the%20overlying%20epidermis%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

Mycosis fungoides (MF) is a primary cutaneous T-cell lymphoma. It is the most common type of cutaneous lymphoma, accounting for almost 50% of all reported cases.10 Classic MF has an indolent course and progresses through several clinical stages. Patches and plaques characterize early stages; lymphadenopathy indicates progression to later stages in which erythroderma may develop with coalescence of patches, plaques, and tumors; and MF present in blood or lymph nodes characterizes the late stage. Each stage of MF is different histologically—from a superficial lichenoid infiltrate with exocytosis of malignant T cells in the patch stage, to more robust epidermotropism and dermal infiltrate in the plaque stage, and finally a dense dermal infiltrate in the late stage.11 The rare syringotropic variant of MF clinically manifests as solitary or multiple erythematous lesions, often with overlying alopecia. Syringotropic MF uniquely exhibits folliculotropism and syringotropism along with syringometaplasia on histologic evaluation (Figure 5).12 The syringometaplasia can be difficult to distinguish from the epithelial nests of LELC, particularly with the lymphocytic background. Immunohistochemical panels for T-cell markers can highlight aberrant T cells in syringotropic MF through their usual loss of CD5 and CD7, in comparison to normal T cells in LELC.11 An elevated CD4:CD8 ratio of 4:1 and molecular analysis for T-cell receptor gene clonal rearrangements also can support the diagnosis of MF.12

Chaiprasit_5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20Syringotropic%20mycosis%20fungoides.%20A%20dermal%20infiltrate%20of%20neoplastic%20T%20cells%20exhibiting%20syringotropism%20can%20be%20observed.%20The%20involved%20eccrine%20glands%20shows%20focal%20areas%20of%20syringometaplasia%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

References
  1. Morteza Abedi S, Salama S, Alowami S. Lymphoepithelioma-like carcinoma of the skin: case report and approach to surgical pathology sign out. Rare Tumors. 2013;5:E47.
  2. Fisher JC, White RM, Hurd DS. Lymphoepithelioma-like carcinoma of the skin: a case of one patient presenting with two primary cutaneous neoplasms. J Am Osteopath Coll Dermatol. 2015;33:40-41.
  3. Welch PQ, Williams SB, Foss RD, et al. Lymphoepithelioma-like carcinoma of head and neck skin: a systematic analysis of 11 cases and review of literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2011;111:78-86.
  4. Yu R, Salama S, Alowami S. Cutaneous lymphadenoma: a rare case and brief review of a diagnostic pitfall. Rare Tumors. 2014;6:5358.
  5. Monteagudo C, Fúnez R, Sánchez-Sendra B, et al. Cutaneous lymphadenoma is a distinct trichoblastoma-like lymphoepithelial tumor with diffuse androgen receptor immunoreactivity, Notch1 ligand in Reed-Sternberg-like Cells, and common EGFR somatic mutations. Am J Surg Pathol. 2021;45:1382-1390.
  6. Stelow EB, Wenig BM. Update from the 4th edition of the World Health Organization classification of head and neck tumours: nasopharynx. Head Neck Pathol. 2017;11:16-22.
  7. Almomani MH, Zulfiqar H, Nagalli S. Nasopharyngeal carcinoma (NPC, lymphoepithelioma). StatPearls Publishing; 2022.
  8. Lassen CB, Lock-Andersen J. Lymphoepithelioma-like carcinoma of the skin: a case with perineural invasion. Plast Reconstr Surg Glob Open. 2014;2:E252.
  9. Motaparthi K, Kapil JP, Velazquez EF. Cutaneous squamous cell carcinoma: review of the eighth edition of the American Joint Committee on Cancer Staging Guidelines, Prognostic Factors, and Histopathologic Variants. Adv Anat Pathol. 2017;24:171-194.
  10. Pileri A, Facchetti F, Rütten A, et al. Syringotropic mycosis fungoides: a rare variant of the disease with peculiar clinicopathologic features. Am J Surg Pathol. 2011;35:100-109.
  11. Ryu HJ, Kim SI, Jang HO, et al. Evaluation of the International Society for Cutaneous Lymphoma Algorithm for the Diagnosis of Early Mycosis Fungoides [published October 15, 2021]. Cells. 2021;10:2758. doi:10.3390/cells10102758
  12. Lehmer LM, Amber KT, de Feraudy SM. Syringotropic mycosis fungoides: a rare form of cutaneous T-cell lymphoma enabling a histopathologic “sigh of relief.” Am J Dermatopathol. 2017;39:920-923.
Article PDF
Author and Disclosure Information

Drs. Chaiprasit, Modi, and Speiser are from the Department of Pathology, Loyola University Medical Center, Maywood, Illinois. Austin C. Casillas is from the Morehouse School of Medicine, Atlanta, Georgia.

The authors report no conflict of interest.

Correspondence: Thanchanok Chaiprasit, MD, Department of Pathology, Loyola University Medical Center, 2160 S First Ave, Building 110, Room 2232, Maywood, IL 60153 (thanchanok.chaiprasit@luhs.org).

Issue
Cutis - 113(4)
Publications
Topics
Page Number
170,174-176
Sections
Author and Disclosure Information

Drs. Chaiprasit, Modi, and Speiser are from the Department of Pathology, Loyola University Medical Center, Maywood, Illinois. Austin C. Casillas is from the Morehouse School of Medicine, Atlanta, Georgia.

The authors report no conflict of interest.

Correspondence: Thanchanok Chaiprasit, MD, Department of Pathology, Loyola University Medical Center, 2160 S First Ave, Building 110, Room 2232, Maywood, IL 60153 (thanchanok.chaiprasit@luhs.org).

Author and Disclosure Information

Drs. Chaiprasit, Modi, and Speiser are from the Department of Pathology, Loyola University Medical Center, Maywood, Illinois. Austin C. Casillas is from the Morehouse School of Medicine, Atlanta, Georgia.

The authors report no conflict of interest.

Correspondence: Thanchanok Chaiprasit, MD, Department of Pathology, Loyola University Medical Center, 2160 S First Ave, Building 110, Room 2232, Maywood, IL 60153 (thanchanok.chaiprasit@luhs.org).

Article PDF
Article PDF

The Diagnosis: Lymphoepithelioma-like Carcinoma

Lymphoepithelioma-like carcinoma (LELC) is a rare, poorly differentiated, primary cutaneous neoplasm that occurs on sun-exposed skin, particularly on the head and neck of elderly individuals. It often manifests as an asymptomatic, slow-growing, flesh-colored or erythematous dermal nodule, though ulceration and tenderness have been reported.1 Histopathologically, these neoplasms often are poorly circumscribed and can infiltrate surrounding subcutaneous and soft tissue. As a biphasic tumor, LELC is characterized by islands, nests, or trabeculae of epithelioid cells within the mid dermis surrounded by a dense lymphocytic infiltrate with plasma cells (Figure 1).1 The epithelial component rarely communicates with the overlying epidermis and is composed of atypical polygonal cells with eosinophilic cytoplasm, vesicular nuclei, prominent nucleoli, and frequent mitosis.2 These epithelial nests can be highlighted by pancytokeratin AE1/AE3 or other epithelial differentiation markers (eg, CAM 5.2, CK5/6, epithelial membrane antigen, high-molecular-weight cytokeratin), while the surrounding lymphocytic infiltrate consists of an admixture of T cells and B cells. Lymphoepithelioma-like carcinomas also can demonstrate sebaceous, eccrine, or follicular differentiations.3 The epithelial nests of LELC also are positive for p63 and epithelial membrane antigen.2

Chaiprasit_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Lymphoepithelial-like%20carcinoma.%20Dermal%20nests%20of%20epithelioid%20cells%20surrounded%20by%20a%20dense%20lymphocytic%20infiltrate%20are%20observed.%20The%20overlying%20epidermis%20is%20uninvolved%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%3C%2Fp%3E

The usual treatment of LELC is wide local excision or Mohs micrographic surgery.1 Despite the poorly differentiated morphology of the tumor, LELC has a generally good prognosis with low metastatic potential and few reports of local recurrence after incomplete excision.3 Patients who are not candidates for surgery as well as recalcitrant cases are managed with radiotherapy.1

Cutaneous lymphadenoma (CL) is a benign adnexal neoplasm that manifests as a small, solitary, fleshcolored nodule usually in the head and neck region.4 Histologically, CL consists of well-circumscribed epithelial nests within the dermis that are peripherally outlined by palisading basaloid cells and filled with clear to eosinophilic epithelioid cells (Figure 2).5 The fibrotic tumor stroma often is infiltrated by numerous intralobular dendritic cells and lymphocytes that occasionally can be arranged in germinal center–like nodules.4 The lymphoepithelial nature of CL can be challenging to distinguish morphologically from LELC, and immunohistochemistry stains may be required. In CL, both the basaloid and epithelioid cells stain positive for pancytokeratin AE1/ AE3, but the peripheral palisaded basaloid cells also stain positive for BerEP4. Additionally, the fibrotic stroma can be highlighted by CD34 and the intralobular dendritic cells by S-100.4

Chaiprasit_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Cutaneous%20lymphadenoma.%20Nests%20of%20clear%20epithelioid%20cells%20with%20peripheral%20basaloid%20cells%20are%20present%20within%20the%20dermis.%20The%20tumor%20stroma%20is%20fibrotic%20with%20lymphocytic%20infiltration%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

Nasopharyngeal carcinoma (NPC), formerly known as lymphoepithelioma, refers to carcinoma arising within the epithelium of the nasopharynx.6 Endemic to China, NPC manifests as an enlarging nasopharyngeal mass, causing clinical symptoms such as nasal obstruction and epistaxis.7 Histologically, nonkeratinizing NPC exhibits a biphasic morphology consisting of epithelioid neoplastic cells and background lymphocytic infiltrates (Figure 3). The epithelial component consists of round to oval neoplastic cells with amphophilic to eosinophilic cytoplasm, vesicular nuclei, and prominent nucleoli.6 Nasopharyngeal carcinoma is associated strongly with the Epstein-Barr virus while LELC is not; thus, Epstein- Barr encoding region in situ hybridization can reliably distinguish these entities. Metastatic NPC is rare but has been reported; therefore, it is highly recommended to perform an otolaryngologic examination in addition to testing for Epstein-Barr virus reactivity as part of a complete evaluation.8

Chaiprasit_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Nonkeratinizing%20nasopharyngeal%20carcinoma%20(formerly%20known%20as%20lymphoepithelioma).%20A%20dense%20lymphocytic%20infiltrate%20is%20present%20in%20the%20background%20of%20basaloid%20epithelial%20nests%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

Cutaneous squamous cell carcinoma (SCC) is a common epidermal malignancy with multiple subtypes and variable morphology. The clinical presentation of SCC is similar to LELC—an enlarging hyperkeratotic papule or nodule on sun-exposed skin that often is ulcerated and tender.9 Histologically, poorly differentiated nonkeratinizing SCC can form nests and trabeculae of epithelioid cells that are stained by epithelial differentiation markers, resembling the epithelioid nests of LELC. Distinguishing between LELC and poorly differentiated SCC with robust inflammatory infiltrate can be challenging (Figure 4). In fact, some experts support LELC as an SCC variant rather than a separate entity.9 However, in contrast to LELC, the dermal nests of SCC usually maintain an epidermal connection and often are associated with an overlying area of SCC in situ or welldifferentiated SCC.3

Chaiprasit_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Poorly%20differentiated%20squamous%20cell%20carcinoma.%20Robust%20inflammation%20is%20present%20as%20well%20as%20infiltrative%20nests%20and%20trabeculae%20of%20neoplastic%20epithelial%20cells%20with%20surrounding%20desmoplastic%20reaction%20and%20brisk%20mixed%20inflammatory%20infiltrate.%20The%20associated%20squamous%20cell%20carcinoma%20in%20situ%20also%20is%20present%20in%20the%20overlying%20epidermis%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

Mycosis fungoides (MF) is a primary cutaneous T-cell lymphoma. It is the most common type of cutaneous lymphoma, accounting for almost 50% of all reported cases.10 Classic MF has an indolent course and progresses through several clinical stages. Patches and plaques characterize early stages; lymphadenopathy indicates progression to later stages in which erythroderma may develop with coalescence of patches, plaques, and tumors; and MF present in blood or lymph nodes characterizes the late stage. Each stage of MF is different histologically—from a superficial lichenoid infiltrate with exocytosis of malignant T cells in the patch stage, to more robust epidermotropism and dermal infiltrate in the plaque stage, and finally a dense dermal infiltrate in the late stage.11 The rare syringotropic variant of MF clinically manifests as solitary or multiple erythematous lesions, often with overlying alopecia. Syringotropic MF uniquely exhibits folliculotropism and syringotropism along with syringometaplasia on histologic evaluation (Figure 5).12 The syringometaplasia can be difficult to distinguish from the epithelial nests of LELC, particularly with the lymphocytic background. Immunohistochemical panels for T-cell markers can highlight aberrant T cells in syringotropic MF through their usual loss of CD5 and CD7, in comparison to normal T cells in LELC.11 An elevated CD4:CD8 ratio of 4:1 and molecular analysis for T-cell receptor gene clonal rearrangements also can support the diagnosis of MF.12

Chaiprasit_5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20Syringotropic%20mycosis%20fungoides.%20A%20dermal%20infiltrate%20of%20neoplastic%20T%20cells%20exhibiting%20syringotropism%20can%20be%20observed.%20The%20involved%20eccrine%20glands%20shows%20focal%20areas%20of%20syringometaplasia%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

The Diagnosis: Lymphoepithelioma-like Carcinoma

Lymphoepithelioma-like carcinoma (LELC) is a rare, poorly differentiated, primary cutaneous neoplasm that occurs on sun-exposed skin, particularly on the head and neck of elderly individuals. It often manifests as an asymptomatic, slow-growing, flesh-colored or erythematous dermal nodule, though ulceration and tenderness have been reported.1 Histopathologically, these neoplasms often are poorly circumscribed and can infiltrate surrounding subcutaneous and soft tissue. As a biphasic tumor, LELC is characterized by islands, nests, or trabeculae of epithelioid cells within the mid dermis surrounded by a dense lymphocytic infiltrate with plasma cells (Figure 1).1 The epithelial component rarely communicates with the overlying epidermis and is composed of atypical polygonal cells with eosinophilic cytoplasm, vesicular nuclei, prominent nucleoli, and frequent mitosis.2 These epithelial nests can be highlighted by pancytokeratin AE1/AE3 or other epithelial differentiation markers (eg, CAM 5.2, CK5/6, epithelial membrane antigen, high-molecular-weight cytokeratin), while the surrounding lymphocytic infiltrate consists of an admixture of T cells and B cells. Lymphoepithelioma-like carcinomas also can demonstrate sebaceous, eccrine, or follicular differentiations.3 The epithelial nests of LELC also are positive for p63 and epithelial membrane antigen.2

Chaiprasit_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Lymphoepithelial-like%20carcinoma.%20Dermal%20nests%20of%20epithelioid%20cells%20surrounded%20by%20a%20dense%20lymphocytic%20infiltrate%20are%20observed.%20The%20overlying%20epidermis%20is%20uninvolved%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%3C%2Fp%3E

The usual treatment of LELC is wide local excision or Mohs micrographic surgery.1 Despite the poorly differentiated morphology of the tumor, LELC has a generally good prognosis with low metastatic potential and few reports of local recurrence after incomplete excision.3 Patients who are not candidates for surgery as well as recalcitrant cases are managed with radiotherapy.1

Cutaneous lymphadenoma (CL) is a benign adnexal neoplasm that manifests as a small, solitary, fleshcolored nodule usually in the head and neck region.4 Histologically, CL consists of well-circumscribed epithelial nests within the dermis that are peripherally outlined by palisading basaloid cells and filled with clear to eosinophilic epithelioid cells (Figure 2).5 The fibrotic tumor stroma often is infiltrated by numerous intralobular dendritic cells and lymphocytes that occasionally can be arranged in germinal center–like nodules.4 The lymphoepithelial nature of CL can be challenging to distinguish morphologically from LELC, and immunohistochemistry stains may be required. In CL, both the basaloid and epithelioid cells stain positive for pancytokeratin AE1/ AE3, but the peripheral palisaded basaloid cells also stain positive for BerEP4. Additionally, the fibrotic stroma can be highlighted by CD34 and the intralobular dendritic cells by S-100.4

Chaiprasit_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Cutaneous%20lymphadenoma.%20Nests%20of%20clear%20epithelioid%20cells%20with%20peripheral%20basaloid%20cells%20are%20present%20within%20the%20dermis.%20The%20tumor%20stroma%20is%20fibrotic%20with%20lymphocytic%20infiltration%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

Nasopharyngeal carcinoma (NPC), formerly known as lymphoepithelioma, refers to carcinoma arising within the epithelium of the nasopharynx.6 Endemic to China, NPC manifests as an enlarging nasopharyngeal mass, causing clinical symptoms such as nasal obstruction and epistaxis.7 Histologically, nonkeratinizing NPC exhibits a biphasic morphology consisting of epithelioid neoplastic cells and background lymphocytic infiltrates (Figure 3). The epithelial component consists of round to oval neoplastic cells with amphophilic to eosinophilic cytoplasm, vesicular nuclei, and prominent nucleoli.6 Nasopharyngeal carcinoma is associated strongly with the Epstein-Barr virus while LELC is not; thus, Epstein- Barr encoding region in situ hybridization can reliably distinguish these entities. Metastatic NPC is rare but has been reported; therefore, it is highly recommended to perform an otolaryngologic examination in addition to testing for Epstein-Barr virus reactivity as part of a complete evaluation.8

Chaiprasit_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Nonkeratinizing%20nasopharyngeal%20carcinoma%20(formerly%20known%20as%20lymphoepithelioma).%20A%20dense%20lymphocytic%20infiltrate%20is%20present%20in%20the%20background%20of%20basaloid%20epithelial%20nests%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

Cutaneous squamous cell carcinoma (SCC) is a common epidermal malignancy with multiple subtypes and variable morphology. The clinical presentation of SCC is similar to LELC—an enlarging hyperkeratotic papule or nodule on sun-exposed skin that often is ulcerated and tender.9 Histologically, poorly differentiated nonkeratinizing SCC can form nests and trabeculae of epithelioid cells that are stained by epithelial differentiation markers, resembling the epithelioid nests of LELC. Distinguishing between LELC and poorly differentiated SCC with robust inflammatory infiltrate can be challenging (Figure 4). In fact, some experts support LELC as an SCC variant rather than a separate entity.9 However, in contrast to LELC, the dermal nests of SCC usually maintain an epidermal connection and often are associated with an overlying area of SCC in situ or welldifferentiated SCC.3

Chaiprasit_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Poorly%20differentiated%20squamous%20cell%20carcinoma.%20Robust%20inflammation%20is%20present%20as%20well%20as%20infiltrative%20nests%20and%20trabeculae%20of%20neoplastic%20epithelial%20cells%20with%20surrounding%20desmoplastic%20reaction%20and%20brisk%20mixed%20inflammatory%20infiltrate.%20The%20associated%20squamous%20cell%20carcinoma%20in%20situ%20also%20is%20present%20in%20the%20overlying%20epidermis%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97100%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

Mycosis fungoides (MF) is a primary cutaneous T-cell lymphoma. It is the most common type of cutaneous lymphoma, accounting for almost 50% of all reported cases.10 Classic MF has an indolent course and progresses through several clinical stages. Patches and plaques characterize early stages; lymphadenopathy indicates progression to later stages in which erythroderma may develop with coalescence of patches, plaques, and tumors; and MF present in blood or lymph nodes characterizes the late stage. Each stage of MF is different histologically—from a superficial lichenoid infiltrate with exocytosis of malignant T cells in the patch stage, to more robust epidermotropism and dermal infiltrate in the plaque stage, and finally a dense dermal infiltrate in the late stage.11 The rare syringotropic variant of MF clinically manifests as solitary or multiple erythematous lesions, often with overlying alopecia. Syringotropic MF uniquely exhibits folliculotropism and syringotropism along with syringometaplasia on histologic evaluation (Figure 5).12 The syringometaplasia can be difficult to distinguish from the epithelial nests of LELC, particularly with the lymphocytic background. Immunohistochemical panels for T-cell markers can highlight aberrant T cells in syringotropic MF through their usual loss of CD5 and CD7, in comparison to normal T cells in LELC.11 An elevated CD4:CD8 ratio of 4:1 and molecular analysis for T-cell receptor gene clonal rearrangements also can support the diagnosis of MF.12

Chaiprasit_5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20Syringotropic%20mycosis%20fungoides.%20A%20dermal%20infiltrate%20of%20neoplastic%20T%20cells%20exhibiting%20syringotropism%20can%20be%20observed.%20The%20involved%20eccrine%20glands%20shows%20focal%20areas%20of%20syringometaplasia%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740%3B%20inset%3A%20original%20magnification%20%C3%97400).%3C%2Fp%3E

References
  1. Morteza Abedi S, Salama S, Alowami S. Lymphoepithelioma-like carcinoma of the skin: case report and approach to surgical pathology sign out. Rare Tumors. 2013;5:E47.
  2. Fisher JC, White RM, Hurd DS. Lymphoepithelioma-like carcinoma of the skin: a case of one patient presenting with two primary cutaneous neoplasms. J Am Osteopath Coll Dermatol. 2015;33:40-41.
  3. Welch PQ, Williams SB, Foss RD, et al. Lymphoepithelioma-like carcinoma of head and neck skin: a systematic analysis of 11 cases and review of literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2011;111:78-86.
  4. Yu R, Salama S, Alowami S. Cutaneous lymphadenoma: a rare case and brief review of a diagnostic pitfall. Rare Tumors. 2014;6:5358.
  5. Monteagudo C, Fúnez R, Sánchez-Sendra B, et al. Cutaneous lymphadenoma is a distinct trichoblastoma-like lymphoepithelial tumor with diffuse androgen receptor immunoreactivity, Notch1 ligand in Reed-Sternberg-like Cells, and common EGFR somatic mutations. Am J Surg Pathol. 2021;45:1382-1390.
  6. Stelow EB, Wenig BM. Update from the 4th edition of the World Health Organization classification of head and neck tumours: nasopharynx. Head Neck Pathol. 2017;11:16-22.
  7. Almomani MH, Zulfiqar H, Nagalli S. Nasopharyngeal carcinoma (NPC, lymphoepithelioma). StatPearls Publishing; 2022.
  8. Lassen CB, Lock-Andersen J. Lymphoepithelioma-like carcinoma of the skin: a case with perineural invasion. Plast Reconstr Surg Glob Open. 2014;2:E252.
  9. Motaparthi K, Kapil JP, Velazquez EF. Cutaneous squamous cell carcinoma: review of the eighth edition of the American Joint Committee on Cancer Staging Guidelines, Prognostic Factors, and Histopathologic Variants. Adv Anat Pathol. 2017;24:171-194.
  10. Pileri A, Facchetti F, Rütten A, et al. Syringotropic mycosis fungoides: a rare variant of the disease with peculiar clinicopathologic features. Am J Surg Pathol. 2011;35:100-109.
  11. Ryu HJ, Kim SI, Jang HO, et al. Evaluation of the International Society for Cutaneous Lymphoma Algorithm for the Diagnosis of Early Mycosis Fungoides [published October 15, 2021]. Cells. 2021;10:2758. doi:10.3390/cells10102758
  12. Lehmer LM, Amber KT, de Feraudy SM. Syringotropic mycosis fungoides: a rare form of cutaneous T-cell lymphoma enabling a histopathologic “sigh of relief.” Am J Dermatopathol. 2017;39:920-923.
References
  1. Morteza Abedi S, Salama S, Alowami S. Lymphoepithelioma-like carcinoma of the skin: case report and approach to surgical pathology sign out. Rare Tumors. 2013;5:E47.
  2. Fisher JC, White RM, Hurd DS. Lymphoepithelioma-like carcinoma of the skin: a case of one patient presenting with two primary cutaneous neoplasms. J Am Osteopath Coll Dermatol. 2015;33:40-41.
  3. Welch PQ, Williams SB, Foss RD, et al. Lymphoepithelioma-like carcinoma of head and neck skin: a systematic analysis of 11 cases and review of literature. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2011;111:78-86.
  4. Yu R, Salama S, Alowami S. Cutaneous lymphadenoma: a rare case and brief review of a diagnostic pitfall. Rare Tumors. 2014;6:5358.
  5. Monteagudo C, Fúnez R, Sánchez-Sendra B, et al. Cutaneous lymphadenoma is a distinct trichoblastoma-like lymphoepithelial tumor with diffuse androgen receptor immunoreactivity, Notch1 ligand in Reed-Sternberg-like Cells, and common EGFR somatic mutations. Am J Surg Pathol. 2021;45:1382-1390.
  6. Stelow EB, Wenig BM. Update from the 4th edition of the World Health Organization classification of head and neck tumours: nasopharynx. Head Neck Pathol. 2017;11:16-22.
  7. Almomani MH, Zulfiqar H, Nagalli S. Nasopharyngeal carcinoma (NPC, lymphoepithelioma). StatPearls Publishing; 2022.
  8. Lassen CB, Lock-Andersen J. Lymphoepithelioma-like carcinoma of the skin: a case with perineural invasion. Plast Reconstr Surg Glob Open. 2014;2:E252.
  9. Motaparthi K, Kapil JP, Velazquez EF. Cutaneous squamous cell carcinoma: review of the eighth edition of the American Joint Committee on Cancer Staging Guidelines, Prognostic Factors, and Histopathologic Variants. Adv Anat Pathol. 2017;24:171-194.
  10. Pileri A, Facchetti F, Rütten A, et al. Syringotropic mycosis fungoides: a rare variant of the disease with peculiar clinicopathologic features. Am J Surg Pathol. 2011;35:100-109.
  11. Ryu HJ, Kim SI, Jang HO, et al. Evaluation of the International Society for Cutaneous Lymphoma Algorithm for the Diagnosis of Early Mycosis Fungoides [published October 15, 2021]. Cells. 2021;10:2758. doi:10.3390/cells10102758
  12. Lehmer LM, Amber KT, de Feraudy SM. Syringotropic mycosis fungoides: a rare form of cutaneous T-cell lymphoma enabling a histopathologic “sigh of relief.” Am J Dermatopathol. 2017;39:920-923.
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
170,174-176
Page Number
170,174-176
Publications
Publications
Topics
Article Type
Display Headline
Tender Dermal Nodule on the Temple
Display Headline
Tender Dermal Nodule on the Temple
Sections
Questionnaire Body

A 77-year-old man presented with a 1.2-cm dermal nodule on the left temple of 1 year’s duration. The lesion had become tender and darker in color. An excision was performed and submitted for histologic examination. Additional immunohistochemistry staining for Epstein-Barr virus was negative.

Chaiprasit_Quiz.jpg
%3Cp%3EH%26amp%3BE%2C%20original%20magnification%20%C3%9720%3B%20inset%3A%20original%20magnification%20%C3%97100.%3C%2Fp%3E

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 04/08/2024 - 11:15
Un-Gate On Date
Mon, 04/08/2024 - 11:15
Use ProPublica
CFC Schedule Remove Status
Mon, 04/08/2024 - 11:15
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
Image
Teambase ID
180026F9.SIG
Disable zoom
Off

Centrifugally Spreading Lymphocutaneous Sporotrichosis: A Rare Cutaneous Manifestation

Article Type
Changed
Mon, 05/06/2024 - 14:26
Display Headline
Centrifugally Spreading Lymphocutaneous Sporotrichosis: A Rare Cutaneous Manifestation

To the Editor:

Sporotrichosis refers to a subacute to chronic fungal infection that usually involves the cutaneous and subcutaneous tissues and is caused by the introduction of Sporothrix, a dimorphic fungus, through the skin. We present a case of chronic atypical lymphocutaneous sporotrichosis.

A 46-year-old man presented to the outpatient dermatology clinic for follow-up for a rash on the right leg that spread to the thigh and became painful and pruritic. It initially developed 8 years prior to the current presentation after he sustained trauma to the leg from an electroshock weapon. One year prior to the current presentation, he had presented to the emergency department and was prescribed doxycycline 100 mg twice daily for 7 days as well as bacitracin ointment. He also was instructed to follow up with dermatology, but a lack of health insurance and other socioeconomic barriers prevented him from seeking dermatologic care. Nine months later, he again presented to the emergency department due to a motor vehicle accident. Computed tomography (CT) of the right leg revealed exophytic dermal masses, inflammatory stranding of the subcutaneous tissue, and right inguinal lymph nodes measuring up to 1.4 cm; there was no osteoarticular involvement. At that time, the patient was applying gentian violet to the skin lesions and taking hydroxyzine 50 mg 3 times daily as needed for pruritus with minimal relief. Financial support was provided for follow-up with dermatology, which occurred almost 5 months later.

At the current presentation, physical examination revealed a large annular plaque with verrucous, scaly, erythematous borders and a hypopigmented atrophic center extending from the medial aspect of the right leg to the posterior thigh. Numerous pink, scaly, crusted nodules were scattered primarily along the periphery, with some evidence of draining sinus tracts. In addition, a fibrotic pink linear plaque extended from the medial right leg to the popliteal fossa, consistent with a keloid. Violet staining along the periphery of the lesion also was appreciated secondary to the application of topical gentian violet (Figure 1).

CT113004006_e_Fig1_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20and%20B%2C%20Annular%20verrucous%20plaque%20with%20central%20hypopigmentation%20extending%20from%20the%20medial%20right%20leg%20to%20the%20posterior%20right%20thigh%20with%20keloid%20formation%20at%20the%20proximal%20edge.%20Violaceous%20staining%20was%20seen%20secondary%20to%20topical%20application%20of%20gentian%20violet.%3C%2Fp%3E

Based on the chronic history and morphology, a diagnosis of a chronic fungal or atypical mycobacterial infection was favored. In particular, chromoblastomycosis, cutaneous tuberculosis (eg, scrofuloderma, lupus vulgaris, tuberculosis verrucosa cutis), and atypical mycobacterial infection were highest on the differential, as these conditions often exhibit annular, nodular, verrucous, and/or atrophic lesions. The nodularity, crusting, and draining sinus tracts also raised the possibility of mycetoma. Given the extension of the lesion from the lower to upper leg, a sporotrichoid infection also was considered but was thought to be less likely based on the annular configuration.

Two 4-mm punch biopsies were taken from a peripheral nodule—one for routine histology and another for bacterial, fungal, and mycobacterial cultures. An ­interferon-gamma release assay also was ordered to evaluate for immune responses indicative of prior Mycobacterium tuberculosis infection, but the patient did not obtain this for unknown reasons. Histology demonstrated pseudoepitheliomatous hyperplasia and necrotizing granulomas, which suggested an infectious etiology, but no organisms were identified on tissue staining and all cultures were negative for growth at 6 weeks. The patient was asked to return at that point, and 4 additional scouting biopsies were performed and sent for routine histology, M tuberculosis nucleic acid amplification testing, and microbiologic cultures (ie, bacterial, mycobacterial, fungal, nocardia, actinomycetes). Within 1 week, a filamentous organism with pigmentation visible on the front and back of a Sabouraud dextrose agar plate was identified on fungal culture (Figure 2). Microscopic evaluation of this mold with lactophenol blue stain revealed thin septate hyphae with conidiophores arising at right angles that bore clusters of microconidia (Figure 3). Sequencing analysis ultimately identified this organism as Sporothrix schenckii. Routine histology demonstrated pseudoepitheliomatous hyperplasia with scattered intraepidermal collections of neutrophils (Figure 4). The dermis showed a dense, superficial, and deep infiltrate composed of lymphocytes, histiocytes, and plasma cells with occasional neutrophils and eosinophils. A Grocott-Gomori methenamine-silver stain revealed a cluster of ovoid yeast forms within the stratum corneum (Figure 5). The patient was referred to infectious disease for follow-up and treatment.

Aggarwal_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Anterior%20view%20of%20darkly%20pigmented%2C%20filamentous%20fungi%20grown%20on%20a%20Sabouraud%20dextrose%20agar%20culture%20plate%20at%2025%20%C2%B0C.%20Pigmentation%20was%20visible%20on%20the%20reverse%20view%20of%20the%20culture%20plate%20demonstrating%20that%20the%20mold%20is%20dematiaceous%20(inset).%3C%2Fp%3E

The patient later visited a community clinic providing dermatologic care for patients without insurance. He was started on itraconazole 200 mg daily for a total of 6 months until dermatologic clearance of the cutaneous lesions was observed. He was followed by the clinic with laboratory tests including a liver function test. At follow-up 8 months later, a repeat biopsy was performed to ensure histologic clearance of the sporotrichosis, which revealed a dermal scar and no evidence of residual infection.

Aggarwal_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Lactophenol%20blue%20stain%20of%20the%20mold%20form%20demonstrated%20septate%20hyphae%20with%20conidiophores%20arising%20at%20right%20angles%20and%20clustered%20microconidia%20resembling%20rose%20petals%20(original%20magnification%20%C3%9760).%20Reference%20bar%20indicates%2020%20%C2%B5m.%3C%2Fp%3E

Sporothrix schenckii was first isolated in 1898 by Benjamin Schenck, a student at Johns Hopkins Medicine (Baltimore, Maryland), and identified by a mycologist as sporotricha.1 Species within the genus Sporothrix are unique in that the fungi are both dimorphic (growing as a mold at 25 °C but as a yeast at 37 °C) and dematiaceous (dark pigmentation from melanin is visible on inspection of the anterior and reverse sides of culture plates). Infection usually occurs when cutaneous or subcutaneous tissues are exposed to the fungus via microabrasions; activities thought to contribute to exposure include gardening, agricultural work, animal husbandry, and feline scratches.2 Although skin trauma frequently is considered the primary route of infection, patient recall is variable, with one study noting that only 37.7% of patients recalled trauma and another study similarly demonstrating a patient recall rate of 25%.3,4

Aggarwal_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Histopathology%20demonstrated%20pseudoepitheliomatous%20hyperplasia%20with%20intraepidermal%20collections%20of%20neutrophils%20and%20a%20dense%20mixed%20inflammatory%20dermal%20infiltrate%20(H%26amp%3BE%2C%20original%20magnification%20%C3%974).%3C%2Fp%3E

Lymphocutaneous sporotrichosis is the most common presentation of the fungal infection,5 and clinical cases may be classified into 1 of 4 categories: (1) lymphangitic lesions—papules at the site of inoculation with spread along the lymphatic channels; (2) localized (fixed) cutaneous lesions—1 or 2 lesions at the inoculation site; (3) disseminated (multifocal) cutaneous lesions; and (4) extracutaneous lesions.6 Extracutaneous manifestations of this infection most notably have been reported as pulmonary disease through inhalation of conidia or through dissemination in immunocompromised hosts.7 Our patient’s infection was categorized as lymphangitic lesions due to spread from the lower to upper leg, albeit in a highly atypical, annular fashion. A review of systems was otherwise negative, and CT ruled out osteoarticular involvement.

Aggarwal_5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20Grocott-Gomori%20methenamine-silver%20stain%20demonstrated%20a%20cluster%20of%20ovoid%20yeast%20forms%20within%20the%20stratum%20corneum%20(original%20magnification%20%C3%9760).%20Reference%20bar%20indicates%2020%20%C2%B5m.%3C%2Fp%3E

In addition to socioeconomic barriers, several factors contributed to a delayed diagnosis in this patient including the annular presentation with central hypopigmentation and atrophy, negative initial microbiological cultures and lack of visualization of organisms on histopathology, and the consequent need for repeat biopsies. For lymphocutaneous sporotrichosis, the typical presentation consists of a papule or ulcerated nodule at the site of inoculation with subsequent linear spread along lymphatic channels. This classic sporotrichoid pattern is a key diagnostic clue for identifying sporotrichosis but was absent at the time our patient presented for medical care. Rather, the sporotrichoid spread seemed to have occurred in a centrifugal fashion up the leg. Few case reports have documented an annular presentation of lymphocutaneous sporotrichosis,8-13 and one report described central atrophy and hypopigmentation.10 Pain and pruritus, which were present in our patient, rarely are documented.9 Finally, the diagnosis of cutaneous fungal infections may require multiple biopsies due to the variable abundance of viable organisms in tissue specimens as well as the fastidious growth characteristics of these organisms. Furthermore, sensitivity often is low for both fungal and mycobacterial cultures, and cultures may take days to weeks to yield growth.14,15 For these reasons, empiric therapy and repeat biopsies often are pursued if clinical suspicion is high enough.16 Our patient returned for multiple scouting biopsies after the initial tissue culture was negative and was even considered for empiric treatment against Mycobacterium prior to positive fungal cultures.

Another unique aspect of our case was the presence of a keloid. It is difficult to know if this keloid was secondary to the trauma the patient sustained in the inciting incident or formed from the fungal infection. Interestingly, it has been hypothesized that fungal infections may contribute to keloid and hypertrophic scar formation.17 In a case series of 3 patients with either keloids or hypertrophic scars and concomitant tinea infection, there was notable improvement in the appearance of the scars 2 weeks after beginning itraconazole therapy.17 However, it is not yet known if a fungal infection can contribute to the pathogenesis of keloid formation.

As with other aspects of this case, the length of time the patient went without diagnosis and treatment was unusual and may help explain the atypical presentation. Although the incubation period for S schenckii can vary, most reports identify patients as seeking medical attention within 1 year of rash onset.18-20 In our case, the patient was not diagnosed until 8 years after his symptoms began, requiring multiple referrals, multiple health system touchpoints, and an institution-specific financial aid program. As such, this case also highlights the potential need for a multidisciplinary team approach when caring for patients with poor access to health care.

In conclusion, this case illustrates a unique presentation of lymphocutaneous sporotrichosis that may mimic other chronic infections and result in delayed diagnosis. Although lymphangitic sporotrichosis generally is recognized as having a linear distribution, mounting evidence from this report and others suggests an annular presentation also is possible. Pruritus or pain is rare but should not preclude a diagnosis of sporotrichosis if present. For patients with limited access to health care resources, it is especially important to involve multiple members of the health care team, including social workers and specialists, to prevent a protracted and severe course of disease.

References
  1. Schenck BR. On refractory subcutaneous abscesses caused by a fungus possibly related to the sporotricha. Bulletin of the Johns Hopkins Hospital. 1898;93:286-290.
  2. de Lima Barros MB, de Almeida Paes R, Schubach AO. Sporothrix schenckii and sporotrichosis. Clin Microbiol Rev. 2011;24:633-654. doi:10.1128/CMR.00007-11
  3. Crevasse L, Ellner PD. An outbreak of sporotrichosis in florida. J Am Med Assoc. 1960;173:29-33. doi:10.1001/jama.1960.03020190031006
  4. Mayorga R, Cáceres A, Toriello C, et al. An endemic area of sporotrichosis in Guatemala [in French]. Sabouraudia. 1978;16:185-198.
  5. Morris-Jones R. Sporotrichosis. Clin Exp Dermatol. 2002;27:427-431. doi:10.1046/j.1365-2230.2002.01087.x
  6. Sampaio SA, Da Lacaz CS. Clinical and statistical studies on sporotrichosis in Sao Paulo (Brazil). Article in German. Hautarzt. 1959;10:490-493.
  7. Ramos-e-Silva M, Vasconcelos C, Carneiro S, et al. Sporotrichosis. Clin Dermatol. 2007;25:181-187. doi:10.1016/j.clindermatol.2006.05.006
  8. Williams BA, Jennings TA, Rushing EC, et al. Sporotrichosis on the face of a 7-year-old boy following a bicycle accident. Pediatr Dermatol. 2013;30:E246-E247. doi:10.1111/j.1525-1470.2011.01696.x
  9. Vaishampayan SS, Borde P. An unusual presentation of sporotrichosis. Indian J Dermatol. 2013;58:409. doi:10.4103/0019-5154.117350
  10. Qin J, Zhang J. Sporotrichosis. N Engl J Med. 2019;380:771. doi:10.1056/NEJMicm1809179
  11. Patel A, Mudenda V, Lakhi S, et al. A 27-year-old severely immunosuppressed female with misleading clinical features of disseminated cutaneous sporotrichosis. Case Rep Dermatol Med. 2016;2016:1-4. doi:10.1155/2016/9403690
  12. de Oliveira-Esteves ICMR, Almeida Rosa da Silva G, Eyer-Silva WA, et al. Rapidly progressive disseminated sporotrichosis as the first presentation of HIV infection in a patient with a very low CD4 cell count. Case Rep Infect Dis. 2017;2017:4713140. doi:10.1155/2017/4713140
  13. Singh S, Bachaspatimayum R, Meetei U, et al. Terbinafine in fixed cutaneous sporotrichosis: a case series. J Clin Diagnostic Res. 2018;12:FR01-FR03. doi:10.7860/JCDR/2018/25315.12223
  14. Guarner J, Brandt ME. Histopathologic diagnosis of fungal infections in the 21st century. Clin Microbiol Rev. 2011;24:247-280. doi:10.1128/CMR.00053-10
  15. Peters F, Batinica M, Plum G, et al. Bug or no bug: challenges in diagnosing cutaneous mycobacterial infections. J Ger Soc Dermatol. 2016;14:1227-1236. doi:10.1111/ddg.13001
  16. Khadka P, Koirala S, Thapaliya J. Cutaneous tuberculosis: clinicopathologic arrays and diagnostic challenges. Dermatol Res Pract. 2018;2018:7201973. doi:10.1155/2018/7201973
  17. Okada E, Maruyama Y. Are keloids and hypertrophic scars caused by fungal infection? . Plast Reconstr Surg. 2007;120:814-815. doi:10.1097/01.prs.0000278813.23244.3f
  18. Pappas PG, Tellez I, Deep AE, et al. Sporotrichosis in Peru: description of an area of hyperendemicity. Clin Infect Dis. 2000;30:65-70. doi:10.1086/313607
  19. McGuinness SL, Boyd R, Kidd S, et al. Epidemiological investigation of an outbreak of cutaneous sporotrichosis, Northern Territory, Australia. BMC Infect Dis. 2016;16:1-7. doi:10.1186/s12879-016-1338-0
  20. Rojas FD, Fernández MS, Lucchelli JM, et al. Cavitary pulmonary sporotrichosis: case report and literature review. Mycopathologia. 2017;182:1119-1123. doi:10.1007/s11046-017-0197-6
Article PDF
Author and Disclosure Information

From The Johns Hopkins University School of Medicine, Baltimore, Maryland. Prachi Aggarwal as well as Drs. Rogers, Grenier, and Alhariri are from the Department of Dermatology; Drs. Gaston and Zhang are from the Department of Pathology; and Dr. Shah is from the Division of Infectious Disease.

The authors report no conflict of interest.

Correspondence: Jihad Alhariri, MD, MPH, Cancer Research Building II, Johns Hopkins University School of Medicine, 1550 Orleans St, Baltimore, MD 21231 (jalhari1@jhmi.edu).

Issue
Cutis - 113(4)
Publications
Topics
Page Number
E6-E9
Sections
Author and Disclosure Information

From The Johns Hopkins University School of Medicine, Baltimore, Maryland. Prachi Aggarwal as well as Drs. Rogers, Grenier, and Alhariri are from the Department of Dermatology; Drs. Gaston and Zhang are from the Department of Pathology; and Dr. Shah is from the Division of Infectious Disease.

The authors report no conflict of interest.

Correspondence: Jihad Alhariri, MD, MPH, Cancer Research Building II, Johns Hopkins University School of Medicine, 1550 Orleans St, Baltimore, MD 21231 (jalhari1@jhmi.edu).

Author and Disclosure Information

From The Johns Hopkins University School of Medicine, Baltimore, Maryland. Prachi Aggarwal as well as Drs. Rogers, Grenier, and Alhariri are from the Department of Dermatology; Drs. Gaston and Zhang are from the Department of Pathology; and Dr. Shah is from the Division of Infectious Disease.

The authors report no conflict of interest.

Correspondence: Jihad Alhariri, MD, MPH, Cancer Research Building II, Johns Hopkins University School of Medicine, 1550 Orleans St, Baltimore, MD 21231 (jalhari1@jhmi.edu).

Article PDF
Article PDF

To the Editor:

Sporotrichosis refers to a subacute to chronic fungal infection that usually involves the cutaneous and subcutaneous tissues and is caused by the introduction of Sporothrix, a dimorphic fungus, through the skin. We present a case of chronic atypical lymphocutaneous sporotrichosis.

A 46-year-old man presented to the outpatient dermatology clinic for follow-up for a rash on the right leg that spread to the thigh and became painful and pruritic. It initially developed 8 years prior to the current presentation after he sustained trauma to the leg from an electroshock weapon. One year prior to the current presentation, he had presented to the emergency department and was prescribed doxycycline 100 mg twice daily for 7 days as well as bacitracin ointment. He also was instructed to follow up with dermatology, but a lack of health insurance and other socioeconomic barriers prevented him from seeking dermatologic care. Nine months later, he again presented to the emergency department due to a motor vehicle accident. Computed tomography (CT) of the right leg revealed exophytic dermal masses, inflammatory stranding of the subcutaneous tissue, and right inguinal lymph nodes measuring up to 1.4 cm; there was no osteoarticular involvement. At that time, the patient was applying gentian violet to the skin lesions and taking hydroxyzine 50 mg 3 times daily as needed for pruritus with minimal relief. Financial support was provided for follow-up with dermatology, which occurred almost 5 months later.

At the current presentation, physical examination revealed a large annular plaque with verrucous, scaly, erythematous borders and a hypopigmented atrophic center extending from the medial aspect of the right leg to the posterior thigh. Numerous pink, scaly, crusted nodules were scattered primarily along the periphery, with some evidence of draining sinus tracts. In addition, a fibrotic pink linear plaque extended from the medial right leg to the popliteal fossa, consistent with a keloid. Violet staining along the periphery of the lesion also was appreciated secondary to the application of topical gentian violet (Figure 1).

CT113004006_e_Fig1_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20and%20B%2C%20Annular%20verrucous%20plaque%20with%20central%20hypopigmentation%20extending%20from%20the%20medial%20right%20leg%20to%20the%20posterior%20right%20thigh%20with%20keloid%20formation%20at%20the%20proximal%20edge.%20Violaceous%20staining%20was%20seen%20secondary%20to%20topical%20application%20of%20gentian%20violet.%3C%2Fp%3E

Based on the chronic history and morphology, a diagnosis of a chronic fungal or atypical mycobacterial infection was favored. In particular, chromoblastomycosis, cutaneous tuberculosis (eg, scrofuloderma, lupus vulgaris, tuberculosis verrucosa cutis), and atypical mycobacterial infection were highest on the differential, as these conditions often exhibit annular, nodular, verrucous, and/or atrophic lesions. The nodularity, crusting, and draining sinus tracts also raised the possibility of mycetoma. Given the extension of the lesion from the lower to upper leg, a sporotrichoid infection also was considered but was thought to be less likely based on the annular configuration.

Two 4-mm punch biopsies were taken from a peripheral nodule—one for routine histology and another for bacterial, fungal, and mycobacterial cultures. An ­interferon-gamma release assay also was ordered to evaluate for immune responses indicative of prior Mycobacterium tuberculosis infection, but the patient did not obtain this for unknown reasons. Histology demonstrated pseudoepitheliomatous hyperplasia and necrotizing granulomas, which suggested an infectious etiology, but no organisms were identified on tissue staining and all cultures were negative for growth at 6 weeks. The patient was asked to return at that point, and 4 additional scouting biopsies were performed and sent for routine histology, M tuberculosis nucleic acid amplification testing, and microbiologic cultures (ie, bacterial, mycobacterial, fungal, nocardia, actinomycetes). Within 1 week, a filamentous organism with pigmentation visible on the front and back of a Sabouraud dextrose agar plate was identified on fungal culture (Figure 2). Microscopic evaluation of this mold with lactophenol blue stain revealed thin septate hyphae with conidiophores arising at right angles that bore clusters of microconidia (Figure 3). Sequencing analysis ultimately identified this organism as Sporothrix schenckii. Routine histology demonstrated pseudoepitheliomatous hyperplasia with scattered intraepidermal collections of neutrophils (Figure 4). The dermis showed a dense, superficial, and deep infiltrate composed of lymphocytes, histiocytes, and plasma cells with occasional neutrophils and eosinophils. A Grocott-Gomori methenamine-silver stain revealed a cluster of ovoid yeast forms within the stratum corneum (Figure 5). The patient was referred to infectious disease for follow-up and treatment.

Aggarwal_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Anterior%20view%20of%20darkly%20pigmented%2C%20filamentous%20fungi%20grown%20on%20a%20Sabouraud%20dextrose%20agar%20culture%20plate%20at%2025%20%C2%B0C.%20Pigmentation%20was%20visible%20on%20the%20reverse%20view%20of%20the%20culture%20plate%20demonstrating%20that%20the%20mold%20is%20dematiaceous%20(inset).%3C%2Fp%3E

The patient later visited a community clinic providing dermatologic care for patients without insurance. He was started on itraconazole 200 mg daily for a total of 6 months until dermatologic clearance of the cutaneous lesions was observed. He was followed by the clinic with laboratory tests including a liver function test. At follow-up 8 months later, a repeat biopsy was performed to ensure histologic clearance of the sporotrichosis, which revealed a dermal scar and no evidence of residual infection.

Aggarwal_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Lactophenol%20blue%20stain%20of%20the%20mold%20form%20demonstrated%20septate%20hyphae%20with%20conidiophores%20arising%20at%20right%20angles%20and%20clustered%20microconidia%20resembling%20rose%20petals%20(original%20magnification%20%C3%9760).%20Reference%20bar%20indicates%2020%20%C2%B5m.%3C%2Fp%3E

Sporothrix schenckii was first isolated in 1898 by Benjamin Schenck, a student at Johns Hopkins Medicine (Baltimore, Maryland), and identified by a mycologist as sporotricha.1 Species within the genus Sporothrix are unique in that the fungi are both dimorphic (growing as a mold at 25 °C but as a yeast at 37 °C) and dematiaceous (dark pigmentation from melanin is visible on inspection of the anterior and reverse sides of culture plates). Infection usually occurs when cutaneous or subcutaneous tissues are exposed to the fungus via microabrasions; activities thought to contribute to exposure include gardening, agricultural work, animal husbandry, and feline scratches.2 Although skin trauma frequently is considered the primary route of infection, patient recall is variable, with one study noting that only 37.7% of patients recalled trauma and another study similarly demonstrating a patient recall rate of 25%.3,4

Aggarwal_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Histopathology%20demonstrated%20pseudoepitheliomatous%20hyperplasia%20with%20intraepidermal%20collections%20of%20neutrophils%20and%20a%20dense%20mixed%20inflammatory%20dermal%20infiltrate%20(H%26amp%3BE%2C%20original%20magnification%20%C3%974).%3C%2Fp%3E

Lymphocutaneous sporotrichosis is the most common presentation of the fungal infection,5 and clinical cases may be classified into 1 of 4 categories: (1) lymphangitic lesions—papules at the site of inoculation with spread along the lymphatic channels; (2) localized (fixed) cutaneous lesions—1 or 2 lesions at the inoculation site; (3) disseminated (multifocal) cutaneous lesions; and (4) extracutaneous lesions.6 Extracutaneous manifestations of this infection most notably have been reported as pulmonary disease through inhalation of conidia or through dissemination in immunocompromised hosts.7 Our patient’s infection was categorized as lymphangitic lesions due to spread from the lower to upper leg, albeit in a highly atypical, annular fashion. A review of systems was otherwise negative, and CT ruled out osteoarticular involvement.

Aggarwal_5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20Grocott-Gomori%20methenamine-silver%20stain%20demonstrated%20a%20cluster%20of%20ovoid%20yeast%20forms%20within%20the%20stratum%20corneum%20(original%20magnification%20%C3%9760).%20Reference%20bar%20indicates%2020%20%C2%B5m.%3C%2Fp%3E

In addition to socioeconomic barriers, several factors contributed to a delayed diagnosis in this patient including the annular presentation with central hypopigmentation and atrophy, negative initial microbiological cultures and lack of visualization of organisms on histopathology, and the consequent need for repeat biopsies. For lymphocutaneous sporotrichosis, the typical presentation consists of a papule or ulcerated nodule at the site of inoculation with subsequent linear spread along lymphatic channels. This classic sporotrichoid pattern is a key diagnostic clue for identifying sporotrichosis but was absent at the time our patient presented for medical care. Rather, the sporotrichoid spread seemed to have occurred in a centrifugal fashion up the leg. Few case reports have documented an annular presentation of lymphocutaneous sporotrichosis,8-13 and one report described central atrophy and hypopigmentation.10 Pain and pruritus, which were present in our patient, rarely are documented.9 Finally, the diagnosis of cutaneous fungal infections may require multiple biopsies due to the variable abundance of viable organisms in tissue specimens as well as the fastidious growth characteristics of these organisms. Furthermore, sensitivity often is low for both fungal and mycobacterial cultures, and cultures may take days to weeks to yield growth.14,15 For these reasons, empiric therapy and repeat biopsies often are pursued if clinical suspicion is high enough.16 Our patient returned for multiple scouting biopsies after the initial tissue culture was negative and was even considered for empiric treatment against Mycobacterium prior to positive fungal cultures.

Another unique aspect of our case was the presence of a keloid. It is difficult to know if this keloid was secondary to the trauma the patient sustained in the inciting incident or formed from the fungal infection. Interestingly, it has been hypothesized that fungal infections may contribute to keloid and hypertrophic scar formation.17 In a case series of 3 patients with either keloids or hypertrophic scars and concomitant tinea infection, there was notable improvement in the appearance of the scars 2 weeks after beginning itraconazole therapy.17 However, it is not yet known if a fungal infection can contribute to the pathogenesis of keloid formation.

As with other aspects of this case, the length of time the patient went without diagnosis and treatment was unusual and may help explain the atypical presentation. Although the incubation period for S schenckii can vary, most reports identify patients as seeking medical attention within 1 year of rash onset.18-20 In our case, the patient was not diagnosed until 8 years after his symptoms began, requiring multiple referrals, multiple health system touchpoints, and an institution-specific financial aid program. As such, this case also highlights the potential need for a multidisciplinary team approach when caring for patients with poor access to health care.

In conclusion, this case illustrates a unique presentation of lymphocutaneous sporotrichosis that may mimic other chronic infections and result in delayed diagnosis. Although lymphangitic sporotrichosis generally is recognized as having a linear distribution, mounting evidence from this report and others suggests an annular presentation also is possible. Pruritus or pain is rare but should not preclude a diagnosis of sporotrichosis if present. For patients with limited access to health care resources, it is especially important to involve multiple members of the health care team, including social workers and specialists, to prevent a protracted and severe course of disease.

To the Editor:

Sporotrichosis refers to a subacute to chronic fungal infection that usually involves the cutaneous and subcutaneous tissues and is caused by the introduction of Sporothrix, a dimorphic fungus, through the skin. We present a case of chronic atypical lymphocutaneous sporotrichosis.

A 46-year-old man presented to the outpatient dermatology clinic for follow-up for a rash on the right leg that spread to the thigh and became painful and pruritic. It initially developed 8 years prior to the current presentation after he sustained trauma to the leg from an electroshock weapon. One year prior to the current presentation, he had presented to the emergency department and was prescribed doxycycline 100 mg twice daily for 7 days as well as bacitracin ointment. He also was instructed to follow up with dermatology, but a lack of health insurance and other socioeconomic barriers prevented him from seeking dermatologic care. Nine months later, he again presented to the emergency department due to a motor vehicle accident. Computed tomography (CT) of the right leg revealed exophytic dermal masses, inflammatory stranding of the subcutaneous tissue, and right inguinal lymph nodes measuring up to 1.4 cm; there was no osteoarticular involvement. At that time, the patient was applying gentian violet to the skin lesions and taking hydroxyzine 50 mg 3 times daily as needed for pruritus with minimal relief. Financial support was provided for follow-up with dermatology, which occurred almost 5 months later.

At the current presentation, physical examination revealed a large annular plaque with verrucous, scaly, erythematous borders and a hypopigmented atrophic center extending from the medial aspect of the right leg to the posterior thigh. Numerous pink, scaly, crusted nodules were scattered primarily along the periphery, with some evidence of draining sinus tracts. In addition, a fibrotic pink linear plaque extended from the medial right leg to the popliteal fossa, consistent with a keloid. Violet staining along the periphery of the lesion also was appreciated secondary to the application of topical gentian violet (Figure 1).

CT113004006_e_Fig1_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20and%20B%2C%20Annular%20verrucous%20plaque%20with%20central%20hypopigmentation%20extending%20from%20the%20medial%20right%20leg%20to%20the%20posterior%20right%20thigh%20with%20keloid%20formation%20at%20the%20proximal%20edge.%20Violaceous%20staining%20was%20seen%20secondary%20to%20topical%20application%20of%20gentian%20violet.%3C%2Fp%3E

Based on the chronic history and morphology, a diagnosis of a chronic fungal or atypical mycobacterial infection was favored. In particular, chromoblastomycosis, cutaneous tuberculosis (eg, scrofuloderma, lupus vulgaris, tuberculosis verrucosa cutis), and atypical mycobacterial infection were highest on the differential, as these conditions often exhibit annular, nodular, verrucous, and/or atrophic lesions. The nodularity, crusting, and draining sinus tracts also raised the possibility of mycetoma. Given the extension of the lesion from the lower to upper leg, a sporotrichoid infection also was considered but was thought to be less likely based on the annular configuration.

Two 4-mm punch biopsies were taken from a peripheral nodule—one for routine histology and another for bacterial, fungal, and mycobacterial cultures. An ­interferon-gamma release assay also was ordered to evaluate for immune responses indicative of prior Mycobacterium tuberculosis infection, but the patient did not obtain this for unknown reasons. Histology demonstrated pseudoepitheliomatous hyperplasia and necrotizing granulomas, which suggested an infectious etiology, but no organisms were identified on tissue staining and all cultures were negative for growth at 6 weeks. The patient was asked to return at that point, and 4 additional scouting biopsies were performed and sent for routine histology, M tuberculosis nucleic acid amplification testing, and microbiologic cultures (ie, bacterial, mycobacterial, fungal, nocardia, actinomycetes). Within 1 week, a filamentous organism with pigmentation visible on the front and back of a Sabouraud dextrose agar plate was identified on fungal culture (Figure 2). Microscopic evaluation of this mold with lactophenol blue stain revealed thin septate hyphae with conidiophores arising at right angles that bore clusters of microconidia (Figure 3). Sequencing analysis ultimately identified this organism as Sporothrix schenckii. Routine histology demonstrated pseudoepitheliomatous hyperplasia with scattered intraepidermal collections of neutrophils (Figure 4). The dermis showed a dense, superficial, and deep infiltrate composed of lymphocytes, histiocytes, and plasma cells with occasional neutrophils and eosinophils. A Grocott-Gomori methenamine-silver stain revealed a cluster of ovoid yeast forms within the stratum corneum (Figure 5). The patient was referred to infectious disease for follow-up and treatment.

Aggarwal_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Anterior%20view%20of%20darkly%20pigmented%2C%20filamentous%20fungi%20grown%20on%20a%20Sabouraud%20dextrose%20agar%20culture%20plate%20at%2025%20%C2%B0C.%20Pigmentation%20was%20visible%20on%20the%20reverse%20view%20of%20the%20culture%20plate%20demonstrating%20that%20the%20mold%20is%20dematiaceous%20(inset).%3C%2Fp%3E

The patient later visited a community clinic providing dermatologic care for patients without insurance. He was started on itraconazole 200 mg daily for a total of 6 months until dermatologic clearance of the cutaneous lesions was observed. He was followed by the clinic with laboratory tests including a liver function test. At follow-up 8 months later, a repeat biopsy was performed to ensure histologic clearance of the sporotrichosis, which revealed a dermal scar and no evidence of residual infection.

Aggarwal_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Lactophenol%20blue%20stain%20of%20the%20mold%20form%20demonstrated%20septate%20hyphae%20with%20conidiophores%20arising%20at%20right%20angles%20and%20clustered%20microconidia%20resembling%20rose%20petals%20(original%20magnification%20%C3%9760).%20Reference%20bar%20indicates%2020%20%C2%B5m.%3C%2Fp%3E

Sporothrix schenckii was first isolated in 1898 by Benjamin Schenck, a student at Johns Hopkins Medicine (Baltimore, Maryland), and identified by a mycologist as sporotricha.1 Species within the genus Sporothrix are unique in that the fungi are both dimorphic (growing as a mold at 25 °C but as a yeast at 37 °C) and dematiaceous (dark pigmentation from melanin is visible on inspection of the anterior and reverse sides of culture plates). Infection usually occurs when cutaneous or subcutaneous tissues are exposed to the fungus via microabrasions; activities thought to contribute to exposure include gardening, agricultural work, animal husbandry, and feline scratches.2 Although skin trauma frequently is considered the primary route of infection, patient recall is variable, with one study noting that only 37.7% of patients recalled trauma and another study similarly demonstrating a patient recall rate of 25%.3,4

Aggarwal_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Histopathology%20demonstrated%20pseudoepitheliomatous%20hyperplasia%20with%20intraepidermal%20collections%20of%20neutrophils%20and%20a%20dense%20mixed%20inflammatory%20dermal%20infiltrate%20(H%26amp%3BE%2C%20original%20magnification%20%C3%974).%3C%2Fp%3E

Lymphocutaneous sporotrichosis is the most common presentation of the fungal infection,5 and clinical cases may be classified into 1 of 4 categories: (1) lymphangitic lesions—papules at the site of inoculation with spread along the lymphatic channels; (2) localized (fixed) cutaneous lesions—1 or 2 lesions at the inoculation site; (3) disseminated (multifocal) cutaneous lesions; and (4) extracutaneous lesions.6 Extracutaneous manifestations of this infection most notably have been reported as pulmonary disease through inhalation of conidia or through dissemination in immunocompromised hosts.7 Our patient’s infection was categorized as lymphangitic lesions due to spread from the lower to upper leg, albeit in a highly atypical, annular fashion. A review of systems was otherwise negative, and CT ruled out osteoarticular involvement.

Aggarwal_5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20Grocott-Gomori%20methenamine-silver%20stain%20demonstrated%20a%20cluster%20of%20ovoid%20yeast%20forms%20within%20the%20stratum%20corneum%20(original%20magnification%20%C3%9760).%20Reference%20bar%20indicates%2020%20%C2%B5m.%3C%2Fp%3E

In addition to socioeconomic barriers, several factors contributed to a delayed diagnosis in this patient including the annular presentation with central hypopigmentation and atrophy, negative initial microbiological cultures and lack of visualization of organisms on histopathology, and the consequent need for repeat biopsies. For lymphocutaneous sporotrichosis, the typical presentation consists of a papule or ulcerated nodule at the site of inoculation with subsequent linear spread along lymphatic channels. This classic sporotrichoid pattern is a key diagnostic clue for identifying sporotrichosis but was absent at the time our patient presented for medical care. Rather, the sporotrichoid spread seemed to have occurred in a centrifugal fashion up the leg. Few case reports have documented an annular presentation of lymphocutaneous sporotrichosis,8-13 and one report described central atrophy and hypopigmentation.10 Pain and pruritus, which were present in our patient, rarely are documented.9 Finally, the diagnosis of cutaneous fungal infections may require multiple biopsies due to the variable abundance of viable organisms in tissue specimens as well as the fastidious growth characteristics of these organisms. Furthermore, sensitivity often is low for both fungal and mycobacterial cultures, and cultures may take days to weeks to yield growth.14,15 For these reasons, empiric therapy and repeat biopsies often are pursued if clinical suspicion is high enough.16 Our patient returned for multiple scouting biopsies after the initial tissue culture was negative and was even considered for empiric treatment against Mycobacterium prior to positive fungal cultures.

Another unique aspect of our case was the presence of a keloid. It is difficult to know if this keloid was secondary to the trauma the patient sustained in the inciting incident or formed from the fungal infection. Interestingly, it has been hypothesized that fungal infections may contribute to keloid and hypertrophic scar formation.17 In a case series of 3 patients with either keloids or hypertrophic scars and concomitant tinea infection, there was notable improvement in the appearance of the scars 2 weeks after beginning itraconazole therapy.17 However, it is not yet known if a fungal infection can contribute to the pathogenesis of keloid formation.

As with other aspects of this case, the length of time the patient went without diagnosis and treatment was unusual and may help explain the atypical presentation. Although the incubation period for S schenckii can vary, most reports identify patients as seeking medical attention within 1 year of rash onset.18-20 In our case, the patient was not diagnosed until 8 years after his symptoms began, requiring multiple referrals, multiple health system touchpoints, and an institution-specific financial aid program. As such, this case also highlights the potential need for a multidisciplinary team approach when caring for patients with poor access to health care.

In conclusion, this case illustrates a unique presentation of lymphocutaneous sporotrichosis that may mimic other chronic infections and result in delayed diagnosis. Although lymphangitic sporotrichosis generally is recognized as having a linear distribution, mounting evidence from this report and others suggests an annular presentation also is possible. Pruritus or pain is rare but should not preclude a diagnosis of sporotrichosis if present. For patients with limited access to health care resources, it is especially important to involve multiple members of the health care team, including social workers and specialists, to prevent a protracted and severe course of disease.

References
  1. Schenck BR. On refractory subcutaneous abscesses caused by a fungus possibly related to the sporotricha. Bulletin of the Johns Hopkins Hospital. 1898;93:286-290.
  2. de Lima Barros MB, de Almeida Paes R, Schubach AO. Sporothrix schenckii and sporotrichosis. Clin Microbiol Rev. 2011;24:633-654. doi:10.1128/CMR.00007-11
  3. Crevasse L, Ellner PD. An outbreak of sporotrichosis in florida. J Am Med Assoc. 1960;173:29-33. doi:10.1001/jama.1960.03020190031006
  4. Mayorga R, Cáceres A, Toriello C, et al. An endemic area of sporotrichosis in Guatemala [in French]. Sabouraudia. 1978;16:185-198.
  5. Morris-Jones R. Sporotrichosis. Clin Exp Dermatol. 2002;27:427-431. doi:10.1046/j.1365-2230.2002.01087.x
  6. Sampaio SA, Da Lacaz CS. Clinical and statistical studies on sporotrichosis in Sao Paulo (Brazil). Article in German. Hautarzt. 1959;10:490-493.
  7. Ramos-e-Silva M, Vasconcelos C, Carneiro S, et al. Sporotrichosis. Clin Dermatol. 2007;25:181-187. doi:10.1016/j.clindermatol.2006.05.006
  8. Williams BA, Jennings TA, Rushing EC, et al. Sporotrichosis on the face of a 7-year-old boy following a bicycle accident. Pediatr Dermatol. 2013;30:E246-E247. doi:10.1111/j.1525-1470.2011.01696.x
  9. Vaishampayan SS, Borde P. An unusual presentation of sporotrichosis. Indian J Dermatol. 2013;58:409. doi:10.4103/0019-5154.117350
  10. Qin J, Zhang J. Sporotrichosis. N Engl J Med. 2019;380:771. doi:10.1056/NEJMicm1809179
  11. Patel A, Mudenda V, Lakhi S, et al. A 27-year-old severely immunosuppressed female with misleading clinical features of disseminated cutaneous sporotrichosis. Case Rep Dermatol Med. 2016;2016:1-4. doi:10.1155/2016/9403690
  12. de Oliveira-Esteves ICMR, Almeida Rosa da Silva G, Eyer-Silva WA, et al. Rapidly progressive disseminated sporotrichosis as the first presentation of HIV infection in a patient with a very low CD4 cell count. Case Rep Infect Dis. 2017;2017:4713140. doi:10.1155/2017/4713140
  13. Singh S, Bachaspatimayum R, Meetei U, et al. Terbinafine in fixed cutaneous sporotrichosis: a case series. J Clin Diagnostic Res. 2018;12:FR01-FR03. doi:10.7860/JCDR/2018/25315.12223
  14. Guarner J, Brandt ME. Histopathologic diagnosis of fungal infections in the 21st century. Clin Microbiol Rev. 2011;24:247-280. doi:10.1128/CMR.00053-10
  15. Peters F, Batinica M, Plum G, et al. Bug or no bug: challenges in diagnosing cutaneous mycobacterial infections. J Ger Soc Dermatol. 2016;14:1227-1236. doi:10.1111/ddg.13001
  16. Khadka P, Koirala S, Thapaliya J. Cutaneous tuberculosis: clinicopathologic arrays and diagnostic challenges. Dermatol Res Pract. 2018;2018:7201973. doi:10.1155/2018/7201973
  17. Okada E, Maruyama Y. Are keloids and hypertrophic scars caused by fungal infection? . Plast Reconstr Surg. 2007;120:814-815. doi:10.1097/01.prs.0000278813.23244.3f
  18. Pappas PG, Tellez I, Deep AE, et al. Sporotrichosis in Peru: description of an area of hyperendemicity. Clin Infect Dis. 2000;30:65-70. doi:10.1086/313607
  19. McGuinness SL, Boyd R, Kidd S, et al. Epidemiological investigation of an outbreak of cutaneous sporotrichosis, Northern Territory, Australia. BMC Infect Dis. 2016;16:1-7. doi:10.1186/s12879-016-1338-0
  20. Rojas FD, Fernández MS, Lucchelli JM, et al. Cavitary pulmonary sporotrichosis: case report and literature review. Mycopathologia. 2017;182:1119-1123. doi:10.1007/s11046-017-0197-6
References
  1. Schenck BR. On refractory subcutaneous abscesses caused by a fungus possibly related to the sporotricha. Bulletin of the Johns Hopkins Hospital. 1898;93:286-290.
  2. de Lima Barros MB, de Almeida Paes R, Schubach AO. Sporothrix schenckii and sporotrichosis. Clin Microbiol Rev. 2011;24:633-654. doi:10.1128/CMR.00007-11
  3. Crevasse L, Ellner PD. An outbreak of sporotrichosis in florida. J Am Med Assoc. 1960;173:29-33. doi:10.1001/jama.1960.03020190031006
  4. Mayorga R, Cáceres A, Toriello C, et al. An endemic area of sporotrichosis in Guatemala [in French]. Sabouraudia. 1978;16:185-198.
  5. Morris-Jones R. Sporotrichosis. Clin Exp Dermatol. 2002;27:427-431. doi:10.1046/j.1365-2230.2002.01087.x
  6. Sampaio SA, Da Lacaz CS. Clinical and statistical studies on sporotrichosis in Sao Paulo (Brazil). Article in German. Hautarzt. 1959;10:490-493.
  7. Ramos-e-Silva M, Vasconcelos C, Carneiro S, et al. Sporotrichosis. Clin Dermatol. 2007;25:181-187. doi:10.1016/j.clindermatol.2006.05.006
  8. Williams BA, Jennings TA, Rushing EC, et al. Sporotrichosis on the face of a 7-year-old boy following a bicycle accident. Pediatr Dermatol. 2013;30:E246-E247. doi:10.1111/j.1525-1470.2011.01696.x
  9. Vaishampayan SS, Borde P. An unusual presentation of sporotrichosis. Indian J Dermatol. 2013;58:409. doi:10.4103/0019-5154.117350
  10. Qin J, Zhang J. Sporotrichosis. N Engl J Med. 2019;380:771. doi:10.1056/NEJMicm1809179
  11. Patel A, Mudenda V, Lakhi S, et al. A 27-year-old severely immunosuppressed female with misleading clinical features of disseminated cutaneous sporotrichosis. Case Rep Dermatol Med. 2016;2016:1-4. doi:10.1155/2016/9403690
  12. de Oliveira-Esteves ICMR, Almeida Rosa da Silva G, Eyer-Silva WA, et al. Rapidly progressive disseminated sporotrichosis as the first presentation of HIV infection in a patient with a very low CD4 cell count. Case Rep Infect Dis. 2017;2017:4713140. doi:10.1155/2017/4713140
  13. Singh S, Bachaspatimayum R, Meetei U, et al. Terbinafine in fixed cutaneous sporotrichosis: a case series. J Clin Diagnostic Res. 2018;12:FR01-FR03. doi:10.7860/JCDR/2018/25315.12223
  14. Guarner J, Brandt ME. Histopathologic diagnosis of fungal infections in the 21st century. Clin Microbiol Rev. 2011;24:247-280. doi:10.1128/CMR.00053-10
  15. Peters F, Batinica M, Plum G, et al. Bug or no bug: challenges in diagnosing cutaneous mycobacterial infections. J Ger Soc Dermatol. 2016;14:1227-1236. doi:10.1111/ddg.13001
  16. Khadka P, Koirala S, Thapaliya J. Cutaneous tuberculosis: clinicopathologic arrays and diagnostic challenges. Dermatol Res Pract. 2018;2018:7201973. doi:10.1155/2018/7201973
  17. Okada E, Maruyama Y. Are keloids and hypertrophic scars caused by fungal infection? . Plast Reconstr Surg. 2007;120:814-815. doi:10.1097/01.prs.0000278813.23244.3f
  18. Pappas PG, Tellez I, Deep AE, et al. Sporotrichosis in Peru: description of an area of hyperendemicity. Clin Infect Dis. 2000;30:65-70. doi:10.1086/313607
  19. McGuinness SL, Boyd R, Kidd S, et al. Epidemiological investigation of an outbreak of cutaneous sporotrichosis, Northern Territory, Australia. BMC Infect Dis. 2016;16:1-7. doi:10.1186/s12879-016-1338-0
  20. Rojas FD, Fernández MS, Lucchelli JM, et al. Cavitary pulmonary sporotrichosis: case report and literature review. Mycopathologia. 2017;182:1119-1123. doi:10.1007/s11046-017-0197-6
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
E6-E9
Page Number
E6-E9
Publications
Publications
Topics
Article Type
Display Headline
Centrifugally Spreading Lymphocutaneous Sporotrichosis: A Rare Cutaneous Manifestation
Display Headline
Centrifugally Spreading Lymphocutaneous Sporotrichosis: A Rare Cutaneous Manifestation
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Aggarwal</fileName> <TBEID>0C02F508.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F508</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Case Letter</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240404T133824</firstPublished> <LastPublished>20240404T133824</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240404T133824</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Aggarwal</byline> <bylineText>Prachi Aggarwal, MD; Andrew Rogers, MD; Liesl Grenier, MD; David C. Gaston, MD, PhD; Sean X. Zhang, MD, PhD; Maunank Shah, MD, PhD; Jihad Alhariri, MD, MPH </bylineText> <bylineFull>Aggarwal</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E6-E9</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>To the Editor:Sporotrichosis refers to a subacute to chronic fungal infection that usually involves the cutaneous and subcutaneous tissues and is caused by the </metaDescription> <articlePDF>300983</articlePDF> <teaserImage/> <title>Centrifugally Spreading Lymphocutaneous Sporotrichosis: A Rare Cutaneous Manifestation</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth/> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>4</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2307</CMSID> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>dermatopathology</keyword> <keyword> infectious disease</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>April 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Case Letter | 2307<pubSubsection/></pubSection> </pubSections> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">44</term> </sections> <topics> <term canonical="true">204</term> <term>234</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026ff.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Centrifugally Spreading Lymphocutaneous Sporotrichosis: A Rare Cutaneous Manifestation</title> <deck/> </itemMeta> <itemContent> <p>To the Editor:<br/><br/>Sporotrichosis refers to a subacute to chronic fungal infection that usually involves the cutaneous and subcutaneous tissues and is caused by the introduction of <i>Sporothrix, </i>a dimorphic fungus, through the skin. We present a case of chronic atypical lymphocutaneous sporotrichosis. </p> <p>A 46-year-old man presented to the outpatient dermatology clinic for follow-up for a rash on the right leg that spread to the thigh and became painful and pruritic. It initially developed 8 years prior to the current presentation after he sustained trauma to the leg from an electroshock weapon. One year prior to the current presentation, he had presented to the emergency department and was prescribed doxycycline 100 mg twice daily for 7 days as well as bacitracin ointment. He also was instructed to follow up with dermatology, but a lack of health insurance and other socioeconomic barriers prevented him from seeking dermatologic care. Nine months later, he again presented to the emergency department due to a motor vehicle accident. Computed tomography (CT) of the right leg revealed exophytic dermal masses, inflammatory stranding of the subcutaneous tissue, and right inguinal lymph nodes measuring up to 1.4 cm; there was no osteoarticular involvement. At that time, the patient was applying gentian violet to the skin lesions and taking hydroxyzine 50 mg 3 times daily as needed for pruritus with minimal relief. Financial support was provided for follow-up with dermatology, which occurred almost 5 months later. <br/><br/>At the current presentation, physical examination revealed a large annular plaque with verrucous, scaly, erythematous borders and a hypopigmented atrophic center extending from the medial aspect of the right leg to the posterior thigh. Numerous pink, scaly, crusted nodules were scattered primarily along the periphery, with some evidence of draining sinus tracts. In addition, a fibrotic pink linear plaque extended from the medial right leg to the popliteal fossa, consistent with a keloid. Violet staining along the periphery of the lesion also was appreciated secondary to the application of topical gentian violet (Figure 1). <br/><br/>Based on the chronic history and morphology, a diagnosis of a chronic fungal or atypical mycobacterial infection was favored. In particular, chromoblastomycosis, cutaneous tuberculosis (eg, scrofuloderma, lupus vulgaris, tuberculosis verrucosa cutis), and atypical mycobacterial infection were highest on the differential, as these conditions often exhibit annular, nodular, verrucous, and/or atrophic lesions. The nodularity, crusting, and draining sinus tracts also raised the possibility of mycetoma. Given the extension of the lesion from the lower to upper leg, a sporotrichoid infection also was considered but was thought to be less likely based on the annular configuration. <br/><br/>Two 4-mm punch biopsies were taken from a peripheral nodule—one for routine histology and another for bacterial, fungal, and mycobacterial cultures. An ­interferon-gamma release assay also was ordered to evaluate for immune responses indicative of prior <i>Mycobacterium tuberculosis</i> infection, but the patient did not obtain this for unknown reasons. Histology demonstrated pseudoepitheliomatous hyperplasia and necrotizing granulomas, which suggested an infectious etiology, but no organisms were identified on tissue staining and all cultures were negative for growth at 6 weeks. The patient was asked to return at that point, and 4 additional scouting biopsies were performed and sent for routine histology, <i>M tuberculosis </i>nucleic acid amplification testing, and microbiologic cultures (ie, bacterial, mycobacterial, fungal, nocardia, actinomycetes). Within 1 week, a filamentous organism with pigmentation visible on the front and back of a Sabouraud dextrose agar plate was identified on fungal culture (Figure 2). Microscopic evaluation of this mold with lactophenol blue stain revealed thin septate hyphae with conidiophores arising at right angles that bore clusters of microconidia (Figure 3). Sequencing analysis ultimately identified this organism as <i>Sporothrix schenckii. </i>Routine histology demonstrated pseudoepitheliomatous hyperplasia with scattered intraepidermal collections of neutrophils (Figure 4). The dermis showed a dense, superficial, and deep infiltrate composed of lymphocytes, histiocytes, and plasma cells with occasional neutrophils and eosinophils. A Grocott-Gomori methenamine-silver stain revealed a cluster of ovoid yeast forms within the stratum corneum (Figure 5). The patient was referred to infectious disease for follow-up and treatment. <br/><br/>The patient later visited a community clinic providing dermatologic care for patients without insurance. He was started on itraconazole 200 mg daily for a total of 6 months until dermatologic clearance of the cutaneous lesions was observed. He was followed by the clinic with laboratory tests including a liver function test. At follow-up 8 months later, a repeat biopsy was performed to ensure histologic clearance of the sporotrichosis, which revealed a dermal scar and no evidence of residual infection. <br/><br/><i>Sporothrix schenckii</i> was first isolated in 1898 by Benjamin Schenck, a student at Johns Hopkins Medicine (Baltimore, Maryland), and identified by a mycologist as sporotricha.<sup>1</sup> Species within the genus <i>Sporothrix</i> are unique in that the fungi are both dimorphic (growing as a mold at 25 <span class="body">°</span>C but as a yeast at 37 <span class="body">°</span>C) and dematiaceous (dark pigmentation from melanin is visible on inspection of the anterior and reverse sides of culture plates). Infection usually occurs when cutaneous or subcutaneous tissues are exposed to the fungus via microabrasions; activities thought to contribute to exposure include gardening, agricultural work, animal husbandry, and feline scratches.<sup>2</sup> Although skin trauma frequently is considered the primary route of infection, patient recall is variable, with one study noting that only 37.7% of patients recalled trauma and another study similarly demonstrating a patient recall rate of 25%.<sup>3,4</sup> <br/><br/>Lymphocutaneous sporotrichosis is the most common presentation of the fungal infection,<sup>5</sup> and clinical cases may be classified into 1 of 4 categories: (1) lymphangitic lesions—papules at the site of inoculation with spread along the lymphatic channels; (2) localized (fixed) cutaneous lesions—1 or 2 lesions at the inoculation site; (3) disseminated (multifocal) cutaneous lesions; and (4) extracutaneous lesions.<sup>6</sup> Extracutaneous manifestations of this infection most notably have been reported as pulmonary disease through inhalation of conidia or through dissemination in immunocompromised hosts.<sup>7</sup> Our patient’s infection was categorized as lymphangitic lesions due to spread from the lower to upper leg, albeit in a highly atypical, annular fashion. A review of systems was otherwise negative, and CT ruled out osteoarticular involvement. <br/><br/>In addition to socioeconomic barriers, several factors contributed to a delayed diagnosis in this patient including the annular presentation with central hypopigmentation and atrophy, negative initial microbiological cultures and lack of visualization of organisms on histopathology, and the consequent need for repeat biopsies. For lymphocutaneous sporotrichosis, the typical presentation consists of a papule or ulcerated nodule at the site of inoculation with subsequent linear spread along lymphatic channels. This classic sporotrichoid pattern is a key diagnostic clue for identifying sporotrichosis but was absent at the time our patient presented for medical care. Rather, the sporotrichoid spread seemed to have occurred in a centrifugal fashion up the leg. Few case reports have documented an annular presentation of lymphocutaneous sporotrichosis,<sup>8-13</sup> and one report described central atrophy and hypopigmentation.<sup>10</sup> Pain and pruritus, which were present in our patient, rarely are documented.<sup>9</sup> Finally, the diagnosis of cutaneous fungal infections may require multiple biopsies due to the variable abundance of viable organisms in tissue specimens as well as the fastidious growth characteristics of these organisms. Furthermore, sensitivity often is low for both fungal and mycobacterial cultures, and cultures may take days to weeks to yield growth.<sup>14,15</sup> For these reasons, empiric therapy and repeat biopsies often are pursued if clinical suspicion is high enough.<sup>16</sup> Our patient returned for multiple scouting biopsies after the initial tissue culture was negative and was even considered for empiric treatment against <i>Mycobacterium</i> prior to positive fungal cultures. <br/><br/>Another unique aspect of our case was the presence of a keloid. It is difficult to know if this keloid was secondary to the trauma the patient sustained in the inciting incident or formed from the fungal infection. Interestingly, it has been hypothesized that fungal infections may contribute to keloid and hypertrophic scar formation.<sup>17</sup> In a case series of 3 patients with either keloids or hypertrophic scars and concomitant tinea infection, there was notable improvement in the appearance of the scars 2 weeks after beginning itraconazole therapy.<sup>17</sup> However, it is not yet known if a fungal infection can contribute to the pathogenesis of keloid formation. <br/><br/>As with other aspects of this case, the length of time the patient went without diagnosis and treatment was unusual and may help explain the atypical presentation. Although the incubation period for <i>S schenckii </i>can vary, most reports identify patients as seeking medical attention within 1 year of rash onset.<sup>18-20</sup> In our case, the patient was not diagnosed until 8 years after his symptoms began, requiring multiple referrals, multiple health system touchpoints, and an institution-specific financial aid program. As such, this case also highlights the potential need for a multidisciplinary team approach when caring for patients with poor access to health care. <br/><br/>In conclusion, this case illustrates a unique presentation of lymphocutaneous sporotrichosis that may mimic other chronic infections and result in delayed diagnosis. Although lymphangitic sporotrichosis generally is recognized as having a linear distribution, mounting evidence from this report and others suggests an annular presentation also is possible. Pruritus or pain is rare but should not preclude a diagnosis of sporotrichosis if present<i>. </i>For patients with limited access to health care resources, it is especially important to involve multiple members of the health care team, including social workers and specialists, to prevent a protracted and severe course of disease. </p> <h2>References </h2> <p class="reference"> 1. Schenck BR. On refractory subcutaneous abscesses caused by a fungus possibly related to the sporotricha. <i>Bulletin of the Johns Hopkins Hospital</i>. 1898;93:286-290.<br/><br/> 2. de Lima Barros MB, de Almeida Paes R, Schubach AO. Sporothrix schenckii and sporotrichosis. <i>Clin Microbiol Rev</i>. 2011;24:633-654. doi:10.1128/CMR.00007-11<br/><br/> 3. Crevasse L, Ellner PD. An outbreak of sporotrichosis in florida. <i>J Am Med Assoc</i>. 1960;173:29-33. doi:10.1001/jama.1960.03020190031006<br/><br/> 4. Mayorga R, Cáceres A, Toriello C, et al. An endemic area of sporotrichosis in Guatemala [in French]. <i>Sabouraudia</i>. 1978;16:185-198. <br/><br/> 5. Morris-Jones R. Sporotrichosis. <i>Clin Exp Dermatol</i>. 2002;27:427-431. doi:10.1046/j.1365-2230.2002.01087.x<br/><br/> 6. Sampaio SA, Da Lacaz CS. Clinical and statistical studies on sporotrichosis in Sao Paulo (Brazil). Article in German. <i>Hautarzt</i>. 1959;10:490-493. <br/><br/> 7. Ramos-e-Silva M, Vasconcelos C, Carneiro S, et al. Sporotrichosis. <i>Clin Dermatol</i>. 2007;25:181-187. doi:10.1016/j.clindermatol.2006.05.006<br/><br/> 8. Williams BA, Jennings TA, Rushing EC, et al. Sporotrichosis on the face of a 7-year-old boy following a bicycle accident. <i>Pediatr Dermatol</i>. 2013;30:E246-E247. doi:10.1111/j.1525-1470.2011.01696.x<br/><br/> 9. Vaishampayan SS, Borde P. An unusual presentation of sporotrichosis. <i>Indian J Dermatol</i>. 2013;58:409. doi:10.4103/0019-5154.117350<br/><br/>10. Qin J, Zhang J. Sporotrichosis. <i>N Engl J Med</i>. 2019;380:771. doi:10.1056/NEJMicm1809179<br/><br/>11. Patel A, Mudenda V, Lakhi S, et al. A 27-year-old severely immunosuppressed female with misleading clinical features of disseminated cutaneous sporotrichosis. <i>Case Rep Dermatol Med</i>. 2016;2016:1-4. doi:10.1155/2016/9403690<br/><br/>12. de Oliveira-Esteves ICMR, Almeida Rosa da Silva G, Eyer-Silva WA, et al. Rapidly progressive disseminated sporotrichosis as the first presentation of HIV infection in a patient with a very low CD4 cell count. <i>Case Rep Infect Dis</i>. 2017;2017:4713140. doi:10.1155/2017/4713140<br/><br/>13. Singh S, Bachaspatimayum R, Meetei U, et al. Terbinafine in fixed cutaneous sporotrichosis: a case series. <i>J Clin Diagnostic Res</i>. 2018;12:FR01-FR03. doi:10.7860/JCDR/2018/25315.12223<br/><br/>14. Guarner J, Brandt ME. Histopathologic diagnosis of fungal infections in the 21st century. <i>Clin Microbiol Rev</i>. 2011;24:247-280. doi:10.1128/CMR.00053-10<br/><br/>15. Peters F, Batinica M, Plum G, et al. Bug or no bug: challenges in diagnosing cutaneous mycobacterial infections. <i>J Ger Soc Dermatol</i>. 2016;14:1227-1236. doi:10.1111/ddg.13001<br/><br/>16. Khadka P, Koirala S, Thapaliya J. Cutaneous tuberculosis: clinicopathologic arrays and diagnostic challenges. <i>Dermatol Res Pract</i>. 2018;2018:7201973. doi:10.1155/2018/7201973<br/><br/>17. Okada E, Maruyama Y. Are keloids and hypertrophic scars caused by fungal infection? . <i>Plast Reconstr Surg</i>. 2007;120:814-815. doi:10.1097/01.prs.0000278813.23244.3f<br/><br/>18. Pappas PG, Tellez I, Deep AE, et al. Sporotrichosis in Peru: description of an area of hyperendemicity. <i>Clin Infect Dis</i>. 2000;30:65-70. doi:10.1086/313607<br/><br/>19. McGuinness SL, Boyd R, Kidd S, et al. Epidemiological investigation of an outbreak of cutaneous sporotrichosis, Northern Territory, Australia. <i>BMC Infect Dis</i>. 2016;16:1-7. doi:10.1186/s12879-016-1338-0<br/><br/>20. Rojas FD, Fernández MS, Lucchelli JM, et al. Cavitary pulmonary sporotrichosis: case report and literature review. <i>Mycopathologia</i>. 2017;182:1119-1123. doi:10.1007/s11046-017-0197-6</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From The Johns Hopkins University School of Medicine, Baltimore, Maryland. Prachi Aggarwal as well as Drs. Rogers, Grenier, and Alhariri are from the Department of Dermatology; Drs. Gaston and Zhang are from the Department of Pathology; and Dr. Shah is from the Division of Infectious Disease.</p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>Correspondence: Jihad Alhariri, MD, MPH, Cancer Research Building II, Johns Hopkins University School of Medicine, 1550 Orleans St, Baltimore, MD 21231 (jalhari1@jhmi.edu).<br/><br/><em>Cutis.</em> 2024 April;113(4):E6-E9. doi:10.12788/cutis.0996</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>An atypical presentation of lymphocutaneous sporotrichosis may pose challenges to timely diagnosis and treatment. </li> <li>Although lymphocutaneous sporotrichosis spreads most commonly in a linear fashion along lymphatic channels, an annular configuration is possible. </li> <li>Initial tissue cultures and histopathology of lymphocutaneous sporotrichosis may not yield a diagnosis, necessitating repeat biopsies when clinical suspicion is high. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • An atypical presentation of lymphocutaneous sporotrichosis may pose challenges to timely diagnosis and treatment.
  • Although lymphocutaneous sporotrichosis spreads most commonly in a linear fashion along lymphatic channels, an annular configuration is possible.
  • Initial tissue cultures and histopathology of lymphocutaneous sporotrichosis may not yield a diagnosis, necessitating repeat biopsies when clinical suspicion is high.
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
Image
Teambase ID
180026FF.SIG
Disable zoom
Off

The Role of Dermatology in Identifying and Reporting a Primary Varicella Outbreak

Article Type
Changed
Thu, 04/04/2024 - 14:33
Display Headline
The Role of Dermatology in Identifying and Reporting a Primary Varicella Outbreak

To the Editor:

Cases of primary varicella-zoster virus (VZV) are relatively uncommon in the United States since the introduction of the varicella vaccine in 1995, with an overall decline in cases of more than 97%.1 Prior to the vaccine, 70% of hospitalizations occurred in children; subsequently, hospitalizations among the pediatric population (aged ≤20 years) declined by 97%. Compared to children, adults and immunocompromised patients with VZV infection may present with more severe disease and experience more complications.1

Most children in the United States are vaccinated against VZV, with 90.3% receiving at least 1 dose by 24 months of age.2 However, many countries do not implement universal varicella vaccination for infants.3 As a result, physicians should remember to include primary varicella in the differential when clinically correlated, especially when evaluating patients who have immigrated to the United States or who may be living in unvaccinated communities. We report 2 cases of primary VZV manifesting in adults to remind readers of the salient clinical features of this disease and how dermatologists play a critical role in early and accurate identification of diseases that can have wide-reaching public health implications.

A 26-year-old man with no relevant medical history presented to the emergency department with an itchy and painful rash of 5 days’ duration that began on the trunk and spread to the face, lips, feet, hands, arms, and legs. He also reported shortness of breath, cough, and chills, and he had a temperature of 100.8 °F (38.2 °C). Physical examination revealed numerous erythematous papules and vesiculopustules, some with central umbilication and some with overlying gold crusts (Figure 1).

Richards_Varicella_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%2026-year-old%20man%20with%20erythematous%20papules%20and%20vesicopustules%20scattered%20diffusely%20on%20the%20chest%20characteristic%20of%20varicella-zoster%20virus.%3C%2Fp%3E

Later that day, a 47-year-old man with no relevant medical history presented to the same emergency department with a rash along with self-reported fever and sore throat of 3 days’ duration. Physical examination found innumerable erythematous vesicopustules scattered on the face, scalp, neck, trunk, arms, and legs, some with a “dew drop on a rose petal” appearance and some with overlying hemorrhagic crust (Figure 2).

Richards_Varicella_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%2047-year-old%20man%20with%20erythematous%20vesicopustules%20in%20variable%20stages%20of%20healing%20widely%20dispersed%20across%20the%20upper%20back%20characteristic%20of%20varicella-zoster%20virus.%3C%2Fp%3E

Although infection was of primary concern for the first patient, the presentation of the second patient prompted specific concern for primary VZV infection in both patients, who were placed on airborne and contact isolation precautions.

Skin biopsies from both patients showed acantholytic blisters, hair follicle necrosis, and marked dermal inflammation (Figure 3). Herpetic viral changes were seen in keratinocytes, with steel-grey nuclei, multinucleated keratinocytes, and chromatin margination. An immunostain for VZV was diffusely positive, and VZV antibody IgG was positive (Figure 4).

CT113003016_e_Fig3_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20A%2C%20Histopathology%20showed%20an%20intraepidermal%20acantholytic%20blister%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%20B%2C%20High-power%20view%20revealed%20the%20classic%20herpetic%20viral%20cytopathic%20effect%20of%20multinucleation%2C%20chromatin%20margination%2C%20and%20nuclear%20molding%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97200).%3C%2Fp%3E

Upon additional questioning, both patients reported recent exposure to VZV-like illnesses in family members without a history of international travel. Neither of the patients was sure of their vaccination status or prior infection history. Both patients received intravenous acyclovir 10 mg/kg administered every 8 hours. Both patients experienced improvement and were discharged after 3 days on oral valacyclovir (1 g 3 times daily for a 7-day treatment course).

Richards_Varicella_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Immunostain%20for%20varicella-zoster%20virus%20was%20positive%20(original%20magnification%20%C3%97100).%3C%2Fp%3E

 

 

The similar presentation and timing of these 2 VZV cases caused concern for an unidentified community outbreak. The infection control team was notified; additionally, per hospital protocol the state health department was alerted as well as the clinicians and staff of the hospital with a request to be vigilant for further cases.

Despite high vaccination rates in the United States, outbreaks of varicella still occur, particularly among unvaccinated individuals, and a robust and efficient response is necessary to control the spread of such outbreaks.4 Many states, including Arkansas where our cases occurred, have laws mandating report of VZV cases to the department of health.5 Dermatologists play an important role in reporting cases, aiding in diagnosis through recognition of the physical examination findings, obtaining appropriate biopsy, and recommending additional laboratory testing.

Typical skin manifestations include a pruritic rash of macules, papules, vesicles, and crusted lesions distributed throughout the trunk, face, arms, and legs. Because new lesions appear over several days, they will be in different stages of healing, resulting in the simultaneous presence of papules, vesicles, and crusted lesions.6 This unique characteristic helps distinguish VZV from other skin diseases such as smallpox or mpox (monkeypox), which generally show lesions in similar stages of evolution.

Biopsy also can aid in identification. Viruses in the herpes family reveal similar histopathologic characteristics, including acantholysis and vesicle formation, intranuclear inclusions with margination of chromatin, multinucleation, and nuclear molding.7 Immunohistochemistry can be used to differentiate VZV from herpes simplex virus; however, neither microscopic examination nor immunohistochemistry distinguish primary VZV infection from herpes zoster (HZ).8

The mpox rash progresses more slowly than a VZV rash and has a centrifugal rather than central distribution that can involve the palms and soles. Lymphadenopathy is a characteristic finding in mpox.9 Rickettsialpox is distinguished from VZV primarily by the appearance of brown or black eschar after the original papulovesicular lesions dry out.10 Atypical hand, foot, and mouth disease can manifest in adults as widespread papulovesicular lesions. This form is associated with coxsackievirus A6 and may require direct fluorescent antibody assay or polymerase chain reaction of keratinocytes to rule out VZV.11

Herpes zoster occurs in older adults with a history of primary VZV.6 It manifests as vesicular lesions confined to 1 or 2 adjacent dermatomes vs the diffuse spread of VZV over the entire body. However, HZ can become disseminated in immunocompromised individuals, making it difficult to clinically distinguish from VZV.6 Serology can be helpful, as high IgM titers indicate an acute primary VZV infection. Subsequently increased IgG titers steadily wane over time and spike during reactivation.12

Dermatology and infectious disease consultations in our cases yielded a preliminary diagnosis through physical examination that was confirmed by biopsy and subsequent laboratory testing, which allowed for a swift response by the infection control team including isolation precautions to control a potential outbreak. Patients with VZV should remain in respiratory isolation until all lesions have crusted over.6

 

 

Individuals who had face-to-face indoor contact for at least 5 minutes or who shared a living space with an infected individual should be assessed for VZV immunity, which is defined as confirmed prior immunization or infection.5,13 Lack of VZV immunity requires postexposure prophylaxis—active immunization for the immunocompetent and passive immunization for the immunocompromised.13 Ultimately, no additional cases were reported in the community where our patients resided.

Immunocompetent children with primary VZV require supportive care only. Oral antiviral therapy is the treatment of choice for immunocompetent adults or anyone at increased risk for complications, while intravenous antivirals are recommended for the immunocompromised or those with VZV-related complications.14 A similar approach is used for HZ. Uncomplicated cases are treated with oral antivirals, and complicated cases (eg, HZ ophthalmicus) are treated with intravenous antivirals.15 Commonly used antivirals include acyclovir, valacyclovir, and famciclovir.14

Our cases highlight the ongoing risk for varicella outbreaks in unvaccinated or undervaccinated communities. Physician vigilance is necessary, and dermatology plays a particularly important role in swift and accurate detection of VZV, as demonstrated in our cases by the recognition of classic physical examination findings of erythematous and vesicular papules in each of the patients. Because primary VZV infection can result in life-threatening complications including hepatitis, encephalitis, and pancreatitis, prompt identification and initiation of therapy is important.6 Similarly, quick notification of public health officials about detected primary VZV cases is vital to containing potential community outbreaks.

References
  1. Centers for Disease Control and Prevention. Chickenpox (varicella) for healthcare professionals. Published October 21, 2022. Accessed March 6, 2024. https://www.cdc.gov/chickenpox/hcp/index.html#vaccination-impact
  2. National Center for Health Statistics. Immunization. Published June 13, 2023. Accessed March 6, 2024. https://www.cdc.gov/nchs/fastats/immunize.htm
  3. Lee YH, Choe YJ, Lee J, et al. Global varicella vaccination programs. Clin Exp Pediatr. 2022;65:555. doi:10.3345/CEP.2021.01564
  4. Leung J, Lopez AS, Marin M. Changing epidemiology of varicella outbreaks in the United States during the Varicella Vaccination Program, 1995–2019. J Infect Dis. 2022;226(suppl 4):S400-S406.
  5. Arkansas Department of Health. Rules Pertaining to Reportable Diseases. Published September 11, 2023. Accessed March 6, 2024. https://www.healthy.arkansas.gov/images/uploads/rules/ReportableDiseaseList.pdf
  6. Pergam S, Limaye A; The AST Infectious Diseases Community of Practice. Varicella zoster virus (VZV). Am J Transplant. 2009;9(suppl 4):S108-S115. doi:10.1111/J.1600-9143.2009.02901.X
  7. Hoyt B, Bhawan J. Histological spectrum of cutaneous herpes infections. Am J Dermatopathol. 2014;36:609-619. doi:10.1097/DAD.0000000000000148
  8. Oumarou Hama H, Aboudharam G, Barbieri R, et al. Immunohistochemical diagnosis of human infectious diseases: a review. Diagn Pathol. 2022;17. doi:10.1186/S13000-022-01197-5
  9. World Health Organization. Mpox (monkeypox). Published April 18, 2023. Accessed March 7, 2024. https://www.who.int/news-room/fact-sheets/detail/monkeypox
  10. Akram SM, Jamil RT, Gossman W. Rickettsia akari (Rickettsialpox). StatPearls [Internet]. Updated May 8, 2023. Accessed February 29, 2024. https://www.ncbi.nlm.nih.gov/books/NBK448081/
  11. Lott JP, Liu K, Landry ML, et al. Atypical hand-foot-mouth disease associated with coxsackievirus A6 infection. J Am Acad Dermatol. 2013;69:736. doi:10.1016/J.JAAD.2013.07.024
  12. Petrun B, Williams V, Brice S. Disseminated varicella-zoster virus in an immunocompetent adult. Dermatol Online J. 2015;21. doi:10.5070/D3213022343
  13. Kimberlin D, Barnett E, Lynfield R, et al. Exposure to specific pathogens. In: Red Book: 2021-2024 Report of the Committee of Infectious Disease. 32nd ed. American Academy of Pediatrics; 2021:1007-1009.
  14. Treatment of varicella (chickenpox) infection. UpToDate [Internet]. Updated February 7, 2024. Accessed March 6, 2024. https://www.uptodate.com/contents/treatment-of-varicella-chickenpox-infection
  15. Treatment of herpes zoster in the immunocompetent host. UpToDate [Internet]. Updated November 29, 2023. Accessed March 6, 2024. https://www.uptodate.com/contents/treatment-of-herpes-zoster
Article PDF
Author and Disclosure Information

From the University of Arkansas for Medical Sciences. Madeline P. Richards is from the College of Medicine, North Little Rock. Dr. Shalin is from the Departments of Dermatology and Pathology, Dr. Guram is from the Department of Dermatology, and Dr. Vyas is from the Department of Internal Medicine, Little Rock.

Madeline P. Richards and Drs. Shalin and Guram report no conflict of interest. Dr. Vyas has served as a paid consultant to the American Association of Hip and Knee Surgeons.

Correspondence: Madeline P. Richards, BS, 324 UAMS Campus Dr, Mail Slot #576, Little Rock, AR 72205 (mprichards@uams.edu).

Issue
Cutis - 113(3)
Publications
Topics
Page Number
E16-E19
Sections
Author and Disclosure Information

From the University of Arkansas for Medical Sciences. Madeline P. Richards is from the College of Medicine, North Little Rock. Dr. Shalin is from the Departments of Dermatology and Pathology, Dr. Guram is from the Department of Dermatology, and Dr. Vyas is from the Department of Internal Medicine, Little Rock.

Madeline P. Richards and Drs. Shalin and Guram report no conflict of interest. Dr. Vyas has served as a paid consultant to the American Association of Hip and Knee Surgeons.

Correspondence: Madeline P. Richards, BS, 324 UAMS Campus Dr, Mail Slot #576, Little Rock, AR 72205 (mprichards@uams.edu).

Author and Disclosure Information

From the University of Arkansas for Medical Sciences. Madeline P. Richards is from the College of Medicine, North Little Rock. Dr. Shalin is from the Departments of Dermatology and Pathology, Dr. Guram is from the Department of Dermatology, and Dr. Vyas is from the Department of Internal Medicine, Little Rock.

Madeline P. Richards and Drs. Shalin and Guram report no conflict of interest. Dr. Vyas has served as a paid consultant to the American Association of Hip and Knee Surgeons.

Correspondence: Madeline P. Richards, BS, 324 UAMS Campus Dr, Mail Slot #576, Little Rock, AR 72205 (mprichards@uams.edu).

Article PDF
Article PDF

To the Editor:

Cases of primary varicella-zoster virus (VZV) are relatively uncommon in the United States since the introduction of the varicella vaccine in 1995, with an overall decline in cases of more than 97%.1 Prior to the vaccine, 70% of hospitalizations occurred in children; subsequently, hospitalizations among the pediatric population (aged ≤20 years) declined by 97%. Compared to children, adults and immunocompromised patients with VZV infection may present with more severe disease and experience more complications.1

Most children in the United States are vaccinated against VZV, with 90.3% receiving at least 1 dose by 24 months of age.2 However, many countries do not implement universal varicella vaccination for infants.3 As a result, physicians should remember to include primary varicella in the differential when clinically correlated, especially when evaluating patients who have immigrated to the United States or who may be living in unvaccinated communities. We report 2 cases of primary VZV manifesting in adults to remind readers of the salient clinical features of this disease and how dermatologists play a critical role in early and accurate identification of diseases that can have wide-reaching public health implications.

A 26-year-old man with no relevant medical history presented to the emergency department with an itchy and painful rash of 5 days’ duration that began on the trunk and spread to the face, lips, feet, hands, arms, and legs. He also reported shortness of breath, cough, and chills, and he had a temperature of 100.8 °F (38.2 °C). Physical examination revealed numerous erythematous papules and vesiculopustules, some with central umbilication and some with overlying gold crusts (Figure 1).

Richards_Varicella_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%2026-year-old%20man%20with%20erythematous%20papules%20and%20vesicopustules%20scattered%20diffusely%20on%20the%20chest%20characteristic%20of%20varicella-zoster%20virus.%3C%2Fp%3E

Later that day, a 47-year-old man with no relevant medical history presented to the same emergency department with a rash along with self-reported fever and sore throat of 3 days’ duration. Physical examination found innumerable erythematous vesicopustules scattered on the face, scalp, neck, trunk, arms, and legs, some with a “dew drop on a rose petal” appearance and some with overlying hemorrhagic crust (Figure 2).

Richards_Varicella_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%2047-year-old%20man%20with%20erythematous%20vesicopustules%20in%20variable%20stages%20of%20healing%20widely%20dispersed%20across%20the%20upper%20back%20characteristic%20of%20varicella-zoster%20virus.%3C%2Fp%3E

Although infection was of primary concern for the first patient, the presentation of the second patient prompted specific concern for primary VZV infection in both patients, who were placed on airborne and contact isolation precautions.

Skin biopsies from both patients showed acantholytic blisters, hair follicle necrosis, and marked dermal inflammation (Figure 3). Herpetic viral changes were seen in keratinocytes, with steel-grey nuclei, multinucleated keratinocytes, and chromatin margination. An immunostain for VZV was diffusely positive, and VZV antibody IgG was positive (Figure 4).

CT113003016_e_Fig3_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20A%2C%20Histopathology%20showed%20an%20intraepidermal%20acantholytic%20blister%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%20B%2C%20High-power%20view%20revealed%20the%20classic%20herpetic%20viral%20cytopathic%20effect%20of%20multinucleation%2C%20chromatin%20margination%2C%20and%20nuclear%20molding%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97200).%3C%2Fp%3E

Upon additional questioning, both patients reported recent exposure to VZV-like illnesses in family members without a history of international travel. Neither of the patients was sure of their vaccination status or prior infection history. Both patients received intravenous acyclovir 10 mg/kg administered every 8 hours. Both patients experienced improvement and were discharged after 3 days on oral valacyclovir (1 g 3 times daily for a 7-day treatment course).

Richards_Varicella_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Immunostain%20for%20varicella-zoster%20virus%20was%20positive%20(original%20magnification%20%C3%97100).%3C%2Fp%3E

 

 

The similar presentation and timing of these 2 VZV cases caused concern for an unidentified community outbreak. The infection control team was notified; additionally, per hospital protocol the state health department was alerted as well as the clinicians and staff of the hospital with a request to be vigilant for further cases.

Despite high vaccination rates in the United States, outbreaks of varicella still occur, particularly among unvaccinated individuals, and a robust and efficient response is necessary to control the spread of such outbreaks.4 Many states, including Arkansas where our cases occurred, have laws mandating report of VZV cases to the department of health.5 Dermatologists play an important role in reporting cases, aiding in diagnosis through recognition of the physical examination findings, obtaining appropriate biopsy, and recommending additional laboratory testing.

Typical skin manifestations include a pruritic rash of macules, papules, vesicles, and crusted lesions distributed throughout the trunk, face, arms, and legs. Because new lesions appear over several days, they will be in different stages of healing, resulting in the simultaneous presence of papules, vesicles, and crusted lesions.6 This unique characteristic helps distinguish VZV from other skin diseases such as smallpox or mpox (monkeypox), which generally show lesions in similar stages of evolution.

Biopsy also can aid in identification. Viruses in the herpes family reveal similar histopathologic characteristics, including acantholysis and vesicle formation, intranuclear inclusions with margination of chromatin, multinucleation, and nuclear molding.7 Immunohistochemistry can be used to differentiate VZV from herpes simplex virus; however, neither microscopic examination nor immunohistochemistry distinguish primary VZV infection from herpes zoster (HZ).8

The mpox rash progresses more slowly than a VZV rash and has a centrifugal rather than central distribution that can involve the palms and soles. Lymphadenopathy is a characteristic finding in mpox.9 Rickettsialpox is distinguished from VZV primarily by the appearance of brown or black eschar after the original papulovesicular lesions dry out.10 Atypical hand, foot, and mouth disease can manifest in adults as widespread papulovesicular lesions. This form is associated with coxsackievirus A6 and may require direct fluorescent antibody assay or polymerase chain reaction of keratinocytes to rule out VZV.11

Herpes zoster occurs in older adults with a history of primary VZV.6 It manifests as vesicular lesions confined to 1 or 2 adjacent dermatomes vs the diffuse spread of VZV over the entire body. However, HZ can become disseminated in immunocompromised individuals, making it difficult to clinically distinguish from VZV.6 Serology can be helpful, as high IgM titers indicate an acute primary VZV infection. Subsequently increased IgG titers steadily wane over time and spike during reactivation.12

Dermatology and infectious disease consultations in our cases yielded a preliminary diagnosis through physical examination that was confirmed by biopsy and subsequent laboratory testing, which allowed for a swift response by the infection control team including isolation precautions to control a potential outbreak. Patients with VZV should remain in respiratory isolation until all lesions have crusted over.6

 

 

Individuals who had face-to-face indoor contact for at least 5 minutes or who shared a living space with an infected individual should be assessed for VZV immunity, which is defined as confirmed prior immunization or infection.5,13 Lack of VZV immunity requires postexposure prophylaxis—active immunization for the immunocompetent and passive immunization for the immunocompromised.13 Ultimately, no additional cases were reported in the community where our patients resided.

Immunocompetent children with primary VZV require supportive care only. Oral antiviral therapy is the treatment of choice for immunocompetent adults or anyone at increased risk for complications, while intravenous antivirals are recommended for the immunocompromised or those with VZV-related complications.14 A similar approach is used for HZ. Uncomplicated cases are treated with oral antivirals, and complicated cases (eg, HZ ophthalmicus) are treated with intravenous antivirals.15 Commonly used antivirals include acyclovir, valacyclovir, and famciclovir.14

Our cases highlight the ongoing risk for varicella outbreaks in unvaccinated or undervaccinated communities. Physician vigilance is necessary, and dermatology plays a particularly important role in swift and accurate detection of VZV, as demonstrated in our cases by the recognition of classic physical examination findings of erythematous and vesicular papules in each of the patients. Because primary VZV infection can result in life-threatening complications including hepatitis, encephalitis, and pancreatitis, prompt identification and initiation of therapy is important.6 Similarly, quick notification of public health officials about detected primary VZV cases is vital to containing potential community outbreaks.

To the Editor:

Cases of primary varicella-zoster virus (VZV) are relatively uncommon in the United States since the introduction of the varicella vaccine in 1995, with an overall decline in cases of more than 97%.1 Prior to the vaccine, 70% of hospitalizations occurred in children; subsequently, hospitalizations among the pediatric population (aged ≤20 years) declined by 97%. Compared to children, adults and immunocompromised patients with VZV infection may present with more severe disease and experience more complications.1

Most children in the United States are vaccinated against VZV, with 90.3% receiving at least 1 dose by 24 months of age.2 However, many countries do not implement universal varicella vaccination for infants.3 As a result, physicians should remember to include primary varicella in the differential when clinically correlated, especially when evaluating patients who have immigrated to the United States or who may be living in unvaccinated communities. We report 2 cases of primary VZV manifesting in adults to remind readers of the salient clinical features of this disease and how dermatologists play a critical role in early and accurate identification of diseases that can have wide-reaching public health implications.

A 26-year-old man with no relevant medical history presented to the emergency department with an itchy and painful rash of 5 days’ duration that began on the trunk and spread to the face, lips, feet, hands, arms, and legs. He also reported shortness of breath, cough, and chills, and he had a temperature of 100.8 °F (38.2 °C). Physical examination revealed numerous erythematous papules and vesiculopustules, some with central umbilication and some with overlying gold crusts (Figure 1).

Richards_Varicella_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%2026-year-old%20man%20with%20erythematous%20papules%20and%20vesicopustules%20scattered%20diffusely%20on%20the%20chest%20characteristic%20of%20varicella-zoster%20virus.%3C%2Fp%3E

Later that day, a 47-year-old man with no relevant medical history presented to the same emergency department with a rash along with self-reported fever and sore throat of 3 days’ duration. Physical examination found innumerable erythematous vesicopustules scattered on the face, scalp, neck, trunk, arms, and legs, some with a “dew drop on a rose petal” appearance and some with overlying hemorrhagic crust (Figure 2).

Richards_Varicella_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%2047-year-old%20man%20with%20erythematous%20vesicopustules%20in%20variable%20stages%20of%20healing%20widely%20dispersed%20across%20the%20upper%20back%20characteristic%20of%20varicella-zoster%20virus.%3C%2Fp%3E

Although infection was of primary concern for the first patient, the presentation of the second patient prompted specific concern for primary VZV infection in both patients, who were placed on airborne and contact isolation precautions.

Skin biopsies from both patients showed acantholytic blisters, hair follicle necrosis, and marked dermal inflammation (Figure 3). Herpetic viral changes were seen in keratinocytes, with steel-grey nuclei, multinucleated keratinocytes, and chromatin margination. An immunostain for VZV was diffusely positive, and VZV antibody IgG was positive (Figure 4).

CT113003016_e_Fig3_AB.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20A%2C%20Histopathology%20showed%20an%20intraepidermal%20acantholytic%20blister%20(H%26amp%3BE%2C%20original%20magnification%20%C3%9740).%20B%2C%20High-power%20view%20revealed%20the%20classic%20herpetic%20viral%20cytopathic%20effect%20of%20multinucleation%2C%20chromatin%20margination%2C%20and%20nuclear%20molding%20(H%26amp%3BE%2C%20original%20magnification%20%C3%97200).%3C%2Fp%3E

Upon additional questioning, both patients reported recent exposure to VZV-like illnesses in family members without a history of international travel. Neither of the patients was sure of their vaccination status or prior infection history. Both patients received intravenous acyclovir 10 mg/kg administered every 8 hours. Both patients experienced improvement and were discharged after 3 days on oral valacyclovir (1 g 3 times daily for a 7-day treatment course).

Richards_Varicella_4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Immunostain%20for%20varicella-zoster%20virus%20was%20positive%20(original%20magnification%20%C3%97100).%3C%2Fp%3E

 

 

The similar presentation and timing of these 2 VZV cases caused concern for an unidentified community outbreak. The infection control team was notified; additionally, per hospital protocol the state health department was alerted as well as the clinicians and staff of the hospital with a request to be vigilant for further cases.

Despite high vaccination rates in the United States, outbreaks of varicella still occur, particularly among unvaccinated individuals, and a robust and efficient response is necessary to control the spread of such outbreaks.4 Many states, including Arkansas where our cases occurred, have laws mandating report of VZV cases to the department of health.5 Dermatologists play an important role in reporting cases, aiding in diagnosis through recognition of the physical examination findings, obtaining appropriate biopsy, and recommending additional laboratory testing.

Typical skin manifestations include a pruritic rash of macules, papules, vesicles, and crusted lesions distributed throughout the trunk, face, arms, and legs. Because new lesions appear over several days, they will be in different stages of healing, resulting in the simultaneous presence of papules, vesicles, and crusted lesions.6 This unique characteristic helps distinguish VZV from other skin diseases such as smallpox or mpox (monkeypox), which generally show lesions in similar stages of evolution.

Biopsy also can aid in identification. Viruses in the herpes family reveal similar histopathologic characteristics, including acantholysis and vesicle formation, intranuclear inclusions with margination of chromatin, multinucleation, and nuclear molding.7 Immunohistochemistry can be used to differentiate VZV from herpes simplex virus; however, neither microscopic examination nor immunohistochemistry distinguish primary VZV infection from herpes zoster (HZ).8

The mpox rash progresses more slowly than a VZV rash and has a centrifugal rather than central distribution that can involve the palms and soles. Lymphadenopathy is a characteristic finding in mpox.9 Rickettsialpox is distinguished from VZV primarily by the appearance of brown or black eschar after the original papulovesicular lesions dry out.10 Atypical hand, foot, and mouth disease can manifest in adults as widespread papulovesicular lesions. This form is associated with coxsackievirus A6 and may require direct fluorescent antibody assay or polymerase chain reaction of keratinocytes to rule out VZV.11

Herpes zoster occurs in older adults with a history of primary VZV.6 It manifests as vesicular lesions confined to 1 or 2 adjacent dermatomes vs the diffuse spread of VZV over the entire body. However, HZ can become disseminated in immunocompromised individuals, making it difficult to clinically distinguish from VZV.6 Serology can be helpful, as high IgM titers indicate an acute primary VZV infection. Subsequently increased IgG titers steadily wane over time and spike during reactivation.12

Dermatology and infectious disease consultations in our cases yielded a preliminary diagnosis through physical examination that was confirmed by biopsy and subsequent laboratory testing, which allowed for a swift response by the infection control team including isolation precautions to control a potential outbreak. Patients with VZV should remain in respiratory isolation until all lesions have crusted over.6

 

 

Individuals who had face-to-face indoor contact for at least 5 minutes or who shared a living space with an infected individual should be assessed for VZV immunity, which is defined as confirmed prior immunization or infection.5,13 Lack of VZV immunity requires postexposure prophylaxis—active immunization for the immunocompetent and passive immunization for the immunocompromised.13 Ultimately, no additional cases were reported in the community where our patients resided.

Immunocompetent children with primary VZV require supportive care only. Oral antiviral therapy is the treatment of choice for immunocompetent adults or anyone at increased risk for complications, while intravenous antivirals are recommended for the immunocompromised or those with VZV-related complications.14 A similar approach is used for HZ. Uncomplicated cases are treated with oral antivirals, and complicated cases (eg, HZ ophthalmicus) are treated with intravenous antivirals.15 Commonly used antivirals include acyclovir, valacyclovir, and famciclovir.14

Our cases highlight the ongoing risk for varicella outbreaks in unvaccinated or undervaccinated communities. Physician vigilance is necessary, and dermatology plays a particularly important role in swift and accurate detection of VZV, as demonstrated in our cases by the recognition of classic physical examination findings of erythematous and vesicular papules in each of the patients. Because primary VZV infection can result in life-threatening complications including hepatitis, encephalitis, and pancreatitis, prompt identification and initiation of therapy is important.6 Similarly, quick notification of public health officials about detected primary VZV cases is vital to containing potential community outbreaks.

References
  1. Centers for Disease Control and Prevention. Chickenpox (varicella) for healthcare professionals. Published October 21, 2022. Accessed March 6, 2024. https://www.cdc.gov/chickenpox/hcp/index.html#vaccination-impact
  2. National Center for Health Statistics. Immunization. Published June 13, 2023. Accessed March 6, 2024. https://www.cdc.gov/nchs/fastats/immunize.htm
  3. Lee YH, Choe YJ, Lee J, et al. Global varicella vaccination programs. Clin Exp Pediatr. 2022;65:555. doi:10.3345/CEP.2021.01564
  4. Leung J, Lopez AS, Marin M. Changing epidemiology of varicella outbreaks in the United States during the Varicella Vaccination Program, 1995–2019. J Infect Dis. 2022;226(suppl 4):S400-S406.
  5. Arkansas Department of Health. Rules Pertaining to Reportable Diseases. Published September 11, 2023. Accessed March 6, 2024. https://www.healthy.arkansas.gov/images/uploads/rules/ReportableDiseaseList.pdf
  6. Pergam S, Limaye A; The AST Infectious Diseases Community of Practice. Varicella zoster virus (VZV). Am J Transplant. 2009;9(suppl 4):S108-S115. doi:10.1111/J.1600-9143.2009.02901.X
  7. Hoyt B, Bhawan J. Histological spectrum of cutaneous herpes infections. Am J Dermatopathol. 2014;36:609-619. doi:10.1097/DAD.0000000000000148
  8. Oumarou Hama H, Aboudharam G, Barbieri R, et al. Immunohistochemical diagnosis of human infectious diseases: a review. Diagn Pathol. 2022;17. doi:10.1186/S13000-022-01197-5
  9. World Health Organization. Mpox (monkeypox). Published April 18, 2023. Accessed March 7, 2024. https://www.who.int/news-room/fact-sheets/detail/monkeypox
  10. Akram SM, Jamil RT, Gossman W. Rickettsia akari (Rickettsialpox). StatPearls [Internet]. Updated May 8, 2023. Accessed February 29, 2024. https://www.ncbi.nlm.nih.gov/books/NBK448081/
  11. Lott JP, Liu K, Landry ML, et al. Atypical hand-foot-mouth disease associated with coxsackievirus A6 infection. J Am Acad Dermatol. 2013;69:736. doi:10.1016/J.JAAD.2013.07.024
  12. Petrun B, Williams V, Brice S. Disseminated varicella-zoster virus in an immunocompetent adult. Dermatol Online J. 2015;21. doi:10.5070/D3213022343
  13. Kimberlin D, Barnett E, Lynfield R, et al. Exposure to specific pathogens. In: Red Book: 2021-2024 Report of the Committee of Infectious Disease. 32nd ed. American Academy of Pediatrics; 2021:1007-1009.
  14. Treatment of varicella (chickenpox) infection. UpToDate [Internet]. Updated February 7, 2024. Accessed March 6, 2024. https://www.uptodate.com/contents/treatment-of-varicella-chickenpox-infection
  15. Treatment of herpes zoster in the immunocompetent host. UpToDate [Internet]. Updated November 29, 2023. Accessed March 6, 2024. https://www.uptodate.com/contents/treatment-of-herpes-zoster
References
  1. Centers for Disease Control and Prevention. Chickenpox (varicella) for healthcare professionals. Published October 21, 2022. Accessed March 6, 2024. https://www.cdc.gov/chickenpox/hcp/index.html#vaccination-impact
  2. National Center for Health Statistics. Immunization. Published June 13, 2023. Accessed March 6, 2024. https://www.cdc.gov/nchs/fastats/immunize.htm
  3. Lee YH, Choe YJ, Lee J, et al. Global varicella vaccination programs. Clin Exp Pediatr. 2022;65:555. doi:10.3345/CEP.2021.01564
  4. Leung J, Lopez AS, Marin M. Changing epidemiology of varicella outbreaks in the United States during the Varicella Vaccination Program, 1995–2019. J Infect Dis. 2022;226(suppl 4):S400-S406.
  5. Arkansas Department of Health. Rules Pertaining to Reportable Diseases. Published September 11, 2023. Accessed March 6, 2024. https://www.healthy.arkansas.gov/images/uploads/rules/ReportableDiseaseList.pdf
  6. Pergam S, Limaye A; The AST Infectious Diseases Community of Practice. Varicella zoster virus (VZV). Am J Transplant. 2009;9(suppl 4):S108-S115. doi:10.1111/J.1600-9143.2009.02901.X
  7. Hoyt B, Bhawan J. Histological spectrum of cutaneous herpes infections. Am J Dermatopathol. 2014;36:609-619. doi:10.1097/DAD.0000000000000148
  8. Oumarou Hama H, Aboudharam G, Barbieri R, et al. Immunohistochemical diagnosis of human infectious diseases: a review. Diagn Pathol. 2022;17. doi:10.1186/S13000-022-01197-5
  9. World Health Organization. Mpox (monkeypox). Published April 18, 2023. Accessed March 7, 2024. https://www.who.int/news-room/fact-sheets/detail/monkeypox
  10. Akram SM, Jamil RT, Gossman W. Rickettsia akari (Rickettsialpox). StatPearls [Internet]. Updated May 8, 2023. Accessed February 29, 2024. https://www.ncbi.nlm.nih.gov/books/NBK448081/
  11. Lott JP, Liu K, Landry ML, et al. Atypical hand-foot-mouth disease associated with coxsackievirus A6 infection. J Am Acad Dermatol. 2013;69:736. doi:10.1016/J.JAAD.2013.07.024
  12. Petrun B, Williams V, Brice S. Disseminated varicella-zoster virus in an immunocompetent adult. Dermatol Online J. 2015;21. doi:10.5070/D3213022343
  13. Kimberlin D, Barnett E, Lynfield R, et al. Exposure to specific pathogens. In: Red Book: 2021-2024 Report of the Committee of Infectious Disease. 32nd ed. American Academy of Pediatrics; 2021:1007-1009.
  14. Treatment of varicella (chickenpox) infection. UpToDate [Internet]. Updated February 7, 2024. Accessed March 6, 2024. https://www.uptodate.com/contents/treatment-of-varicella-chickenpox-infection
  15. Treatment of herpes zoster in the immunocompetent host. UpToDate [Internet]. Updated November 29, 2023. Accessed March 6, 2024. https://www.uptodate.com/contents/treatment-of-herpes-zoster
Issue
Cutis - 113(3)
Issue
Cutis - 113(3)
Page Number
E16-E19
Page Number
E16-E19
Publications
Publications
Topics
Article Type
Display Headline
The Role of Dermatology in Identifying and Reporting a Primary Varicella Outbreak
Display Headline
The Role of Dermatology in Identifying and Reporting a Primary Varicella Outbreak
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Richards Varicella</fileName> <TBEID>0C02F3B9.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F3B9</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Case Letter</storyname> <articleType>1</articleType> <TBLocation>Published-CT</TBLocation> <QCDate/> <firstPublished>20240404T120352</firstPublished> <LastPublished>20240404T120743</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240404T120351</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Richards</byline> <bylineText>Madeline P. Richards, BS; Sara C. Shalin, MD, PhD; Mavinder Guram, MD; Keyur Vyas, MD</bylineText> <bylineFull>Richards</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E16-E19</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>To the Editor:Cases of primary varicella-zoster virus (VZV) are relatively uncommon in the United States since the introduction of the varicella vaccine in 1995</metaDescription> <articlePDF>301008</articlePDF> <teaserImage/> <title>The Role of Dermatology in Identifying and Reporting a Primary Varicella Outbreak</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>2</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>March</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>3</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2307</CMSID> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>dermatopathology</keyword> <keyword> diversity in medicine</keyword> <keyword> infectious disease</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>March 2024</pubIssueName> <pubArticleType>Departments | 2159</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Case Letter | 2307<pubSubsection/></pubSection> </pubSections> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">44</term> </sections> <topics> <term canonical="true">204</term> <term>66772</term> <term>234</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002701.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>The Role of Dermatology in Identifying and Reporting a Primary Varicella Outbreak</title> <deck/> </itemMeta> <itemContent> <p>To the Editor:<br/><br/>Cases of primary varicella-zoster virus (VZV) are relatively uncommon in the United States since the introduction of the varicella vaccine in 1995, with an overall decline in cases of more than 97%.<sup>1</sup> Prior to the vaccine, 70% of hospitalizations occurred in children; subsequently, hospitalizations among the pediatric population (aged ≤20 years) declined by 97%. Compared to children, adults and immunocompromised patients with VZV infection may present with more severe disease and experience more complications.<sup>1</sup> </p> <p>Most children in the United States are vaccinated against VZV, with 90.3% receiving at least 1 dose by 24 months of age.<sup>2</sup> However, many countries do not implement universal varicella vaccination for infants.<sup>3</sup> As a result, physicians should remember to include primary varicella in the differential when clinically correlated, especially when evaluating patients who have immigrated to the United States or who may be living in unvaccinated communities. We report 2 cases of primary VZV manifesting in adults to remind readers of the salient clinical features of this disease and how dermatologists play a critical role in early and accurate identification of diseases that can have wide-reaching public health implications.<br/><br/>A 26-year-old man with no relevant medical history presented to the emergency department with an itchy and painful rash of 5 days’ duration that began on the trunk and spread to the face, lips, feet, hands, arms, and legs. He also reported shortness of breath, cough, and chills, and he had a temperature of 100.8 <span class="body">°</span>F (38.2 <span class="body">°</span>C). Physical examination revealed numerous erythematous papules and vesiculopustules, some with central umbilication and some with overlying gold crusts (Figure 1). <br/><br/>Later that day, a 47-year-old man with no relevant medical history presented to the same emergency department with a rash along with self-reported fever and sore throat of 3 days’ duration. Physical examination found innumerable erythematous vesicopustules scattered on the face, scalp, neck, trunk, arms, and legs, some with a “dew drop on a rose petal” appearance and some with overlying hemorrhagic crust (Figure 2). <br/><br/>Although infection was of primary concern for the first patient, the presentation of the second patient prompted specific concern for primary VZV infection in both patients, who were placed on airborne and contact isolation precautions.<br/><br/>Skin biopsies from both patients showed acantholytic blisters, hair follicle necrosis, and marked dermal inflammation (Figure 3). Herpetic viral changes were seen in keratinocytes, with steel-grey nuclei, multinucleated keratinocytes, and chromatin margination. An immunostain for VZV was diffusely positive, and VZV antibody IgG was positive (Figure 4). <br/><br/>Upon additional questioning, both patients reported recent exposure to VZV-like illnesses in family members without a history of international travel. Neither of the patients was sure of their vaccination status or prior infection history. Both patients received intravenous acyclovir 10 mg/kg administered every 8 hours. Both patients experienced improvement and were discharged after 3 days on oral valacyclovir (1 g 3 times daily for a 7-day treatment course). <br/><br/>The similar presentation and timing of these 2 VZV cases caused concern for an unidentified community outbreak. The infection control team was notified; additionally, per hospital protocol the state health department was alerted as well as the clinicians and staff of the hospital with a request to be vigilant for further cases.<br/><br/>Despite high vaccination rates in the United States, outbreaks of varicella still occur, particularly among unvaccinated individuals, and a robust and efficient response is necessary to control the spread of such outbreaks.<sup>4</sup> Many states, including Arkansas where our cases occurred, have laws mandating report of VZV cases to the department of health.<sup>5</sup> Dermatologists play an important role in reporting cases, aiding in diagnosis through recognition of the physical examination findings, obtaining appropriate biopsy, and recommending additional laboratory testing. <br/><br/>Typical skin manifestations include a pruritic rash of macules, papules, vesicles, and crusted lesions distributed throughout the trunk, face, arms, and legs. Because new lesions appear over several days, they will be in different stages of healing, resulting in the simultaneous presence of papules, vesicles, and crusted lesions.<sup>6</sup> This unique characteristic helps distinguish VZV from other skin diseases such as smallpox or mpox (monkeypox), which generally show lesions in similar stages of evolution. <br/><br/>Biopsy also can aid in identification. Viruses in the herpes family reveal similar histopathologic characteristics, including acantholysis and vesicle formation, intranuclear inclusions with margination of chromatin, multinucleation, and nuclear molding.<sup>7</sup> Immunohistochemistry can be used to differentiate VZV from herpes simplex virus; however, neither microscopic examination nor immunohistochemistry distinguish primary VZV infection from herpes zoster (HZ).<sup>8</sup> <br/><br/>The mpox rash progresses more slowly than a VZV rash and has a centrifugal rather than central distribution that can involve the palms and soles. Lymphadenopathy is a characteristic finding in mpox.<sup>9</sup> Rickettsialpox is distinguished from VZV primarily by the appearance of brown or black eschar after the original papulovesicular lesions dry out.<sup>10</sup> Atypical hand, foot, and mouth disease can manifest in adults as widespread papulovesicular lesions. This form is associated with coxsackievirus A6 and may require direct fluorescent antibody assay or polymerase chain reaction of keratinocytes to rule out VZV.<sup>11</sup> <br/><br/>Herpes zoster occurs in older adults with a history of primary VZV.<sup>6</sup> It manifests as vesicular lesions confined to 1 or 2 adjacent dermatomes vs the diffuse spread of VZV over the entire body. However, HZ can become disseminated in immunocompromised individuals, making it difficult to clinically distinguish from VZV.<sup>6</sup> Serology can be helpful, as high IgM titers indicate an acute primary VZV infection. Subsequently increased IgG titers steadily wane over time and spike during reactivation.<sup>12</sup> <br/><br/>Dermatology and infectious disease consultations in our cases yielded a preliminary diagnosis through physical examination that was confirmed by biopsy and subsequent laboratory testing, which allowed for a swift response by the infection control team including isolation precautions to control a potential outbreak. Patients with VZV should remain in respiratory isolation until all lesions have crusted over.<sup>6</sup> <br/><br/>Individuals who had face-to-face indoor contact for at least 5 minutes or who shared a living space with an infected individual should be assessed for VZV immunity, which is defined as confirmed prior immunization or infection.<sup>5,13</sup> Lack of VZV immunity requires postexposure prophylaxis—active immunization for the immunocompetent and passive immunization for the immunocompromised.<sup>13</sup> Ultimately, no additional cases were reported in the community where our patients resided. <br/><br/>Immunocompetent children with primary VZV require supportive care only. Oral antiviral therapy is the treatment of choice for immunocompetent adults or anyone at increased risk for complications, while intravenous antivirals are recommended for the immunocompromised or those with VZV-related complications.<sup>14</sup> A similar approach is used for HZ. Uncomplicated cases are treated with oral antivirals, and complicated cases (eg, HZ ophthalmicus) are treated with intravenous antivirals.<sup>15</sup> Commonly used antivirals include acyclovir, valacyclovir, and famciclovir.<sup>14</sup> <br/><br/>Our cases highlight the ongoing risk for varicella outbreaks in unvaccinated or undervaccinated communities. Physician vigilance is necessary, and dermatology plays a particularly important role in swift and accurate detection of VZV, as demonstrated in our cases by the recognition of classic physical examination findings of erythematous and vesicular papules in each of the patients. Because primary VZV infection can result in life-threatening complications including hepatitis, encephalitis, and pancreatitis, prompt identification and initiation of therapy is important.<sup>6</sup> Similarly, quick notification of public health officials about detected primary VZV cases is vital to containing potential community outbreaks. </p> <h2>References</h2> <p class="reference"> 1. Centers for Disease Control and Prevention. Chickenpox (varicella) for healthcare professionals. Published October 21, 2022. Accessed March 6, 2024. https://www.cdc.gov/chickenpox/hcp/index.html#vaccination-impact <br/><br/> 2. National Center for Health Statistics. Immunization. Published June 13, 2023. Accessed March 6, 2024. https://www.cdc.gov/nchs/fastats/immunize.htm <br/><br/> 3. Lee YH, Choe YJ, Lee J, et al. Global varicella vaccination programs. <i>Clin Exp Pediatr</i>. 2022;65:555. doi:10.3345/CEP.2021.01564<br/><br/> 4. Leung J, Lopez AS, Marin M. Changing epidemiology of varicella outbreaks in the United States during the Varicella Vaccination Program, 1995–2019. <i>J Infect Dis</i>. 2022;226(suppl 4):S400-S406.<br/><br/> 5. Arkansas Department of Health. <i>Rules Pertaining to Reportable Diseases</i>. Published September 11, 2023. Accessed March 6, 2024. https://www.healthy.arkansas.gov/images/uploads/rules/ReportableDiseaseList.pdf <br/><br/> 6. Pergam S, Limaye A; The AST Infectious Diseases Community of Practice. Varicella zoster virus (VZV). <i>Am J Transplant</i>. 2009;9(suppl 4):S108-S115. doi:10.1111/J.1600-9143.2009.02901.X<br/><br/> 7. Hoyt B, Bhawan J. Histological spectrum of cutaneous herpes infections. <i>Am J Dermatopathol</i>. 2014;36:609-619. doi:10.1097/DAD.0000000000000148<br/><br/> 8. Oumarou Hama H, Aboudharam G, Barbieri R, et al. Immunohistochemical diagnosis of human infectious diseases: a review. <i>Diagn Pathol</i>. 2022;17. doi:10.1186/S13000-022-01197-5<br/><br/> 9. World Health Organization. Mpox (monkeypox)<i>.</i> Published April 18, 2023. Accessed March 7, 2024. https://www.who.int/news-room/fact-sheets/detail/monkeypox<br/><br/>10. Akram SM, Jamil RT, Gossman W. <i>Rickettsia akari</i> (Rickettsialpox). StatPearls [Internet]. Updated May 8, 2023. Accessed February 29, 2024. https://www.ncbi.nlm.nih.gov/books/NBK448081/<br/><br/>11. Lott JP, Liu K, Landry ML, et al. Atypical hand-foot-mouth disease associated with coxsackievirus A6 infection. <i>J Am Acad Dermatol</i>. 2013;69:736. doi:10.1016/J.JAAD.2013.07.024<br/><br/>12. Petrun B, Williams V, Brice S. Disseminated varicella-zoster virus in an immunocompetent adult. <i>Dermatol Online J</i>. 2015;21. doi:10.5070/D3213022343<br/><br/>13. Kimberlin D, Barnett E, Lynfield R, et al. Exposure to specific pathogens. In: <i>Red Book: 2021-2024 Report of the Committee of Infectious Disease</i>. 32nd ed. American Academy of Pediatrics; 2021:1007-1009.<br/><br/>14. Treatment of varicella (chickenpox) infection. UpToDate [Internet]. Updated February 7, 2024. Accessed March 6, 2024. https://www.uptodate.com/contents/treatment-of-varicella-chickenpox-infection<br/><br/>15. Treatment of herpes zoster in the immunocompetent host. UpToDate [Internet]. Updated November 29, 2023. Accessed March 6, 2024. https://www.uptodate.com/contents/treatment-of-herpes-zoster</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From the University of Arkansas for Medical Sciences. Madeline P. Richards is from the College of Medicine, North Little Rock. Dr. Shalin is from the Departments of Dermatology and Pathology, Dr. Guram is from the Department of Dermatology, and Dr. Vyas is from the Department of Internal Medicine, Little Rock. </p> <p class="disclosure">Madeline P. Richards and Drs. Shalin and Guram report no conflict of interest. Dr. Vyas has served as a paid consultant to the American Association of Hip and Knee Surgeons. <br/><br/>Correspondence: Madeline P. Richards, BS, 324 UAMS Campus Dr, Mail Slot #576, Little Rock, AR 72205 (mprichards@uams.edu). <br/><br/><i>Cutis</i>. 2024 March;113(3):E16-E19. doi:10.12788/cutis.0981 </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Primary varicella is a relatively infrequent occurrence since the introduction of vaccination, creating the need for a reminder on the importance of including it in the differential when clinically appropriate. </li> <li>When outbreaks do happen, typically among unvaccinated communities, swift identification via physical examination and histology is imperative to allow infection control teams and public health officials to quickly take action. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Primary varicella is a relatively infrequent occurrence since the introduction of vaccination, creating the need for a reminder on the importance of including it in the differential when clinically appropriate.
  • When outbreaks do happen, typically among unvaccinated communities, swift identification via physical examination and histology is imperative to allow infection control teams and public health officials to quickly take action.
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
Image
Teambase ID
18002701.SIG
Disable zoom
Off