User login
Hidradenitis Suppurativa
THE PRESENTATION
Severe long-standing hidradenitis suppurativa (Hurley stage III) with architectural changes, ropy scarring, granulation tissue, and purulent discharge in the axilla of a 35-year-old Black man (A) and a 42-year-old Hispanic woman with a light skin tone (B).
Hidradenitis suppurativa (HS) is a chronic inflammatory condition of the follicular epithelium that most commonly is found in the axillae and buttocks, as well as the inguinal, perianal, and submammary areas. It is characterized by firm and tender chronic nodules, abscesses complicated by sinus tracts, fistulae, and scarring thought to be related to follicular occlusion. Double-open comedones also may be seen.
The Hurley staging system is widely used to characterize the extent of disease in HS patients:
- Stage I (mild): nodule(s) and abscess(es) without sinus tracts (tunnels) or scarring;
- Stage II (moderate): recurrent nodule(s) and abscess(es) with a limited number of sinus tracts (tunnels) and/or scarring; and
- Stage III (severe): multiple or extensive sinus tracts (tunnels), abscesses, and/or scarring across the entire area.
Epidemiology
Hidradenitis suppurativa is most common in adults and African American patients. It has a prevalence of 1.3% in African Americans.1 When it occurs in children, it generally develops after the onset of puberty. The incidence is higher in females as well as individuals with a history of smoking and obesity (a higher body mass index).2-5
Key clinical features in people with darker skin tones
The erythema associated with HS may be difficult to see in darker skin tones, but violaceous, dark brown, and gray lesions may be present. When active HS lesions subside, intense hyperpigmentation may be left behind, and in some skin tones a pink or violaceous lesion may be apparent.
Worth noting
Hidradenitis suppurativa is disfiguring and has a negative impact on quality of life, including social relationships. Mental health support and screening tools are useful. Pain also is a common concern and may warrant referral to a pain specialist.6 In early disease, HS lesions can be misdiagnosed as an infection that recurs in the same location.
Treatments for HS include oral antibiotics (ie, tetracyclines, rifampin, clindamycin), topical antibiotics, immunosuppressing biologics, metformin, and spironolactone.7 Surgical interventions may be considered earlier in HS management and vary based on the location and severity of the lesions.8
Patients with HS are at risk for developing squamous cell carcinoma in scars even many years later9; therefore, patients should perform skin checks and be referred to a dermatologist. Squamous cell carcinoma is most commonly found on the buttocks of men with HS and has a poor prognosis.
Health disparity highlight
Although those of African American and African descent have the highest rates of HS,1 the clinical trials for adalimumab (the only biologic approved for HS) enrolled a low number of Black patients.
Thirty HS comorbidities have been identified. Garg et al10 recommended that dermatologists perform examinations for comorbid conditions involving the skin and conduct a simple review of systems for extracutaneous comorbidities. Access to medical care is essential, and health care system barriers affect the ability of some patients to receive adequate continuity of care.
The diagnosis of HS often is delayed due to lack of HS knowledge about the condition in the medical community at large and delayed presentation to a dermatologist.
- Sachdeva M, Shah M, Alavi A. Race-specific prevalence of hidradenitis suppurativa [published online November 11, 2020]. J Cutan Med Surg. 2021;25:177-187. doi:10.1177/1203475420972348
- Zouboulis CC, Goyal M, Byrd AS. Hidradenitis suppurativa in skin of colour. Exp Dermatol. 2021;30(suppl 1):27-30. doi:10.1111 /exd.14341
- Shalom G, Cohen AD. The epidemiology of hidradenitis suppurativa: what do we know? Br J Dermatol. 2019;180:712-713.
- Theut Riis P, Pedersen OB, Sigsgaard V, et al. Prevalence of patients with self-reported hidradenitis suppurativa in a cohort of Danish blood donors: a cross-sectional study. Br J Dermatol. 2019;180:774-781.
- Jemec GB, Kimball AB. Hidradenitis suppurativa: epidemiology and scope of the problem. J Am Acad Dermatol. 2015;73 (5 suppl 1):S4-S7.
- Savage KT, Singh V, Patel ZS, et al. Pain management in hidradenitis suppurativa and a proposed treatment algorithm [published online September 17, 2020]. J Am Acad Dermatol. 2021;85:187-199. doi:10.1016/j.jaad.2020.09.039
- Alikhan A, Sayed C, Alavi A, et al. North American clinical management guidelines for hidradenitis suppurativa: a publication from the United States and Canadian Hidradenitis Suppurativa Foundations: part II: topical, intralesional, and systemic medical management [published online March 11, 2019]. J Am Acad Dermatol. 2019;81:91-101.
- Vellaichamy G, Braunberger TL, Nahhas AF, et al. Surgical procedures for hidradenitis suppurativa. Cutis. 2018;102:13-16.
- Jung JM, Lee KH, Kim Y-J, et al. Assessment of overall and specific cancer risks in patients with hidradenitis suppurativa. JAMA Dermatol. 2020;156:844-853.
- Garg A, Malviya N, Strunk A, et al. Comorbidity screening in hidradenitis suppurativa: evidence-based recommendations from the US and Canadian Hidradenitis Suppurativa Foundations [published online January 23, 2021]. J Am Acad Dermatol. 2022;86:1092-1101. doi:10.1016/j. jaad.2021.01.059
THE PRESENTATION
Severe long-standing hidradenitis suppurativa (Hurley stage III) with architectural changes, ropy scarring, granulation tissue, and purulent discharge in the axilla of a 35-year-old Black man (A) and a 42-year-old Hispanic woman with a light skin tone (B).
Hidradenitis suppurativa (HS) is a chronic inflammatory condition of the follicular epithelium that most commonly is found in the axillae and buttocks, as well as the inguinal, perianal, and submammary areas. It is characterized by firm and tender chronic nodules, abscesses complicated by sinus tracts, fistulae, and scarring thought to be related to follicular occlusion. Double-open comedones also may be seen.
The Hurley staging system is widely used to characterize the extent of disease in HS patients:
- Stage I (mild): nodule(s) and abscess(es) without sinus tracts (tunnels) or scarring;
- Stage II (moderate): recurrent nodule(s) and abscess(es) with a limited number of sinus tracts (tunnels) and/or scarring; and
- Stage III (severe): multiple or extensive sinus tracts (tunnels), abscesses, and/or scarring across the entire area.
Epidemiology
Hidradenitis suppurativa is most common in adults and African American patients. It has a prevalence of 1.3% in African Americans.1 When it occurs in children, it generally develops after the onset of puberty. The incidence is higher in females as well as individuals with a history of smoking and obesity (a higher body mass index).2-5
Key clinical features in people with darker skin tones
The erythema associated with HS may be difficult to see in darker skin tones, but violaceous, dark brown, and gray lesions may be present. When active HS lesions subside, intense hyperpigmentation may be left behind, and in some skin tones a pink or violaceous lesion may be apparent.
Worth noting
Hidradenitis suppurativa is disfiguring and has a negative impact on quality of life, including social relationships. Mental health support and screening tools are useful. Pain also is a common concern and may warrant referral to a pain specialist.6 In early disease, HS lesions can be misdiagnosed as an infection that recurs in the same location.
Treatments for HS include oral antibiotics (ie, tetracyclines, rifampin, clindamycin), topical antibiotics, immunosuppressing biologics, metformin, and spironolactone.7 Surgical interventions may be considered earlier in HS management and vary based on the location and severity of the lesions.8
Patients with HS are at risk for developing squamous cell carcinoma in scars even many years later9; therefore, patients should perform skin checks and be referred to a dermatologist. Squamous cell carcinoma is most commonly found on the buttocks of men with HS and has a poor prognosis.
Health disparity highlight
Although those of African American and African descent have the highest rates of HS,1 the clinical trials for adalimumab (the only biologic approved for HS) enrolled a low number of Black patients.
Thirty HS comorbidities have been identified. Garg et al10 recommended that dermatologists perform examinations for comorbid conditions involving the skin and conduct a simple review of systems for extracutaneous comorbidities. Access to medical care is essential, and health care system barriers affect the ability of some patients to receive adequate continuity of care.
The diagnosis of HS often is delayed due to lack of HS knowledge about the condition in the medical community at large and delayed presentation to a dermatologist.
THE PRESENTATION
Severe long-standing hidradenitis suppurativa (Hurley stage III) with architectural changes, ropy scarring, granulation tissue, and purulent discharge in the axilla of a 35-year-old Black man (A) and a 42-year-old Hispanic woman with a light skin tone (B).
Hidradenitis suppurativa (HS) is a chronic inflammatory condition of the follicular epithelium that most commonly is found in the axillae and buttocks, as well as the inguinal, perianal, and submammary areas. It is characterized by firm and tender chronic nodules, abscesses complicated by sinus tracts, fistulae, and scarring thought to be related to follicular occlusion. Double-open comedones also may be seen.
The Hurley staging system is widely used to characterize the extent of disease in HS patients:
- Stage I (mild): nodule(s) and abscess(es) without sinus tracts (tunnels) or scarring;
- Stage II (moderate): recurrent nodule(s) and abscess(es) with a limited number of sinus tracts (tunnels) and/or scarring; and
- Stage III (severe): multiple or extensive sinus tracts (tunnels), abscesses, and/or scarring across the entire area.
Epidemiology
Hidradenitis suppurativa is most common in adults and African American patients. It has a prevalence of 1.3% in African Americans.1 When it occurs in children, it generally develops after the onset of puberty. The incidence is higher in females as well as individuals with a history of smoking and obesity (a higher body mass index).2-5
Key clinical features in people with darker skin tones
The erythema associated with HS may be difficult to see in darker skin tones, but violaceous, dark brown, and gray lesions may be present. When active HS lesions subside, intense hyperpigmentation may be left behind, and in some skin tones a pink or violaceous lesion may be apparent.
Worth noting
Hidradenitis suppurativa is disfiguring and has a negative impact on quality of life, including social relationships. Mental health support and screening tools are useful. Pain also is a common concern and may warrant referral to a pain specialist.6 In early disease, HS lesions can be misdiagnosed as an infection that recurs in the same location.
Treatments for HS include oral antibiotics (ie, tetracyclines, rifampin, clindamycin), topical antibiotics, immunosuppressing biologics, metformin, and spironolactone.7 Surgical interventions may be considered earlier in HS management and vary based on the location and severity of the lesions.8
Patients with HS are at risk for developing squamous cell carcinoma in scars even many years later9; therefore, patients should perform skin checks and be referred to a dermatologist. Squamous cell carcinoma is most commonly found on the buttocks of men with HS and has a poor prognosis.
Health disparity highlight
Although those of African American and African descent have the highest rates of HS,1 the clinical trials for adalimumab (the only biologic approved for HS) enrolled a low number of Black patients.
Thirty HS comorbidities have been identified. Garg et al10 recommended that dermatologists perform examinations for comorbid conditions involving the skin and conduct a simple review of systems for extracutaneous comorbidities. Access to medical care is essential, and health care system barriers affect the ability of some patients to receive adequate continuity of care.
The diagnosis of HS often is delayed due to lack of HS knowledge about the condition in the medical community at large and delayed presentation to a dermatologist.
- Sachdeva M, Shah M, Alavi A. Race-specific prevalence of hidradenitis suppurativa [published online November 11, 2020]. J Cutan Med Surg. 2021;25:177-187. doi:10.1177/1203475420972348
- Zouboulis CC, Goyal M, Byrd AS. Hidradenitis suppurativa in skin of colour. Exp Dermatol. 2021;30(suppl 1):27-30. doi:10.1111 /exd.14341
- Shalom G, Cohen AD. The epidemiology of hidradenitis suppurativa: what do we know? Br J Dermatol. 2019;180:712-713.
- Theut Riis P, Pedersen OB, Sigsgaard V, et al. Prevalence of patients with self-reported hidradenitis suppurativa in a cohort of Danish blood donors: a cross-sectional study. Br J Dermatol. 2019;180:774-781.
- Jemec GB, Kimball AB. Hidradenitis suppurativa: epidemiology and scope of the problem. J Am Acad Dermatol. 2015;73 (5 suppl 1):S4-S7.
- Savage KT, Singh V, Patel ZS, et al. Pain management in hidradenitis suppurativa and a proposed treatment algorithm [published online September 17, 2020]. J Am Acad Dermatol. 2021;85:187-199. doi:10.1016/j.jaad.2020.09.039
- Alikhan A, Sayed C, Alavi A, et al. North American clinical management guidelines for hidradenitis suppurativa: a publication from the United States and Canadian Hidradenitis Suppurativa Foundations: part II: topical, intralesional, and systemic medical management [published online March 11, 2019]. J Am Acad Dermatol. 2019;81:91-101.
- Vellaichamy G, Braunberger TL, Nahhas AF, et al. Surgical procedures for hidradenitis suppurativa. Cutis. 2018;102:13-16.
- Jung JM, Lee KH, Kim Y-J, et al. Assessment of overall and specific cancer risks in patients with hidradenitis suppurativa. JAMA Dermatol. 2020;156:844-853.
- Garg A, Malviya N, Strunk A, et al. Comorbidity screening in hidradenitis suppurativa: evidence-based recommendations from the US and Canadian Hidradenitis Suppurativa Foundations [published online January 23, 2021]. J Am Acad Dermatol. 2022;86:1092-1101. doi:10.1016/j. jaad.2021.01.059
- Sachdeva M, Shah M, Alavi A. Race-specific prevalence of hidradenitis suppurativa [published online November 11, 2020]. J Cutan Med Surg. 2021;25:177-187. doi:10.1177/1203475420972348
- Zouboulis CC, Goyal M, Byrd AS. Hidradenitis suppurativa in skin of colour. Exp Dermatol. 2021;30(suppl 1):27-30. doi:10.1111 /exd.14341
- Shalom G, Cohen AD. The epidemiology of hidradenitis suppurativa: what do we know? Br J Dermatol. 2019;180:712-713.
- Theut Riis P, Pedersen OB, Sigsgaard V, et al. Prevalence of patients with self-reported hidradenitis suppurativa in a cohort of Danish blood donors: a cross-sectional study. Br J Dermatol. 2019;180:774-781.
- Jemec GB, Kimball AB. Hidradenitis suppurativa: epidemiology and scope of the problem. J Am Acad Dermatol. 2015;73 (5 suppl 1):S4-S7.
- Savage KT, Singh V, Patel ZS, et al. Pain management in hidradenitis suppurativa and a proposed treatment algorithm [published online September 17, 2020]. J Am Acad Dermatol. 2021;85:187-199. doi:10.1016/j.jaad.2020.09.039
- Alikhan A, Sayed C, Alavi A, et al. North American clinical management guidelines for hidradenitis suppurativa: a publication from the United States and Canadian Hidradenitis Suppurativa Foundations: part II: topical, intralesional, and systemic medical management [published online March 11, 2019]. J Am Acad Dermatol. 2019;81:91-101.
- Vellaichamy G, Braunberger TL, Nahhas AF, et al. Surgical procedures for hidradenitis suppurativa. Cutis. 2018;102:13-16.
- Jung JM, Lee KH, Kim Y-J, et al. Assessment of overall and specific cancer risks in patients with hidradenitis suppurativa. JAMA Dermatol. 2020;156:844-853.
- Garg A, Malviya N, Strunk A, et al. Comorbidity screening in hidradenitis suppurativa: evidence-based recommendations from the US and Canadian Hidradenitis Suppurativa Foundations [published online January 23, 2021]. J Am Acad Dermatol. 2022;86:1092-1101. doi:10.1016/j. jaad.2021.01.059
Erythematous Papule on the Nasal Ala
The Diagnosis: Cutaneous Lymphoid Hyperplasia
Cutaneous lymphoid hyperplasia (CLH)(also known as pseudolymphoma or lymphocytoma cutis) is a benign inflammatory condition that typically presents as a flesh-colored to erythematous or violaceous papule or nodule on the head or neck. Cutaneous lymphoid hyperplasia may arise in response to an antigenic stimulus, such as an insect bite, infectious agent (eg, Borrelia species), medication, or foreign body (eg, tattoos and piercings).1,2 Given the benign nature and potential for spontaneous resolution, treatment is conservative; however, high-potency topical steroids, cryosurgery, surgical excision, or local radiotherapy may lead to improvement.3 Our patient was started on clobetasol ointment 0.05% and topical tacrolimus 0.1%. After 3 months of use, she reported lesion improvement, but a new lesion appeared on the nose superior to the original. She was offered a steroid injection and liquid nitrogen freezing but was lost to follow-up.
The histopathologic features of CLH are variable and can resemble a cutaneous B- or T-cell lymphoma (quiz images). If there is B-cell predominance, histopathology typically shows a dense dermal infiltrate of lymphocytes admixed with sparse histiocytes, eosinophils, and plasma cells. Multiple germinal-center phenotype lymphoid follicles also may be seen.4 Histopathology of T-cell–predominant CLH commonly shows CD4+ T helper lymphocytes admixed with CD8+ T cells within the dermis with possible papillary dermal edema and red cell extravasation.5 Immunohistochemical stains for CD3, CD4, CD8, and CD20 usually are positive. Most lymphocytes are CD3+ T cells. Admixed clusters of CD20+ B cells may be present.
Angiolymphoid hyperplasia with eosinophilia is a vascular tumor of the skin composed of endothelial cells and inflammatory cells.6,7 The condition presents as single or multiple flesh-colored to purple papules most commonly on the face, scalp, and ears.8 Histologically, lesions appear as well-circumscribed collections of blood vessels composed of plump endothelial cells and an inflammatory infiltrate with lymphocytes and eosinophils (Figure 1A). Endothelial cells also may have an epithelioid appearance.7 Apparent fenestrations—holes within endothelial cells—may be present (Figure 1B). Surgical excision is the preferred treatment of angiolymphoid hyperplasia with eosinophilia. Success with laser and cryosurgery also has been reported.
Granuloma faciale typically presents as a solitary redbrown papule or plaque on the face. Linear arborizing vessels and dilated follicular openings with brown globules frequently are seen on dermoscopy.9 Although it may resemble CLH clinically, the histopathology of granuloma faciale is characterized by a perivascular and interstitial dermal infiltrate of numerous eosinophils admixed with lymphocytes, plasma cells, and neutrophils underneath a grenz zone (Figure 2).10 Leukocytoclastic vasculitis may be seen in early lesions, and lesions can show variable angiocentric fibrosis.11 Treatment options include intralesional triamcinolone, topical steroids or calcineurin inhibitors, topical psoralen plus UVA, surgical excision, and laser therapy, but outcomes are variable.12
Leukemia cutis is a malignant hematopoietic skin infiltration that presents as multiple pink to red-brown, firm, hemorrhagic papules most frequently involving the head, neck, and trunk.13 Rarely, lesions of leukemia cutis may present as ulcers or bullae. Most lesions occur at presentation of systemic leukemia or in the setting of established leukemia. The cutaneous involvement portends a poor prognosis, strongly correlating with additional extramedullary leukemic involvement.14 Histologic features vary based on the specific type of leukemia (eg, acute myelogenous leukemia). Generally, neoplastic infiltration of the dermis and subcutaneous tissue in a nodular, diffuse, perivascular, or interstitial pattern is seen (Figure 3).15 Leukemia cutis typically resolves after successful treatment of the underlying leukemia.
Mycosis fungoides (MF) is the most common type of cutaneous T-cell lymphoma. In its early stages, MF presents as erythematous, brown, scaly patches and plaques. With progression to the tumor stage of disease, clonal expansion of CD4+ T cells leads to the development of purple papules and nodules.16 Microscopic findings of MF are dependent on the stage of disease. Early patch lesions show superficial or lichenoid lymphocytic infiltration of the epidermal basal layer.17 In the plaque stage, dermal infiltrates and epidermotropism become more pronounced, with increased atypical lymphocytes with cerebriform nuclei and interspersed inflammatory cells (Figure 4). In the tumor stage, lymphocytic infiltrates may involve the entirety of the dermis or extend into the subcutaneous tissue, and malignant cells become larger in size.17 Mycosis fungoides lesions typically stain positive for helper T-cell markers with a minority staining positive for CD8.
- Zhou LL, Mistry N. Cutaneous lymphoid hyperplasia (pseudolymphoma). CMAJ. 2018;190:E398.
- Lackey JN, Xia Y, Cho S, et al. Cutaneous lymphoid hyperplasia: a case report and brief review of the literature. Cutis. 2007;79:445-448.
- Albrecht J, Fine LA, Piette W. Drug-associated lymphoma and pseudolymphoma: recognition and management. Dermatol Clin. 2007;25:233-244, vii.
- Arai E, Shimizu M, Hirose T. A review of 55 cases of cutaneous lymphoid hyperplasia: reassessment of the histopathologic findings leading to reclassification of 4 lesions as cutaneous marginal zone lymphoma and 19 as pseudolymphomatous folliculitis. Hum Pathol. 2005;36:505-511.
- Bergman R, Khamaysi Z, Sahar D, et al. Cutaneous lymphoid hyperplasia presenting as a solitary facial nodule: clinical, histopathological, immunophenotypical, and molecular studies. Arch Dermatol. 2006;142:1561-1566.
- Wells GC, Whimster IW. Subcutaneous angiolymphoid hyperplasia with eosinophilia. Br J Dermatol. 1969;81:1-14.
- Guo R, Gavino AC. Angiolymphoid hyperplasia with eosinophilia. Arch Pathol Lab Med. 2015;139:683-686.
- Olsen TG, Helwig EB. Angiolymphoid hyperplasia with eosinophilia. a clinicopathologic study of 116 patients. J Am Acad Dermatol. 1985;12:781-796.
- Lallas A, Sidiropoulos T, Lefaki I, et al. Photo letter to the editor: dermoscopy of granuloma faciale. J Dermatol Case Rep. 2012;6:59-60.
- Oliveira CC, Ianhez PE, Marques SA, et al. Granuloma faciale: clinical, morphological and immunohistochemical aspects in a series of 10 patients. An Bras Dermatol. 2016;91:803-807.
- Marcoval J, Moreno A, Peyr J. Granuloma faciale: a clinicopathological study of 11 cases. J Am Acad Dermatol. 2004;51:269-273.
- Lindhaus C, Elsner P. Granuloma faciale treatment: a systematic review. Acta Derm Venereol. 2018;98:14-18.
- Haidari W, Strowd LC. Clinical characterization of leukemia cutis presentation. Cutis. 2019;104:326-330; E3.
- Rao AG, Danturty I. Leukemia cutis. Indian J Dermatol. 2012;57:504.
- Desch JK, Smoller BR. The spectrum of cutaneous disease in leukemias. J Cutan Pathol. 1993;20:407-410.
- Yamashita T, Abbade LP, Marques ME, et al. Mycosis fungoides and Sezary syndrome: clinical, histopathological and immunohistochemical review and update. An Bras Dermatol. 2012;87:817-828; quiz 829-830.
- Smoller BR, Bishop K, Glusac E, et al. Reassessment of histologic parameters in the diagnosis of mycosis fungoides. Am J Surg Pathol. 1995;19:1423-1430.
The Diagnosis: Cutaneous Lymphoid Hyperplasia
Cutaneous lymphoid hyperplasia (CLH)(also known as pseudolymphoma or lymphocytoma cutis) is a benign inflammatory condition that typically presents as a flesh-colored to erythematous or violaceous papule or nodule on the head or neck. Cutaneous lymphoid hyperplasia may arise in response to an antigenic stimulus, such as an insect bite, infectious agent (eg, Borrelia species), medication, or foreign body (eg, tattoos and piercings).1,2 Given the benign nature and potential for spontaneous resolution, treatment is conservative; however, high-potency topical steroids, cryosurgery, surgical excision, or local radiotherapy may lead to improvement.3 Our patient was started on clobetasol ointment 0.05% and topical tacrolimus 0.1%. After 3 months of use, she reported lesion improvement, but a new lesion appeared on the nose superior to the original. She was offered a steroid injection and liquid nitrogen freezing but was lost to follow-up.
The histopathologic features of CLH are variable and can resemble a cutaneous B- or T-cell lymphoma (quiz images). If there is B-cell predominance, histopathology typically shows a dense dermal infiltrate of lymphocytes admixed with sparse histiocytes, eosinophils, and plasma cells. Multiple germinal-center phenotype lymphoid follicles also may be seen.4 Histopathology of T-cell–predominant CLH commonly shows CD4+ T helper lymphocytes admixed with CD8+ T cells within the dermis with possible papillary dermal edema and red cell extravasation.5 Immunohistochemical stains for CD3, CD4, CD8, and CD20 usually are positive. Most lymphocytes are CD3+ T cells. Admixed clusters of CD20+ B cells may be present.
Angiolymphoid hyperplasia with eosinophilia is a vascular tumor of the skin composed of endothelial cells and inflammatory cells.6,7 The condition presents as single or multiple flesh-colored to purple papules most commonly on the face, scalp, and ears.8 Histologically, lesions appear as well-circumscribed collections of blood vessels composed of plump endothelial cells and an inflammatory infiltrate with lymphocytes and eosinophils (Figure 1A). Endothelial cells also may have an epithelioid appearance.7 Apparent fenestrations—holes within endothelial cells—may be present (Figure 1B). Surgical excision is the preferred treatment of angiolymphoid hyperplasia with eosinophilia. Success with laser and cryosurgery also has been reported.
Granuloma faciale typically presents as a solitary redbrown papule or plaque on the face. Linear arborizing vessels and dilated follicular openings with brown globules frequently are seen on dermoscopy.9 Although it may resemble CLH clinically, the histopathology of granuloma faciale is characterized by a perivascular and interstitial dermal infiltrate of numerous eosinophils admixed with lymphocytes, plasma cells, and neutrophils underneath a grenz zone (Figure 2).10 Leukocytoclastic vasculitis may be seen in early lesions, and lesions can show variable angiocentric fibrosis.11 Treatment options include intralesional triamcinolone, topical steroids or calcineurin inhibitors, topical psoralen plus UVA, surgical excision, and laser therapy, but outcomes are variable.12
Leukemia cutis is a malignant hematopoietic skin infiltration that presents as multiple pink to red-brown, firm, hemorrhagic papules most frequently involving the head, neck, and trunk.13 Rarely, lesions of leukemia cutis may present as ulcers or bullae. Most lesions occur at presentation of systemic leukemia or in the setting of established leukemia. The cutaneous involvement portends a poor prognosis, strongly correlating with additional extramedullary leukemic involvement.14 Histologic features vary based on the specific type of leukemia (eg, acute myelogenous leukemia). Generally, neoplastic infiltration of the dermis and subcutaneous tissue in a nodular, diffuse, perivascular, or interstitial pattern is seen (Figure 3).15 Leukemia cutis typically resolves after successful treatment of the underlying leukemia.
Mycosis fungoides (MF) is the most common type of cutaneous T-cell lymphoma. In its early stages, MF presents as erythematous, brown, scaly patches and plaques. With progression to the tumor stage of disease, clonal expansion of CD4+ T cells leads to the development of purple papules and nodules.16 Microscopic findings of MF are dependent on the stage of disease. Early patch lesions show superficial or lichenoid lymphocytic infiltration of the epidermal basal layer.17 In the plaque stage, dermal infiltrates and epidermotropism become more pronounced, with increased atypical lymphocytes with cerebriform nuclei and interspersed inflammatory cells (Figure 4). In the tumor stage, lymphocytic infiltrates may involve the entirety of the dermis or extend into the subcutaneous tissue, and malignant cells become larger in size.17 Mycosis fungoides lesions typically stain positive for helper T-cell markers with a minority staining positive for CD8.
The Diagnosis: Cutaneous Lymphoid Hyperplasia
Cutaneous lymphoid hyperplasia (CLH)(also known as pseudolymphoma or lymphocytoma cutis) is a benign inflammatory condition that typically presents as a flesh-colored to erythematous or violaceous papule or nodule on the head or neck. Cutaneous lymphoid hyperplasia may arise in response to an antigenic stimulus, such as an insect bite, infectious agent (eg, Borrelia species), medication, or foreign body (eg, tattoos and piercings).1,2 Given the benign nature and potential for spontaneous resolution, treatment is conservative; however, high-potency topical steroids, cryosurgery, surgical excision, or local radiotherapy may lead to improvement.3 Our patient was started on clobetasol ointment 0.05% and topical tacrolimus 0.1%. After 3 months of use, she reported lesion improvement, but a new lesion appeared on the nose superior to the original. She was offered a steroid injection and liquid nitrogen freezing but was lost to follow-up.
The histopathologic features of CLH are variable and can resemble a cutaneous B- or T-cell lymphoma (quiz images). If there is B-cell predominance, histopathology typically shows a dense dermal infiltrate of lymphocytes admixed with sparse histiocytes, eosinophils, and plasma cells. Multiple germinal-center phenotype lymphoid follicles also may be seen.4 Histopathology of T-cell–predominant CLH commonly shows CD4+ T helper lymphocytes admixed with CD8+ T cells within the dermis with possible papillary dermal edema and red cell extravasation.5 Immunohistochemical stains for CD3, CD4, CD8, and CD20 usually are positive. Most lymphocytes are CD3+ T cells. Admixed clusters of CD20+ B cells may be present.
Angiolymphoid hyperplasia with eosinophilia is a vascular tumor of the skin composed of endothelial cells and inflammatory cells.6,7 The condition presents as single or multiple flesh-colored to purple papules most commonly on the face, scalp, and ears.8 Histologically, lesions appear as well-circumscribed collections of blood vessels composed of plump endothelial cells and an inflammatory infiltrate with lymphocytes and eosinophils (Figure 1A). Endothelial cells also may have an epithelioid appearance.7 Apparent fenestrations—holes within endothelial cells—may be present (Figure 1B). Surgical excision is the preferred treatment of angiolymphoid hyperplasia with eosinophilia. Success with laser and cryosurgery also has been reported.
Granuloma faciale typically presents as a solitary redbrown papule or plaque on the face. Linear arborizing vessels and dilated follicular openings with brown globules frequently are seen on dermoscopy.9 Although it may resemble CLH clinically, the histopathology of granuloma faciale is characterized by a perivascular and interstitial dermal infiltrate of numerous eosinophils admixed with lymphocytes, plasma cells, and neutrophils underneath a grenz zone (Figure 2).10 Leukocytoclastic vasculitis may be seen in early lesions, and lesions can show variable angiocentric fibrosis.11 Treatment options include intralesional triamcinolone, topical steroids or calcineurin inhibitors, topical psoralen plus UVA, surgical excision, and laser therapy, but outcomes are variable.12
Leukemia cutis is a malignant hematopoietic skin infiltration that presents as multiple pink to red-brown, firm, hemorrhagic papules most frequently involving the head, neck, and trunk.13 Rarely, lesions of leukemia cutis may present as ulcers or bullae. Most lesions occur at presentation of systemic leukemia or in the setting of established leukemia. The cutaneous involvement portends a poor prognosis, strongly correlating with additional extramedullary leukemic involvement.14 Histologic features vary based on the specific type of leukemia (eg, acute myelogenous leukemia). Generally, neoplastic infiltration of the dermis and subcutaneous tissue in a nodular, diffuse, perivascular, or interstitial pattern is seen (Figure 3).15 Leukemia cutis typically resolves after successful treatment of the underlying leukemia.
Mycosis fungoides (MF) is the most common type of cutaneous T-cell lymphoma. In its early stages, MF presents as erythematous, brown, scaly patches and plaques. With progression to the tumor stage of disease, clonal expansion of CD4+ T cells leads to the development of purple papules and nodules.16 Microscopic findings of MF are dependent on the stage of disease. Early patch lesions show superficial or lichenoid lymphocytic infiltration of the epidermal basal layer.17 In the plaque stage, dermal infiltrates and epidermotropism become more pronounced, with increased atypical lymphocytes with cerebriform nuclei and interspersed inflammatory cells (Figure 4). In the tumor stage, lymphocytic infiltrates may involve the entirety of the dermis or extend into the subcutaneous tissue, and malignant cells become larger in size.17 Mycosis fungoides lesions typically stain positive for helper T-cell markers with a minority staining positive for CD8.
- Zhou LL, Mistry N. Cutaneous lymphoid hyperplasia (pseudolymphoma). CMAJ. 2018;190:E398.
- Lackey JN, Xia Y, Cho S, et al. Cutaneous lymphoid hyperplasia: a case report and brief review of the literature. Cutis. 2007;79:445-448.
- Albrecht J, Fine LA, Piette W. Drug-associated lymphoma and pseudolymphoma: recognition and management. Dermatol Clin. 2007;25:233-244, vii.
- Arai E, Shimizu M, Hirose T. A review of 55 cases of cutaneous lymphoid hyperplasia: reassessment of the histopathologic findings leading to reclassification of 4 lesions as cutaneous marginal zone lymphoma and 19 as pseudolymphomatous folliculitis. Hum Pathol. 2005;36:505-511.
- Bergman R, Khamaysi Z, Sahar D, et al. Cutaneous lymphoid hyperplasia presenting as a solitary facial nodule: clinical, histopathological, immunophenotypical, and molecular studies. Arch Dermatol. 2006;142:1561-1566.
- Wells GC, Whimster IW. Subcutaneous angiolymphoid hyperplasia with eosinophilia. Br J Dermatol. 1969;81:1-14.
- Guo R, Gavino AC. Angiolymphoid hyperplasia with eosinophilia. Arch Pathol Lab Med. 2015;139:683-686.
- Olsen TG, Helwig EB. Angiolymphoid hyperplasia with eosinophilia. a clinicopathologic study of 116 patients. J Am Acad Dermatol. 1985;12:781-796.
- Lallas A, Sidiropoulos T, Lefaki I, et al. Photo letter to the editor: dermoscopy of granuloma faciale. J Dermatol Case Rep. 2012;6:59-60.
- Oliveira CC, Ianhez PE, Marques SA, et al. Granuloma faciale: clinical, morphological and immunohistochemical aspects in a series of 10 patients. An Bras Dermatol. 2016;91:803-807.
- Marcoval J, Moreno A, Peyr J. Granuloma faciale: a clinicopathological study of 11 cases. J Am Acad Dermatol. 2004;51:269-273.
- Lindhaus C, Elsner P. Granuloma faciale treatment: a systematic review. Acta Derm Venereol. 2018;98:14-18.
- Haidari W, Strowd LC. Clinical characterization of leukemia cutis presentation. Cutis. 2019;104:326-330; E3.
- Rao AG, Danturty I. Leukemia cutis. Indian J Dermatol. 2012;57:504.
- Desch JK, Smoller BR. The spectrum of cutaneous disease in leukemias. J Cutan Pathol. 1993;20:407-410.
- Yamashita T, Abbade LP, Marques ME, et al. Mycosis fungoides and Sezary syndrome: clinical, histopathological and immunohistochemical review and update. An Bras Dermatol. 2012;87:817-828; quiz 829-830.
- Smoller BR, Bishop K, Glusac E, et al. Reassessment of histologic parameters in the diagnosis of mycosis fungoides. Am J Surg Pathol. 1995;19:1423-1430.
- Zhou LL, Mistry N. Cutaneous lymphoid hyperplasia (pseudolymphoma). CMAJ. 2018;190:E398.
- Lackey JN, Xia Y, Cho S, et al. Cutaneous lymphoid hyperplasia: a case report and brief review of the literature. Cutis. 2007;79:445-448.
- Albrecht J, Fine LA, Piette W. Drug-associated lymphoma and pseudolymphoma: recognition and management. Dermatol Clin. 2007;25:233-244, vii.
- Arai E, Shimizu M, Hirose T. A review of 55 cases of cutaneous lymphoid hyperplasia: reassessment of the histopathologic findings leading to reclassification of 4 lesions as cutaneous marginal zone lymphoma and 19 as pseudolymphomatous folliculitis. Hum Pathol. 2005;36:505-511.
- Bergman R, Khamaysi Z, Sahar D, et al. Cutaneous lymphoid hyperplasia presenting as a solitary facial nodule: clinical, histopathological, immunophenotypical, and molecular studies. Arch Dermatol. 2006;142:1561-1566.
- Wells GC, Whimster IW. Subcutaneous angiolymphoid hyperplasia with eosinophilia. Br J Dermatol. 1969;81:1-14.
- Guo R, Gavino AC. Angiolymphoid hyperplasia with eosinophilia. Arch Pathol Lab Med. 2015;139:683-686.
- Olsen TG, Helwig EB. Angiolymphoid hyperplasia with eosinophilia. a clinicopathologic study of 116 patients. J Am Acad Dermatol. 1985;12:781-796.
- Lallas A, Sidiropoulos T, Lefaki I, et al. Photo letter to the editor: dermoscopy of granuloma faciale. J Dermatol Case Rep. 2012;6:59-60.
- Oliveira CC, Ianhez PE, Marques SA, et al. Granuloma faciale: clinical, morphological and immunohistochemical aspects in a series of 10 patients. An Bras Dermatol. 2016;91:803-807.
- Marcoval J, Moreno A, Peyr J. Granuloma faciale: a clinicopathological study of 11 cases. J Am Acad Dermatol. 2004;51:269-273.
- Lindhaus C, Elsner P. Granuloma faciale treatment: a systematic review. Acta Derm Venereol. 2018;98:14-18.
- Haidari W, Strowd LC. Clinical characterization of leukemia cutis presentation. Cutis. 2019;104:326-330; E3.
- Rao AG, Danturty I. Leukemia cutis. Indian J Dermatol. 2012;57:504.
- Desch JK, Smoller BR. The spectrum of cutaneous disease in leukemias. J Cutan Pathol. 1993;20:407-410.
- Yamashita T, Abbade LP, Marques ME, et al. Mycosis fungoides and Sezary syndrome: clinical, histopathological and immunohistochemical review and update. An Bras Dermatol. 2012;87:817-828; quiz 829-830.
- Smoller BR, Bishop K, Glusac E, et al. Reassessment of histologic parameters in the diagnosis of mycosis fungoides. Am J Surg Pathol. 1995;19:1423-1430.
A 35-year-old woman presented with a slowly growing, smooth, erythematous papule of 2 months’ duration on the left nasal ala surrounding a piercing (top, inset) that had been performed 4 years prior. A tangential biopsy was obtained for histopathologic evaluation.
Novel study offers clues to sex bias in lupus
Systemic lupus erythematosus (SLE), or lupus, shows a marked sex bias, affecting about nine females for every one male, according to Susan Kovats, PhD, who studies sex differences in immunity at the Oklahoma Medical Research Foundation in Oklahoma City. This characteristic of lupus suggests that hormones are involved in the pathogenesis of the disease. It also suggests, Dr. Kovats said, that the X chromosome might play a role.
Though studies since the 1970s have indicated a significant role for hormones, the issue is still complex and not well understood, and relatively little research has been done on the molecular mechanisms that might be responsible. This may be because of difficulties with influencing the immune system in vitro, said George A. Robinson, PhD, of University College London’s Centre for Rheumatology.
But Dr. Robinson and his team found a unique way of investigating the role of sex chromosomes and hormones in the inflammatory profiles across subjects of different sex, gender, age, and disease status. In research published online in The Lancet Rheumatology, Dr. Robinson and his team looked at immune cells taken from both cisgender men and women and transgender men and women, and thus were able to “get a more physiological view of what sex hormones are doing to the immune system,” he said.
Dr. Kovats agreed that it was a useful approach. “The transgender people provided an opportunity to effectively separate sex hormone levels from chromosome content,” she said in an interview.
Methods and findings
Peripheral blood mononuclear cell (PBMC) samples were taken from cisgender individuals with and without juvenile-onset lupus and assessed for 28 immune-cell subsets, including different T-cell, B-cell, and monotype subsets. Subjects included 39 postpubertal cisgender men and women (17 men and 22 women) who did not have juvenile-onset lupus, and 35 postpubertal cisgender men and women (12 men and 23 women) who did have juvenile onset lupus. All were aged 16-25 years. The transgender group included five transgender men and five transgender women (aged 18-19) who were undergoing gender-affirming sex hormone treatment.
The analysis found that one of the key differences between young postpubertal cisgender men and age-matched cisgender women was that the men had significantly elevated frequencies of regulatory T cells (T-reg cells), and the T-reg cells from young cisgender men had greater suppressive capacity in vitro than did those from cisgender women. In addition, RNA sequencing data from isolated T-reg cells showed the transcriptomic signature of the cisgender men’s T-regs were significantly enriched for genes in the P13K-AKT signaling pathway. The frequency of T-reg cells was not influenced by sex hormones, but their transcriptomic profile was affected.
“These results are beginning to give us an indication of which genes might be differentially regulated by sex hormones and how these are associated with autoimmunity,” Dr. Robinson said. “We’ve also found that, depending on whether you’re a cisgender man or woman, you may have a different pathogenic process to developing lupus. It’s not necessarily that one mechanism drives the disease across both sexes.”
New approaches, better insights
Dr. Kovats was particularly impressed by the methods of this study. “It was a natural study, the kind of thing we can usually do only in mice,” she said.
“One problem with studies on the effects of hormones in disease is that historically researchers have not paid that much attention to the actual hormone levels in the humans they studied. They might look at 100 women and 100 men, roughly between the ages of 20 and 50. We’re starting to see more, but there aren’t a lot of studies correlating numbers of cells in blood with actual hormone levels in the person. And as we know, just because someone’s a certain age doesn’t mean that they have a textbook hormone level. Early menopause, birth-control pills, many things can affect those levels.”
The researchers hope that these findings will shed light on the mechanisms that create sexual bias in autoimmune diseases, particularly lupus, as well as help researchers to better understand the innate and adaptive immunological differences between men and women. It could also be useful in the clinical setting, Dr. Robinson said. Because of the extreme sex bias in lupus, doctors see far more women with the illness than men. When they do see men with lupus, they need to be able to consider how the patient’s sex affects the development and course of the disease. “I think that people need to start looking at patients as clinically different, depending on their sex and gender,” he said. Information like that analyzed in this study could help with that. This could be especially important because as Dr. Kovats pointed out, although men get lupus far less often than women, when they do have it, they tend to have more severe disease.
Help from machines
This study was groundbreaking in another area as well. The researchers used machine learning to analyze the data. “We’ve started working a lot more with these analysis methods to try to answer as much as we can with these smaller data sets,” Dr. Robinson said. “Rather than the conventional analysis that we would typically perform, we’re able to use machine learning and artificial intelligence to try and learn from the data and increase the numbers that we’re working with by using a training data set. This allows us to interrogate the data with a lot more precision.”
The authors declared no competing interests.
Systemic lupus erythematosus (SLE), or lupus, shows a marked sex bias, affecting about nine females for every one male, according to Susan Kovats, PhD, who studies sex differences in immunity at the Oklahoma Medical Research Foundation in Oklahoma City. This characteristic of lupus suggests that hormones are involved in the pathogenesis of the disease. It also suggests, Dr. Kovats said, that the X chromosome might play a role.
Though studies since the 1970s have indicated a significant role for hormones, the issue is still complex and not well understood, and relatively little research has been done on the molecular mechanisms that might be responsible. This may be because of difficulties with influencing the immune system in vitro, said George A. Robinson, PhD, of University College London’s Centre for Rheumatology.
But Dr. Robinson and his team found a unique way of investigating the role of sex chromosomes and hormones in the inflammatory profiles across subjects of different sex, gender, age, and disease status. In research published online in The Lancet Rheumatology, Dr. Robinson and his team looked at immune cells taken from both cisgender men and women and transgender men and women, and thus were able to “get a more physiological view of what sex hormones are doing to the immune system,” he said.
Dr. Kovats agreed that it was a useful approach. “The transgender people provided an opportunity to effectively separate sex hormone levels from chromosome content,” she said in an interview.
Methods and findings
Peripheral blood mononuclear cell (PBMC) samples were taken from cisgender individuals with and without juvenile-onset lupus and assessed for 28 immune-cell subsets, including different T-cell, B-cell, and monotype subsets. Subjects included 39 postpubertal cisgender men and women (17 men and 22 women) who did not have juvenile-onset lupus, and 35 postpubertal cisgender men and women (12 men and 23 women) who did have juvenile onset lupus. All were aged 16-25 years. The transgender group included five transgender men and five transgender women (aged 18-19) who were undergoing gender-affirming sex hormone treatment.
The analysis found that one of the key differences between young postpubertal cisgender men and age-matched cisgender women was that the men had significantly elevated frequencies of regulatory T cells (T-reg cells), and the T-reg cells from young cisgender men had greater suppressive capacity in vitro than did those from cisgender women. In addition, RNA sequencing data from isolated T-reg cells showed the transcriptomic signature of the cisgender men’s T-regs were significantly enriched for genes in the P13K-AKT signaling pathway. The frequency of T-reg cells was not influenced by sex hormones, but their transcriptomic profile was affected.
“These results are beginning to give us an indication of which genes might be differentially regulated by sex hormones and how these are associated with autoimmunity,” Dr. Robinson said. “We’ve also found that, depending on whether you’re a cisgender man or woman, you may have a different pathogenic process to developing lupus. It’s not necessarily that one mechanism drives the disease across both sexes.”
New approaches, better insights
Dr. Kovats was particularly impressed by the methods of this study. “It was a natural study, the kind of thing we can usually do only in mice,” she said.
“One problem with studies on the effects of hormones in disease is that historically researchers have not paid that much attention to the actual hormone levels in the humans they studied. They might look at 100 women and 100 men, roughly between the ages of 20 and 50. We’re starting to see more, but there aren’t a lot of studies correlating numbers of cells in blood with actual hormone levels in the person. And as we know, just because someone’s a certain age doesn’t mean that they have a textbook hormone level. Early menopause, birth-control pills, many things can affect those levels.”
The researchers hope that these findings will shed light on the mechanisms that create sexual bias in autoimmune diseases, particularly lupus, as well as help researchers to better understand the innate and adaptive immunological differences between men and women. It could also be useful in the clinical setting, Dr. Robinson said. Because of the extreme sex bias in lupus, doctors see far more women with the illness than men. When they do see men with lupus, they need to be able to consider how the patient’s sex affects the development and course of the disease. “I think that people need to start looking at patients as clinically different, depending on their sex and gender,” he said. Information like that analyzed in this study could help with that. This could be especially important because as Dr. Kovats pointed out, although men get lupus far less often than women, when they do have it, they tend to have more severe disease.
Help from machines
This study was groundbreaking in another area as well. The researchers used machine learning to analyze the data. “We’ve started working a lot more with these analysis methods to try to answer as much as we can with these smaller data sets,” Dr. Robinson said. “Rather than the conventional analysis that we would typically perform, we’re able to use machine learning and artificial intelligence to try and learn from the data and increase the numbers that we’re working with by using a training data set. This allows us to interrogate the data with a lot more precision.”
The authors declared no competing interests.
Systemic lupus erythematosus (SLE), or lupus, shows a marked sex bias, affecting about nine females for every one male, according to Susan Kovats, PhD, who studies sex differences in immunity at the Oklahoma Medical Research Foundation in Oklahoma City. This characteristic of lupus suggests that hormones are involved in the pathogenesis of the disease. It also suggests, Dr. Kovats said, that the X chromosome might play a role.
Though studies since the 1970s have indicated a significant role for hormones, the issue is still complex and not well understood, and relatively little research has been done on the molecular mechanisms that might be responsible. This may be because of difficulties with influencing the immune system in vitro, said George A. Robinson, PhD, of University College London’s Centre for Rheumatology.
But Dr. Robinson and his team found a unique way of investigating the role of sex chromosomes and hormones in the inflammatory profiles across subjects of different sex, gender, age, and disease status. In research published online in The Lancet Rheumatology, Dr. Robinson and his team looked at immune cells taken from both cisgender men and women and transgender men and women, and thus were able to “get a more physiological view of what sex hormones are doing to the immune system,” he said.
Dr. Kovats agreed that it was a useful approach. “The transgender people provided an opportunity to effectively separate sex hormone levels from chromosome content,” she said in an interview.
Methods and findings
Peripheral blood mononuclear cell (PBMC) samples were taken from cisgender individuals with and without juvenile-onset lupus and assessed for 28 immune-cell subsets, including different T-cell, B-cell, and monotype subsets. Subjects included 39 postpubertal cisgender men and women (17 men and 22 women) who did not have juvenile-onset lupus, and 35 postpubertal cisgender men and women (12 men and 23 women) who did have juvenile onset lupus. All were aged 16-25 years. The transgender group included five transgender men and five transgender women (aged 18-19) who were undergoing gender-affirming sex hormone treatment.
The analysis found that one of the key differences between young postpubertal cisgender men and age-matched cisgender women was that the men had significantly elevated frequencies of regulatory T cells (T-reg cells), and the T-reg cells from young cisgender men had greater suppressive capacity in vitro than did those from cisgender women. In addition, RNA sequencing data from isolated T-reg cells showed the transcriptomic signature of the cisgender men’s T-regs were significantly enriched for genes in the P13K-AKT signaling pathway. The frequency of T-reg cells was not influenced by sex hormones, but their transcriptomic profile was affected.
“These results are beginning to give us an indication of which genes might be differentially regulated by sex hormones and how these are associated with autoimmunity,” Dr. Robinson said. “We’ve also found that, depending on whether you’re a cisgender man or woman, you may have a different pathogenic process to developing lupus. It’s not necessarily that one mechanism drives the disease across both sexes.”
New approaches, better insights
Dr. Kovats was particularly impressed by the methods of this study. “It was a natural study, the kind of thing we can usually do only in mice,” she said.
“One problem with studies on the effects of hormones in disease is that historically researchers have not paid that much attention to the actual hormone levels in the humans they studied. They might look at 100 women and 100 men, roughly between the ages of 20 and 50. We’re starting to see more, but there aren’t a lot of studies correlating numbers of cells in blood with actual hormone levels in the person. And as we know, just because someone’s a certain age doesn’t mean that they have a textbook hormone level. Early menopause, birth-control pills, many things can affect those levels.”
The researchers hope that these findings will shed light on the mechanisms that create sexual bias in autoimmune diseases, particularly lupus, as well as help researchers to better understand the innate and adaptive immunological differences between men and women. It could also be useful in the clinical setting, Dr. Robinson said. Because of the extreme sex bias in lupus, doctors see far more women with the illness than men. When they do see men with lupus, they need to be able to consider how the patient’s sex affects the development and course of the disease. “I think that people need to start looking at patients as clinically different, depending on their sex and gender,” he said. Information like that analyzed in this study could help with that. This could be especially important because as Dr. Kovats pointed out, although men get lupus far less often than women, when they do have it, they tend to have more severe disease.
Help from machines
This study was groundbreaking in another area as well. The researchers used machine learning to analyze the data. “We’ve started working a lot more with these analysis methods to try to answer as much as we can with these smaller data sets,” Dr. Robinson said. “Rather than the conventional analysis that we would typically perform, we’re able to use machine learning and artificial intelligence to try and learn from the data and increase the numbers that we’re working with by using a training data set. This allows us to interrogate the data with a lot more precision.”
The authors declared no competing interests.
FROM THE LANCET RHEUMATOLOGY
Can Atopic Dermatitis and Allergic Contact Dermatitis Coexist?
Atopic dermatitis (AD) and allergic contact dermatitis (ACD) are 2 common inflammatory skin conditions that may have similar clinical presentations. Historically, it was thought that these conditions could not be diagnosed simultaneously due to their differing immune mechanisms; however, this belief has been challenged by recent evidence suggesting a more nuanced relationship between the 2 disease processes. In this review, we examine the complex interplay between AD and ACD and explain how shifts in conventional understanding of the 2 conditions shaped our evolving recognition of their ability to coexist.
Epidemiology of AD and ACD
Atopic dermatitis is the most common inflammatory skin disease in children and adolescents, with an estimated prevalence reaching 21%.1 In 60% of cases, onset of AD will occur within the first year of life, and 90% of cases begin within the first 5 years.2 Resolution may occur by adulthood; however, AD may continue to impact up to 8% to 9% of adults, with an increased prevalence in those older than 75 years.1 This may represent an underestimation of the burden of adult AD; one systematic review of 17 studies found that the pooled proportion of adult-onset AD was greater than 25%.3
In contrast, ACD previously was assumed to be a disease that more commonly impacted adults and only rarely children, primarily due to an early misconception that children were not frequently exposed to contact allergens and their immune systems were too immature to react to them even if exposed.4,5 However, it is now known that children do have risk factors for development of ACD, including a thinner stratum corneum and potentially a more absorbent skin surface.4 In addition, a 2022 study by the North American Contact Dermatitis Group (NACDG) found similar rates of ACD in children (n=1871) and adults (n=41,699) referred for patch testing (55.2% and 57.3%, respectively) as well as similar rates of having at least 1 relevant positive patch test (49.2% and 52.2%).6
In opposition to traditional beliefs, these findings highlight that AD and ACD can occur across age groups.
Immune Mechanism
The pathogenesis of AD represents a multifactorial process involving the immune system, cutaneous flora, genetic predisposition, and surrounding environment. Immunologically, acute AD is driven by a predominantly TH2 helper T-cell response with high levels of IL-4, IL-5, and IL-137; TH22, TH17, and TH1 also have been implicated.8 Notably, TH17 is found in high levels during the acute eczema phase, while TH1 and TH22are associated with the chronic phase.7
The pathophysiology of ACD is not completely understood. The classic paradigm involves 2 phases: sensitization and elicitation. Sensitization involves antigen-presenting cells that take up allergens absorbed by the skin to present them in regional lymph nodes where antigen-specific T lymphocytes are generated. Elicitation occurs upon re-exposure to the allergen, at which time the primed T lymphocytes are recruited to the skin, causing inflammation.9 Allergic contact dermatitis initially was thought to be driven by TH1 cytokines and IL-17 but now is understood to be more complex.10 Studies have revealed immune polarization of contact allergens, demonstrating that nickel primarily induces a TH1/TH17 response, whereas fragrance and rubber accelerators skew to TH2; TH9 and TH22 also may be involved depending on the causative allergen.11,12
Of note, the immunologic differences between AD and ACD led early investigators to believe that patients with AD were relatively protected from ACD.13 However, as previously described, there are several overlapping cytokines between AD and ACD. Furthermore, research has revealed that risk of contact sensitization might be increased in the chronic eczema phase due to the shared TH1 pathway.14 Barrier-disrupted skin (such as that in AD) also may increase the cytokine response and the density of antigen-presenting cells, leading to a proallergic state.15 This suggests that the immunologic pathways of AD and ACD are more intertwined than was previously understood.
Underlying Risk Factors
Skin barrier dysfunction is a key step in the pathogenesis of AD. Patients with AD commonly have loss-of-function mutations in the filaggrin gene, a protein that is key to the function of the stratum corneum. Loss of this protein may not only impact the immune response as previously noted but also may lead to increased transepidermal water loss and bacterial colonization.16 Interestingly, a 2014 review examined how this mutation could lead to an increased risk of sensitization to bivalent metal ions via an impaired chelating ability of the skin.17 Furthermore, a 2016 study conducted in Dutch construction workers revealed an increased risk for contact dermatitis (irritant and allergic) for those with a loss-of-function filaggrin mutation.18
Importantly, this same mutation may explain why patients with AD tend to have increased skin colonization by Staphylococcus aureus. The abundance of S aureus and the relative decrease in the diversity of other microorganisms on the skin may be associated with increased AD severity.19 Likewise, S aureus may play a role in the pathogenesis of ACD via production of its exotoxin directed at the T-cell receptor V beta 17 region. In particular, this receptor has been associated with nickel sensitization.17
Another risk factor to consider is increased exposure to contact sensitizers when treating AD. For instance, management often includes use of over-the-counter emollients, natural or botanical remedies with purported benefits for AD, cleansers, and detergents. However, these products can contain some of the most prevalent contact allergens seen in those with AD, including methyl-isothiazolinone, formaldehyde releasers, and fragrance.20 Topical corticosteroids also are frequently used, and ACD to steroid molecules can occur, particularly to tixocortol-21-pivalate (a marker for class A corticosteroids) and budesonide (a marker for class B corticosteroids).21 Other allergens (eg, benzyl alcohol, propylene glycol) also may be found as inactive ingredients of topical corticosteroids.22 These exposures may place AD patients at risk for ACD.
The Coexistence of AD and ACD
Given the overlapping epidemiology, immunology, and potentially increased risk for the development of ACD in patients with AD, it would be reasonable to assume that the 2 diagnoses could coexist; however, is there clinical data to support this idea? Based on recent database reviews, the answer appears to be yes.20,23-26 An analysis from the Pediatric Contact Dermatitis Registry revealed that 30% of 1142 pediatric patch test cases analyzed were diagnosed as AD and ACD simultaneously.24 The NACDG found similar results in its 2021 review, as 29.5% of children (n=1648) and 20.7% of adults (n=36,834) had a concurrent diagnosis of AD and ACD.20 Notably, older results from these databases also demonstrated an association between the 2 conditions.23,25,26
It remains unclear whether the prevalence of ACD is higher in those with or without AD. A comprehensive systematic review conducted in 2017 examined this topic through analysis of 74 studies. The results demonstrated a similar prevalence of contact sensitization in individuals with and without AD.27 Another systematic review of 31 studies conducted in 2017 found a higher prevalence for ACD in children without AD; however, the authors noted that the included studies were too variable (eg, size, design, allergens tested) to draw definitive conclusions.28
Even though there is no clear overall increased risk for ACD in patients with AD, research has suggested that certain allergens may be more prevalent in the setting of AD. An NACDG study found that adults with AD had increased odds of reacting to 10 of the top 25 NACDG screening allergens compared to those without AD.20 Other studies have found that AD patients may be more likely to become sensitized to certain allergens, such as fragrance and lanolin.14
Considerations for Management
Diagnosis of ACD in patients with AD can be challenging because these conditions may present similarly with chronic, pruritic, inflammatory patches and plaques. Chronic ACD may be misdiagnosed as AD if patch testing is not performed.29 Given the prevalence of ACD in the setting of AD, there should be a low threshold to pursue patch testing, especially when dermatitis is recalcitrant to standard therapies or presents in an atypical distribution (ie, perioral, predominantly head/neck, hand and foot, isolated eyelid involvement, buttocks).4,30 Various allergen series are available for patch testing adults and children including the NACDG Standard Series, American Contact Dermatitis Society Core Allergen Series, or the Pediatric Baseline Series.31-33
If potentially relevant allergens are uncovered by patch testing, patients should be counseled on avoidance strategies. However, allergen avoidance may not always lead to complete symptom resolution, especially if AD is present concomitantly with ACD. Therefore, use of topical or systemic therapies still may be required. Topical corticosteroids can be used when dermatitis is acute and localized. Systemic corticosteroids are utilized for both diagnoses when cases are more severe or extensive, but their adverse-effect profile limits long-term use. Other systemic treatments, including conventional agents (ie, azathioprine, cyclosporine, methotrexate, mycophenolate mofetil), biologics, and small molecule inhibitors also may be considered for severe cases.34,35 Dupilumab, a monoclonal antibody targeting IL-4/IL-13, is approved for use in moderate to severe AD in patients 6 months and older. Recent evidence has suggested that dupilumab also may be an effective off-label treatment choice for ACD when allergen avoidance alone is insufficient.36 Studies have been conducted on secukinumab, a monoclonal antibody against IL-17; however, it has not been shown to be effective in either AD or ACD.37,38 This indicates that targeted biologics may not always be successful in treating these diagnoses, likely due to their complex immune pathways. Finally, there is an emerging role for JAK inhibitors. Three are approved for AD: topical ruxolitinib, oral abrocitinib, and oral upadacitinib.39 Further investigation is needed to determine the efficacy of JAK inhibitors in ACD.
Final Interpretation
Evolving evidence shows that AD and ACD can occur at the same time despite the historical perspective that their immune pathways were too polarized for this to happen. Atopic dermatitis may be an important risk factor for subsequent development of ACD. Management should include a low threshold to perform patch testing, while pharmacotherapies utilized in the treatment of both conditions should be considered.
- Chan LN, Magyari A, Ye M, et al. The epidemiology of atopic dermatitis in older adults: a population-based study in the United Kingdom. PLoS One. 2021;16:E0258219. doi:10.1371/journal.pone.0258219
- Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: section 1. diagnosis and assessment of atopic dermatitis [published online November 27, 2013]. J Am Acad Dermatol. 2014;70:338-351. doi:10.1016/j.jaad.2013.10.010
- Lee HH, Patel KR, Singam V, et al. A systematic review and meta-analysis of the prevalence and phenotype of adult-onset atopic dermatitis [published online June 2, 2018]. J Am Acad Dermatol. 2019;80:1526-1532.e7. doi:10.1016/j.jaad.2018.05.1241
- Borok J, Matiz C, Goldenberg A, et al. Contact dermatitis in atopic dermatitis children—past, present, and future. Clin Rev Allergy Immunol. 2019;56:86-98. doi:10.1007/s12016-018-8711-2
- Goldenberg A, Silverberg N, Silverberg JI, et al. Pediatric allergic contact dermatitis: lessons for better care. J Allergy Clin Immunol Pract. 2015;3:661-667; quiz 668. doi:10.1016/j.jaip.2015.02.007
- Silverberg JI, Hou A, Warshaw EM, et al. Age-related differences in patch testing results among children: analysis of North American Contact Dermatitis Group data, 2001-2018 [published online July 24, 2021]. J Am Acad Dermatol. 2022;86:818-826. doi:10.1016/j.jaad.2021.07.030
- Tokura Y, Phadungsaksawasdi P, Ito T. Atopic dermatitis as Th2 disease revisited. J Cutan Immunol Allergy. 2018;1:158-164. doi:10.1002/cia2.12033
- Brunner PM, Guttman-Yassky E, Leung DY. The immunology of atopic dermatitis and its reversibility with broad-spectrum and targeted therapies. J Allergy Clin Immunol. 2017;139(suppl 4):S65-S76. doi:10.1016/j.jaci.2017.01.011
- Murphy PB, Atwater AR, Mueller M. Allergic Contact Dermatitis. StatPearls Publishing; 2021. https://www.ncbi.nlm.nih.gov/books/NBK532866/
- He D, Wu L, Kim HK, et al. IL-17 and IFN-gamma mediate the elicitation of contact hypersensitivity responses by different mechanisms and both are required for optimal responses [published online June 24, 2009]. J Immunol. 2009;183:1463-1470. doi:10.4049/jimmunol.0804108.
- Dhingra N, Shemer A, Correa da Rosa J, et al. Molecular profiling of contact dermatitis skin identifies allergen-dependent differences in immune response [published April 25, 2014]. J Allergy Clin Immunol. 2014;134:362-372. doi:10.1016/j.jaci.2014.03.009
- Owen JL, Vakharia PP, Silverberg JI. The role and diagnosis of allergic contact dermatitis in patients with atopic dermatitis. Am J Clin Dermatol. 2018;19:293-302. doi:10.1007/s40257-017-0340-7
- Uehara M, Sawai T. A longitudinal study of contact sensitivity in patients with atopic dermatitis. Arch Dermatol. 1989;125:366-368.
- Yüksel YT, Nørreslet LB, Thyssen JP. Allergic contact dermatitis in patients with atopic dermatitis. Curr Derm Rep. 2021;10:67-76.
- Gittler JK, Krueger JG, Guttman-Yassky E. Atopic dermatitis results in intrinsic barrier and immune abnormalities: implications for contact dermatitis [published online August 28, 2012]. J Allergy Clin Immunol. 2013;131:300-313. doi:10.1016/j.jaci.2012.06.048
- Drislane C, Irvine AD. The role of filaggrin in atopic dermatitis and allergic disease [published online October 14, 2019]. Ann Allergy Asthma Immunol. 2020;124:36-43. doi:10.1016/j.anai.2019.10.008
- Thyssen JP, McFadden JP, Kimber I. The multiple factors affectingthe association between atopic dermatitis and contact sensitization [published online December 26, 2013]. Allergy. 2014;69:28-36. doi:10.1111/all.12358
- Timmerman JG, Heederik D, Spee T, et al. Contact dermatitis in the construction industry: the role of filaggrin loss-of-function mutations [published online December 12, 2015]. Br J Dermatol. 2016;174:348-355. doi:10.1111/bjd.14215
- Edslev SM, Agner T, Andersen PS. Skin microbiome in atopic dermatitis. Acta Derm Venereol. 2020;100:adv00164. doi:
10.2340/00015555-3514 - Silverberg JI, Hou A, Warshaw EM, et al. Prevalence and trend of allergen sensitization in adults and children with atopic dermatitis referred for patch testing, North American Contact Dermatitis Group data, 2001-2016 [published online March 27, 2021]. J Allergy Clin Immunol Pract. 2021;9:2853-2866.e14. doi:10.1016/j.jaip.2021.03.028
- Pratt MD, Mufti A, Lipson J, et al. Patch test reactions to corticosteroids: retrospective analysis from the North American Contact Dermatitis Group 2007-2014. Dermatitis. 2017;28:58-63. doi:10.1097/DER.0000000000000251
- Xiong M, Peterson MY, Hylwa S. Allergic contact dermatitis from benzyl alcohol in hydrocortisone cream [published online January 14, 2022]. Contact Dermatitis. 2022;86:424-425. doi:10.1111/cod.14042
- Goldenberg A, Mousdicas N, Silverberg N, et al. Pediatric Contact Dermatitis Registry inaugural case data. Dermatitis. 2016;27:293-302. doi:10.1097/DER.0000000000000214
- Jacob SE, McGowan M, Silverberg NB, et al. Pediatric Contact Dermatitis Registry data on contact allergy in children with atopic dermatitis. JAMA Dermatol. 2017;153:765-770. doi:10.1001/jamadermatol.2016.6136
- Zug KA, McGinley-Smith D, Warshaw EM, et al. Contact allergy in children referred for patch testing: North American Contact Dermatitis Group data, 2001-2004. Arch Dermatol. 2008;144:1329-1336. doi:10.1001/archderm.144.10.1329
- Zug KA, Pham AK, Belsito DV, et al. Patch testing in children from 2005 to 2012: results from the North American contact dermatitis group. Dermatitis. 2014;25:345-355. doi:10.1097/DER.0000000000000083
- Hamann CR, Hamann D, Egeberg A, et al. Association between atopic dermatitis and contact sensitization: a systematic review and meta-analysis [published online April 6, 2017]. J Am Acad Dermatol. 2017;77:70-78. doi:10.1016/j.jaad.2017.02.001
- Simonsen AB, Johansen JD, Deleuran M, et al. Contact allergy in children with atopic dermatitis: a systematic review [published online June 12, 2017]. Br J Dermatol. 2017;177:395-405. doi:10.1111/bjd.15628
- Chen R, Raffi J, Murase JE. Tocopherol allergic dermatitis masquerading as lifelong atopic dermatitis. Dermatitis. 2020;31:E3-E4. doi:10.1097/DER.0000000000000543
- Tam I, Yu J. Pediatric contact dermatitis: what’s new. Curr Opin Pediatr. 2020;32:524-530. doi:10.1097/MOP.0000000000000919
- Cohen DE, Rao S, Brancaccio RR. Use of the North American Contact Dermatitis Group Standard 65-allergen series alone in the evaluation of allergic contact dermatitis: a series of 794 patients. Dermatitis. 2008;19:137-141.
- Schalock PC, Dunnick CA, Nedorost S, et al. American Contact Dermatitis Society Core Allergen Series: 2020 update. Dermatitis. 2020;31:279-282. doi:10.1097/DER.0000000000000621
- Yu J, Atwater AR, Brod B, et al. Pediatric baseline patch test series: Pediatric Contact Dermatitis Workgroup. Dermatitis. 2018;29:206-212. doi:10.1097/DER.0000000000000385
- Bußmann C, Novak N. Systemic therapy of atopic dermatitis. Allergol Select. 2017;1:1-8. doi:10.5414/ALX01285E
- Sung CT, McGowan MA, Machler BC, et al. Systemic treatments for allergic contact dermatitis. Dermatitis. 2019;30:46-53. doi:10.1097/DER.0000000000000435
- Johnson H, Adler BL, Yu J. Dupilumab for allergic contact dermatitis: an overview of its use and impact on patch testing. Cutis. 2022;109:265-267, E4-E5. doi:10.12788/cutis.0519
- Todberg T, Zachariae C, Krustrup D, et al. The effect of treatment with anti-interleukin-17 in patients with allergic contact dermatitis. Contact Dermatitis. 2018;78:431-432. doi:10.1111/cod.12988
- Ungar B, Pavel AB, Li R, et al. Phase 2 randomized, double-blind study of IL-17 targeting with secukinumab in atopic dermatitis [published online May 16, 2020]. J Allergy Clin Immunol. 2021;147:394-397. doi:10.1016/j.jaci.2020.04.055
- Perche PO, Cook MK, Feldman SR. Abrocitinib: a new FDA-approved drug for moderate-to-severe atopic dermatitis [published online May 19, 2022]. Ann Pharmacother. doi:10.1177/10600280221096713
Atopic dermatitis (AD) and allergic contact dermatitis (ACD) are 2 common inflammatory skin conditions that may have similar clinical presentations. Historically, it was thought that these conditions could not be diagnosed simultaneously due to their differing immune mechanisms; however, this belief has been challenged by recent evidence suggesting a more nuanced relationship between the 2 disease processes. In this review, we examine the complex interplay between AD and ACD and explain how shifts in conventional understanding of the 2 conditions shaped our evolving recognition of their ability to coexist.
Epidemiology of AD and ACD
Atopic dermatitis is the most common inflammatory skin disease in children and adolescents, with an estimated prevalence reaching 21%.1 In 60% of cases, onset of AD will occur within the first year of life, and 90% of cases begin within the first 5 years.2 Resolution may occur by adulthood; however, AD may continue to impact up to 8% to 9% of adults, with an increased prevalence in those older than 75 years.1 This may represent an underestimation of the burden of adult AD; one systematic review of 17 studies found that the pooled proportion of adult-onset AD was greater than 25%.3
In contrast, ACD previously was assumed to be a disease that more commonly impacted adults and only rarely children, primarily due to an early misconception that children were not frequently exposed to contact allergens and their immune systems were too immature to react to them even if exposed.4,5 However, it is now known that children do have risk factors for development of ACD, including a thinner stratum corneum and potentially a more absorbent skin surface.4 In addition, a 2022 study by the North American Contact Dermatitis Group (NACDG) found similar rates of ACD in children (n=1871) and adults (n=41,699) referred for patch testing (55.2% and 57.3%, respectively) as well as similar rates of having at least 1 relevant positive patch test (49.2% and 52.2%).6
In opposition to traditional beliefs, these findings highlight that AD and ACD can occur across age groups.
Immune Mechanism
The pathogenesis of AD represents a multifactorial process involving the immune system, cutaneous flora, genetic predisposition, and surrounding environment. Immunologically, acute AD is driven by a predominantly TH2 helper T-cell response with high levels of IL-4, IL-5, and IL-137; TH22, TH17, and TH1 also have been implicated.8 Notably, TH17 is found in high levels during the acute eczema phase, while TH1 and TH22are associated with the chronic phase.7
The pathophysiology of ACD is not completely understood. The classic paradigm involves 2 phases: sensitization and elicitation. Sensitization involves antigen-presenting cells that take up allergens absorbed by the skin to present them in regional lymph nodes where antigen-specific T lymphocytes are generated. Elicitation occurs upon re-exposure to the allergen, at which time the primed T lymphocytes are recruited to the skin, causing inflammation.9 Allergic contact dermatitis initially was thought to be driven by TH1 cytokines and IL-17 but now is understood to be more complex.10 Studies have revealed immune polarization of contact allergens, demonstrating that nickel primarily induces a TH1/TH17 response, whereas fragrance and rubber accelerators skew to TH2; TH9 and TH22 also may be involved depending on the causative allergen.11,12
Of note, the immunologic differences between AD and ACD led early investigators to believe that patients with AD were relatively protected from ACD.13 However, as previously described, there are several overlapping cytokines between AD and ACD. Furthermore, research has revealed that risk of contact sensitization might be increased in the chronic eczema phase due to the shared TH1 pathway.14 Barrier-disrupted skin (such as that in AD) also may increase the cytokine response and the density of antigen-presenting cells, leading to a proallergic state.15 This suggests that the immunologic pathways of AD and ACD are more intertwined than was previously understood.
Underlying Risk Factors
Skin barrier dysfunction is a key step in the pathogenesis of AD. Patients with AD commonly have loss-of-function mutations in the filaggrin gene, a protein that is key to the function of the stratum corneum. Loss of this protein may not only impact the immune response as previously noted but also may lead to increased transepidermal water loss and bacterial colonization.16 Interestingly, a 2014 review examined how this mutation could lead to an increased risk of sensitization to bivalent metal ions via an impaired chelating ability of the skin.17 Furthermore, a 2016 study conducted in Dutch construction workers revealed an increased risk for contact dermatitis (irritant and allergic) for those with a loss-of-function filaggrin mutation.18
Importantly, this same mutation may explain why patients with AD tend to have increased skin colonization by Staphylococcus aureus. The abundance of S aureus and the relative decrease in the diversity of other microorganisms on the skin may be associated with increased AD severity.19 Likewise, S aureus may play a role in the pathogenesis of ACD via production of its exotoxin directed at the T-cell receptor V beta 17 region. In particular, this receptor has been associated with nickel sensitization.17
Another risk factor to consider is increased exposure to contact sensitizers when treating AD. For instance, management often includes use of over-the-counter emollients, natural or botanical remedies with purported benefits for AD, cleansers, and detergents. However, these products can contain some of the most prevalent contact allergens seen in those with AD, including methyl-isothiazolinone, formaldehyde releasers, and fragrance.20 Topical corticosteroids also are frequently used, and ACD to steroid molecules can occur, particularly to tixocortol-21-pivalate (a marker for class A corticosteroids) and budesonide (a marker for class B corticosteroids).21 Other allergens (eg, benzyl alcohol, propylene glycol) also may be found as inactive ingredients of topical corticosteroids.22 These exposures may place AD patients at risk for ACD.
The Coexistence of AD and ACD
Given the overlapping epidemiology, immunology, and potentially increased risk for the development of ACD in patients with AD, it would be reasonable to assume that the 2 diagnoses could coexist; however, is there clinical data to support this idea? Based on recent database reviews, the answer appears to be yes.20,23-26 An analysis from the Pediatric Contact Dermatitis Registry revealed that 30% of 1142 pediatric patch test cases analyzed were diagnosed as AD and ACD simultaneously.24 The NACDG found similar results in its 2021 review, as 29.5% of children (n=1648) and 20.7% of adults (n=36,834) had a concurrent diagnosis of AD and ACD.20 Notably, older results from these databases also demonstrated an association between the 2 conditions.23,25,26
It remains unclear whether the prevalence of ACD is higher in those with or without AD. A comprehensive systematic review conducted in 2017 examined this topic through analysis of 74 studies. The results demonstrated a similar prevalence of contact sensitization in individuals with and without AD.27 Another systematic review of 31 studies conducted in 2017 found a higher prevalence for ACD in children without AD; however, the authors noted that the included studies were too variable (eg, size, design, allergens tested) to draw definitive conclusions.28
Even though there is no clear overall increased risk for ACD in patients with AD, research has suggested that certain allergens may be more prevalent in the setting of AD. An NACDG study found that adults with AD had increased odds of reacting to 10 of the top 25 NACDG screening allergens compared to those without AD.20 Other studies have found that AD patients may be more likely to become sensitized to certain allergens, such as fragrance and lanolin.14
Considerations for Management
Diagnosis of ACD in patients with AD can be challenging because these conditions may present similarly with chronic, pruritic, inflammatory patches and plaques. Chronic ACD may be misdiagnosed as AD if patch testing is not performed.29 Given the prevalence of ACD in the setting of AD, there should be a low threshold to pursue patch testing, especially when dermatitis is recalcitrant to standard therapies or presents in an atypical distribution (ie, perioral, predominantly head/neck, hand and foot, isolated eyelid involvement, buttocks).4,30 Various allergen series are available for patch testing adults and children including the NACDG Standard Series, American Contact Dermatitis Society Core Allergen Series, or the Pediatric Baseline Series.31-33
If potentially relevant allergens are uncovered by patch testing, patients should be counseled on avoidance strategies. However, allergen avoidance may not always lead to complete symptom resolution, especially if AD is present concomitantly with ACD. Therefore, use of topical or systemic therapies still may be required. Topical corticosteroids can be used when dermatitis is acute and localized. Systemic corticosteroids are utilized for both diagnoses when cases are more severe or extensive, but their adverse-effect profile limits long-term use. Other systemic treatments, including conventional agents (ie, azathioprine, cyclosporine, methotrexate, mycophenolate mofetil), biologics, and small molecule inhibitors also may be considered for severe cases.34,35 Dupilumab, a monoclonal antibody targeting IL-4/IL-13, is approved for use in moderate to severe AD in patients 6 months and older. Recent evidence has suggested that dupilumab also may be an effective off-label treatment choice for ACD when allergen avoidance alone is insufficient.36 Studies have been conducted on secukinumab, a monoclonal antibody against IL-17; however, it has not been shown to be effective in either AD or ACD.37,38 This indicates that targeted biologics may not always be successful in treating these diagnoses, likely due to their complex immune pathways. Finally, there is an emerging role for JAK inhibitors. Three are approved for AD: topical ruxolitinib, oral abrocitinib, and oral upadacitinib.39 Further investigation is needed to determine the efficacy of JAK inhibitors in ACD.
Final Interpretation
Evolving evidence shows that AD and ACD can occur at the same time despite the historical perspective that their immune pathways were too polarized for this to happen. Atopic dermatitis may be an important risk factor for subsequent development of ACD. Management should include a low threshold to perform patch testing, while pharmacotherapies utilized in the treatment of both conditions should be considered.
Atopic dermatitis (AD) and allergic contact dermatitis (ACD) are 2 common inflammatory skin conditions that may have similar clinical presentations. Historically, it was thought that these conditions could not be diagnosed simultaneously due to their differing immune mechanisms; however, this belief has been challenged by recent evidence suggesting a more nuanced relationship between the 2 disease processes. In this review, we examine the complex interplay between AD and ACD and explain how shifts in conventional understanding of the 2 conditions shaped our evolving recognition of their ability to coexist.
Epidemiology of AD and ACD
Atopic dermatitis is the most common inflammatory skin disease in children and adolescents, with an estimated prevalence reaching 21%.1 In 60% of cases, onset of AD will occur within the first year of life, and 90% of cases begin within the first 5 years.2 Resolution may occur by adulthood; however, AD may continue to impact up to 8% to 9% of adults, with an increased prevalence in those older than 75 years.1 This may represent an underestimation of the burden of adult AD; one systematic review of 17 studies found that the pooled proportion of adult-onset AD was greater than 25%.3
In contrast, ACD previously was assumed to be a disease that more commonly impacted adults and only rarely children, primarily due to an early misconception that children were not frequently exposed to contact allergens and their immune systems were too immature to react to them even if exposed.4,5 However, it is now known that children do have risk factors for development of ACD, including a thinner stratum corneum and potentially a more absorbent skin surface.4 In addition, a 2022 study by the North American Contact Dermatitis Group (NACDG) found similar rates of ACD in children (n=1871) and adults (n=41,699) referred for patch testing (55.2% and 57.3%, respectively) as well as similar rates of having at least 1 relevant positive patch test (49.2% and 52.2%).6
In opposition to traditional beliefs, these findings highlight that AD and ACD can occur across age groups.
Immune Mechanism
The pathogenesis of AD represents a multifactorial process involving the immune system, cutaneous flora, genetic predisposition, and surrounding environment. Immunologically, acute AD is driven by a predominantly TH2 helper T-cell response with high levels of IL-4, IL-5, and IL-137; TH22, TH17, and TH1 also have been implicated.8 Notably, TH17 is found in high levels during the acute eczema phase, while TH1 and TH22are associated with the chronic phase.7
The pathophysiology of ACD is not completely understood. The classic paradigm involves 2 phases: sensitization and elicitation. Sensitization involves antigen-presenting cells that take up allergens absorbed by the skin to present them in regional lymph nodes where antigen-specific T lymphocytes are generated. Elicitation occurs upon re-exposure to the allergen, at which time the primed T lymphocytes are recruited to the skin, causing inflammation.9 Allergic contact dermatitis initially was thought to be driven by TH1 cytokines and IL-17 but now is understood to be more complex.10 Studies have revealed immune polarization of contact allergens, demonstrating that nickel primarily induces a TH1/TH17 response, whereas fragrance and rubber accelerators skew to TH2; TH9 and TH22 also may be involved depending on the causative allergen.11,12
Of note, the immunologic differences between AD and ACD led early investigators to believe that patients with AD were relatively protected from ACD.13 However, as previously described, there are several overlapping cytokines between AD and ACD. Furthermore, research has revealed that risk of contact sensitization might be increased in the chronic eczema phase due to the shared TH1 pathway.14 Barrier-disrupted skin (such as that in AD) also may increase the cytokine response and the density of antigen-presenting cells, leading to a proallergic state.15 This suggests that the immunologic pathways of AD and ACD are more intertwined than was previously understood.
Underlying Risk Factors
Skin barrier dysfunction is a key step in the pathogenesis of AD. Patients with AD commonly have loss-of-function mutations in the filaggrin gene, a protein that is key to the function of the stratum corneum. Loss of this protein may not only impact the immune response as previously noted but also may lead to increased transepidermal water loss and bacterial colonization.16 Interestingly, a 2014 review examined how this mutation could lead to an increased risk of sensitization to bivalent metal ions via an impaired chelating ability of the skin.17 Furthermore, a 2016 study conducted in Dutch construction workers revealed an increased risk for contact dermatitis (irritant and allergic) for those with a loss-of-function filaggrin mutation.18
Importantly, this same mutation may explain why patients with AD tend to have increased skin colonization by Staphylococcus aureus. The abundance of S aureus and the relative decrease in the diversity of other microorganisms on the skin may be associated with increased AD severity.19 Likewise, S aureus may play a role in the pathogenesis of ACD via production of its exotoxin directed at the T-cell receptor V beta 17 region. In particular, this receptor has been associated with nickel sensitization.17
Another risk factor to consider is increased exposure to contact sensitizers when treating AD. For instance, management often includes use of over-the-counter emollients, natural or botanical remedies with purported benefits for AD, cleansers, and detergents. However, these products can contain some of the most prevalent contact allergens seen in those with AD, including methyl-isothiazolinone, formaldehyde releasers, and fragrance.20 Topical corticosteroids also are frequently used, and ACD to steroid molecules can occur, particularly to tixocortol-21-pivalate (a marker for class A corticosteroids) and budesonide (a marker for class B corticosteroids).21 Other allergens (eg, benzyl alcohol, propylene glycol) also may be found as inactive ingredients of topical corticosteroids.22 These exposures may place AD patients at risk for ACD.
The Coexistence of AD and ACD
Given the overlapping epidemiology, immunology, and potentially increased risk for the development of ACD in patients with AD, it would be reasonable to assume that the 2 diagnoses could coexist; however, is there clinical data to support this idea? Based on recent database reviews, the answer appears to be yes.20,23-26 An analysis from the Pediatric Contact Dermatitis Registry revealed that 30% of 1142 pediatric patch test cases analyzed were diagnosed as AD and ACD simultaneously.24 The NACDG found similar results in its 2021 review, as 29.5% of children (n=1648) and 20.7% of adults (n=36,834) had a concurrent diagnosis of AD and ACD.20 Notably, older results from these databases also demonstrated an association between the 2 conditions.23,25,26
It remains unclear whether the prevalence of ACD is higher in those with or without AD. A comprehensive systematic review conducted in 2017 examined this topic through analysis of 74 studies. The results demonstrated a similar prevalence of contact sensitization in individuals with and without AD.27 Another systematic review of 31 studies conducted in 2017 found a higher prevalence for ACD in children without AD; however, the authors noted that the included studies were too variable (eg, size, design, allergens tested) to draw definitive conclusions.28
Even though there is no clear overall increased risk for ACD in patients with AD, research has suggested that certain allergens may be more prevalent in the setting of AD. An NACDG study found that adults with AD had increased odds of reacting to 10 of the top 25 NACDG screening allergens compared to those without AD.20 Other studies have found that AD patients may be more likely to become sensitized to certain allergens, such as fragrance and lanolin.14
Considerations for Management
Diagnosis of ACD in patients with AD can be challenging because these conditions may present similarly with chronic, pruritic, inflammatory patches and plaques. Chronic ACD may be misdiagnosed as AD if patch testing is not performed.29 Given the prevalence of ACD in the setting of AD, there should be a low threshold to pursue patch testing, especially when dermatitis is recalcitrant to standard therapies or presents in an atypical distribution (ie, perioral, predominantly head/neck, hand and foot, isolated eyelid involvement, buttocks).4,30 Various allergen series are available for patch testing adults and children including the NACDG Standard Series, American Contact Dermatitis Society Core Allergen Series, or the Pediatric Baseline Series.31-33
If potentially relevant allergens are uncovered by patch testing, patients should be counseled on avoidance strategies. However, allergen avoidance may not always lead to complete symptom resolution, especially if AD is present concomitantly with ACD. Therefore, use of topical or systemic therapies still may be required. Topical corticosteroids can be used when dermatitis is acute and localized. Systemic corticosteroids are utilized for both diagnoses when cases are more severe or extensive, but their adverse-effect profile limits long-term use. Other systemic treatments, including conventional agents (ie, azathioprine, cyclosporine, methotrexate, mycophenolate mofetil), biologics, and small molecule inhibitors also may be considered for severe cases.34,35 Dupilumab, a monoclonal antibody targeting IL-4/IL-13, is approved for use in moderate to severe AD in patients 6 months and older. Recent evidence has suggested that dupilumab also may be an effective off-label treatment choice for ACD when allergen avoidance alone is insufficient.36 Studies have been conducted on secukinumab, a monoclonal antibody against IL-17; however, it has not been shown to be effective in either AD or ACD.37,38 This indicates that targeted biologics may not always be successful in treating these diagnoses, likely due to their complex immune pathways. Finally, there is an emerging role for JAK inhibitors. Three are approved for AD: topical ruxolitinib, oral abrocitinib, and oral upadacitinib.39 Further investigation is needed to determine the efficacy of JAK inhibitors in ACD.
Final Interpretation
Evolving evidence shows that AD and ACD can occur at the same time despite the historical perspective that their immune pathways were too polarized for this to happen. Atopic dermatitis may be an important risk factor for subsequent development of ACD. Management should include a low threshold to perform patch testing, while pharmacotherapies utilized in the treatment of both conditions should be considered.
- Chan LN, Magyari A, Ye M, et al. The epidemiology of atopic dermatitis in older adults: a population-based study in the United Kingdom. PLoS One. 2021;16:E0258219. doi:10.1371/journal.pone.0258219
- Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: section 1. diagnosis and assessment of atopic dermatitis [published online November 27, 2013]. J Am Acad Dermatol. 2014;70:338-351. doi:10.1016/j.jaad.2013.10.010
- Lee HH, Patel KR, Singam V, et al. A systematic review and meta-analysis of the prevalence and phenotype of adult-onset atopic dermatitis [published online June 2, 2018]. J Am Acad Dermatol. 2019;80:1526-1532.e7. doi:10.1016/j.jaad.2018.05.1241
- Borok J, Matiz C, Goldenberg A, et al. Contact dermatitis in atopic dermatitis children—past, present, and future. Clin Rev Allergy Immunol. 2019;56:86-98. doi:10.1007/s12016-018-8711-2
- Goldenberg A, Silverberg N, Silverberg JI, et al. Pediatric allergic contact dermatitis: lessons for better care. J Allergy Clin Immunol Pract. 2015;3:661-667; quiz 668. doi:10.1016/j.jaip.2015.02.007
- Silverberg JI, Hou A, Warshaw EM, et al. Age-related differences in patch testing results among children: analysis of North American Contact Dermatitis Group data, 2001-2018 [published online July 24, 2021]. J Am Acad Dermatol. 2022;86:818-826. doi:10.1016/j.jaad.2021.07.030
- Tokura Y, Phadungsaksawasdi P, Ito T. Atopic dermatitis as Th2 disease revisited. J Cutan Immunol Allergy. 2018;1:158-164. doi:10.1002/cia2.12033
- Brunner PM, Guttman-Yassky E, Leung DY. The immunology of atopic dermatitis and its reversibility with broad-spectrum and targeted therapies. J Allergy Clin Immunol. 2017;139(suppl 4):S65-S76. doi:10.1016/j.jaci.2017.01.011
- Murphy PB, Atwater AR, Mueller M. Allergic Contact Dermatitis. StatPearls Publishing; 2021. https://www.ncbi.nlm.nih.gov/books/NBK532866/
- He D, Wu L, Kim HK, et al. IL-17 and IFN-gamma mediate the elicitation of contact hypersensitivity responses by different mechanisms and both are required for optimal responses [published online June 24, 2009]. J Immunol. 2009;183:1463-1470. doi:10.4049/jimmunol.0804108.
- Dhingra N, Shemer A, Correa da Rosa J, et al. Molecular profiling of contact dermatitis skin identifies allergen-dependent differences in immune response [published April 25, 2014]. J Allergy Clin Immunol. 2014;134:362-372. doi:10.1016/j.jaci.2014.03.009
- Owen JL, Vakharia PP, Silverberg JI. The role and diagnosis of allergic contact dermatitis in patients with atopic dermatitis. Am J Clin Dermatol. 2018;19:293-302. doi:10.1007/s40257-017-0340-7
- Uehara M, Sawai T. A longitudinal study of contact sensitivity in patients with atopic dermatitis. Arch Dermatol. 1989;125:366-368.
- Yüksel YT, Nørreslet LB, Thyssen JP. Allergic contact dermatitis in patients with atopic dermatitis. Curr Derm Rep. 2021;10:67-76.
- Gittler JK, Krueger JG, Guttman-Yassky E. Atopic dermatitis results in intrinsic barrier and immune abnormalities: implications for contact dermatitis [published online August 28, 2012]. J Allergy Clin Immunol. 2013;131:300-313. doi:10.1016/j.jaci.2012.06.048
- Drislane C, Irvine AD. The role of filaggrin in atopic dermatitis and allergic disease [published online October 14, 2019]. Ann Allergy Asthma Immunol. 2020;124:36-43. doi:10.1016/j.anai.2019.10.008
- Thyssen JP, McFadden JP, Kimber I. The multiple factors affectingthe association between atopic dermatitis and contact sensitization [published online December 26, 2013]. Allergy. 2014;69:28-36. doi:10.1111/all.12358
- Timmerman JG, Heederik D, Spee T, et al. Contact dermatitis in the construction industry: the role of filaggrin loss-of-function mutations [published online December 12, 2015]. Br J Dermatol. 2016;174:348-355. doi:10.1111/bjd.14215
- Edslev SM, Agner T, Andersen PS. Skin microbiome in atopic dermatitis. Acta Derm Venereol. 2020;100:adv00164. doi:
10.2340/00015555-3514 - Silverberg JI, Hou A, Warshaw EM, et al. Prevalence and trend of allergen sensitization in adults and children with atopic dermatitis referred for patch testing, North American Contact Dermatitis Group data, 2001-2016 [published online March 27, 2021]. J Allergy Clin Immunol Pract. 2021;9:2853-2866.e14. doi:10.1016/j.jaip.2021.03.028
- Pratt MD, Mufti A, Lipson J, et al. Patch test reactions to corticosteroids: retrospective analysis from the North American Contact Dermatitis Group 2007-2014. Dermatitis. 2017;28:58-63. doi:10.1097/DER.0000000000000251
- Xiong M, Peterson MY, Hylwa S. Allergic contact dermatitis from benzyl alcohol in hydrocortisone cream [published online January 14, 2022]. Contact Dermatitis. 2022;86:424-425. doi:10.1111/cod.14042
- Goldenberg A, Mousdicas N, Silverberg N, et al. Pediatric Contact Dermatitis Registry inaugural case data. Dermatitis. 2016;27:293-302. doi:10.1097/DER.0000000000000214
- Jacob SE, McGowan M, Silverberg NB, et al. Pediatric Contact Dermatitis Registry data on contact allergy in children with atopic dermatitis. JAMA Dermatol. 2017;153:765-770. doi:10.1001/jamadermatol.2016.6136
- Zug KA, McGinley-Smith D, Warshaw EM, et al. Contact allergy in children referred for patch testing: North American Contact Dermatitis Group data, 2001-2004. Arch Dermatol. 2008;144:1329-1336. doi:10.1001/archderm.144.10.1329
- Zug KA, Pham AK, Belsito DV, et al. Patch testing in children from 2005 to 2012: results from the North American contact dermatitis group. Dermatitis. 2014;25:345-355. doi:10.1097/DER.0000000000000083
- Hamann CR, Hamann D, Egeberg A, et al. Association between atopic dermatitis and contact sensitization: a systematic review and meta-analysis [published online April 6, 2017]. J Am Acad Dermatol. 2017;77:70-78. doi:10.1016/j.jaad.2017.02.001
- Simonsen AB, Johansen JD, Deleuran M, et al. Contact allergy in children with atopic dermatitis: a systematic review [published online June 12, 2017]. Br J Dermatol. 2017;177:395-405. doi:10.1111/bjd.15628
- Chen R, Raffi J, Murase JE. Tocopherol allergic dermatitis masquerading as lifelong atopic dermatitis. Dermatitis. 2020;31:E3-E4. doi:10.1097/DER.0000000000000543
- Tam I, Yu J. Pediatric contact dermatitis: what’s new. Curr Opin Pediatr. 2020;32:524-530. doi:10.1097/MOP.0000000000000919
- Cohen DE, Rao S, Brancaccio RR. Use of the North American Contact Dermatitis Group Standard 65-allergen series alone in the evaluation of allergic contact dermatitis: a series of 794 patients. Dermatitis. 2008;19:137-141.
- Schalock PC, Dunnick CA, Nedorost S, et al. American Contact Dermatitis Society Core Allergen Series: 2020 update. Dermatitis. 2020;31:279-282. doi:10.1097/DER.0000000000000621
- Yu J, Atwater AR, Brod B, et al. Pediatric baseline patch test series: Pediatric Contact Dermatitis Workgroup. Dermatitis. 2018;29:206-212. doi:10.1097/DER.0000000000000385
- Bußmann C, Novak N. Systemic therapy of atopic dermatitis. Allergol Select. 2017;1:1-8. doi:10.5414/ALX01285E
- Sung CT, McGowan MA, Machler BC, et al. Systemic treatments for allergic contact dermatitis. Dermatitis. 2019;30:46-53. doi:10.1097/DER.0000000000000435
- Johnson H, Adler BL, Yu J. Dupilumab for allergic contact dermatitis: an overview of its use and impact on patch testing. Cutis. 2022;109:265-267, E4-E5. doi:10.12788/cutis.0519
- Todberg T, Zachariae C, Krustrup D, et al. The effect of treatment with anti-interleukin-17 in patients with allergic contact dermatitis. Contact Dermatitis. 2018;78:431-432. doi:10.1111/cod.12988
- Ungar B, Pavel AB, Li R, et al. Phase 2 randomized, double-blind study of IL-17 targeting with secukinumab in atopic dermatitis [published online May 16, 2020]. J Allergy Clin Immunol. 2021;147:394-397. doi:10.1016/j.jaci.2020.04.055
- Perche PO, Cook MK, Feldman SR. Abrocitinib: a new FDA-approved drug for moderate-to-severe atopic dermatitis [published online May 19, 2022]. Ann Pharmacother. doi:10.1177/10600280221096713
- Chan LN, Magyari A, Ye M, et al. The epidemiology of atopic dermatitis in older adults: a population-based study in the United Kingdom. PLoS One. 2021;16:E0258219. doi:10.1371/journal.pone.0258219
- Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: section 1. diagnosis and assessment of atopic dermatitis [published online November 27, 2013]. J Am Acad Dermatol. 2014;70:338-351. doi:10.1016/j.jaad.2013.10.010
- Lee HH, Patel KR, Singam V, et al. A systematic review and meta-analysis of the prevalence and phenotype of adult-onset atopic dermatitis [published online June 2, 2018]. J Am Acad Dermatol. 2019;80:1526-1532.e7. doi:10.1016/j.jaad.2018.05.1241
- Borok J, Matiz C, Goldenberg A, et al. Contact dermatitis in atopic dermatitis children—past, present, and future. Clin Rev Allergy Immunol. 2019;56:86-98. doi:10.1007/s12016-018-8711-2
- Goldenberg A, Silverberg N, Silverberg JI, et al. Pediatric allergic contact dermatitis: lessons for better care. J Allergy Clin Immunol Pract. 2015;3:661-667; quiz 668. doi:10.1016/j.jaip.2015.02.007
- Silverberg JI, Hou A, Warshaw EM, et al. Age-related differences in patch testing results among children: analysis of North American Contact Dermatitis Group data, 2001-2018 [published online July 24, 2021]. J Am Acad Dermatol. 2022;86:818-826. doi:10.1016/j.jaad.2021.07.030
- Tokura Y, Phadungsaksawasdi P, Ito T. Atopic dermatitis as Th2 disease revisited. J Cutan Immunol Allergy. 2018;1:158-164. doi:10.1002/cia2.12033
- Brunner PM, Guttman-Yassky E, Leung DY. The immunology of atopic dermatitis and its reversibility with broad-spectrum and targeted therapies. J Allergy Clin Immunol. 2017;139(suppl 4):S65-S76. doi:10.1016/j.jaci.2017.01.011
- Murphy PB, Atwater AR, Mueller M. Allergic Contact Dermatitis. StatPearls Publishing; 2021. https://www.ncbi.nlm.nih.gov/books/NBK532866/
- He D, Wu L, Kim HK, et al. IL-17 and IFN-gamma mediate the elicitation of contact hypersensitivity responses by different mechanisms and both are required for optimal responses [published online June 24, 2009]. J Immunol. 2009;183:1463-1470. doi:10.4049/jimmunol.0804108.
- Dhingra N, Shemer A, Correa da Rosa J, et al. Molecular profiling of contact dermatitis skin identifies allergen-dependent differences in immune response [published April 25, 2014]. J Allergy Clin Immunol. 2014;134:362-372. doi:10.1016/j.jaci.2014.03.009
- Owen JL, Vakharia PP, Silverberg JI. The role and diagnosis of allergic contact dermatitis in patients with atopic dermatitis. Am J Clin Dermatol. 2018;19:293-302. doi:10.1007/s40257-017-0340-7
- Uehara M, Sawai T. A longitudinal study of contact sensitivity in patients with atopic dermatitis. Arch Dermatol. 1989;125:366-368.
- Yüksel YT, Nørreslet LB, Thyssen JP. Allergic contact dermatitis in patients with atopic dermatitis. Curr Derm Rep. 2021;10:67-76.
- Gittler JK, Krueger JG, Guttman-Yassky E. Atopic dermatitis results in intrinsic barrier and immune abnormalities: implications for contact dermatitis [published online August 28, 2012]. J Allergy Clin Immunol. 2013;131:300-313. doi:10.1016/j.jaci.2012.06.048
- Drislane C, Irvine AD. The role of filaggrin in atopic dermatitis and allergic disease [published online October 14, 2019]. Ann Allergy Asthma Immunol. 2020;124:36-43. doi:10.1016/j.anai.2019.10.008
- Thyssen JP, McFadden JP, Kimber I. The multiple factors affectingthe association between atopic dermatitis and contact sensitization [published online December 26, 2013]. Allergy. 2014;69:28-36. doi:10.1111/all.12358
- Timmerman JG, Heederik D, Spee T, et al. Contact dermatitis in the construction industry: the role of filaggrin loss-of-function mutations [published online December 12, 2015]. Br J Dermatol. 2016;174:348-355. doi:10.1111/bjd.14215
- Edslev SM, Agner T, Andersen PS. Skin microbiome in atopic dermatitis. Acta Derm Venereol. 2020;100:adv00164. doi:
10.2340/00015555-3514 - Silverberg JI, Hou A, Warshaw EM, et al. Prevalence and trend of allergen sensitization in adults and children with atopic dermatitis referred for patch testing, North American Contact Dermatitis Group data, 2001-2016 [published online March 27, 2021]. J Allergy Clin Immunol Pract. 2021;9:2853-2866.e14. doi:10.1016/j.jaip.2021.03.028
- Pratt MD, Mufti A, Lipson J, et al. Patch test reactions to corticosteroids: retrospective analysis from the North American Contact Dermatitis Group 2007-2014. Dermatitis. 2017;28:58-63. doi:10.1097/DER.0000000000000251
- Xiong M, Peterson MY, Hylwa S. Allergic contact dermatitis from benzyl alcohol in hydrocortisone cream [published online January 14, 2022]. Contact Dermatitis. 2022;86:424-425. doi:10.1111/cod.14042
- Goldenberg A, Mousdicas N, Silverberg N, et al. Pediatric Contact Dermatitis Registry inaugural case data. Dermatitis. 2016;27:293-302. doi:10.1097/DER.0000000000000214
- Jacob SE, McGowan M, Silverberg NB, et al. Pediatric Contact Dermatitis Registry data on contact allergy in children with atopic dermatitis. JAMA Dermatol. 2017;153:765-770. doi:10.1001/jamadermatol.2016.6136
- Zug KA, McGinley-Smith D, Warshaw EM, et al. Contact allergy in children referred for patch testing: North American Contact Dermatitis Group data, 2001-2004. Arch Dermatol. 2008;144:1329-1336. doi:10.1001/archderm.144.10.1329
- Zug KA, Pham AK, Belsito DV, et al. Patch testing in children from 2005 to 2012: results from the North American contact dermatitis group. Dermatitis. 2014;25:345-355. doi:10.1097/DER.0000000000000083
- Hamann CR, Hamann D, Egeberg A, et al. Association between atopic dermatitis and contact sensitization: a systematic review and meta-analysis [published online April 6, 2017]. J Am Acad Dermatol. 2017;77:70-78. doi:10.1016/j.jaad.2017.02.001
- Simonsen AB, Johansen JD, Deleuran M, et al. Contact allergy in children with atopic dermatitis: a systematic review [published online June 12, 2017]. Br J Dermatol. 2017;177:395-405. doi:10.1111/bjd.15628
- Chen R, Raffi J, Murase JE. Tocopherol allergic dermatitis masquerading as lifelong atopic dermatitis. Dermatitis. 2020;31:E3-E4. doi:10.1097/DER.0000000000000543
- Tam I, Yu J. Pediatric contact dermatitis: what’s new. Curr Opin Pediatr. 2020;32:524-530. doi:10.1097/MOP.0000000000000919
- Cohen DE, Rao S, Brancaccio RR. Use of the North American Contact Dermatitis Group Standard 65-allergen series alone in the evaluation of allergic contact dermatitis: a series of 794 patients. Dermatitis. 2008;19:137-141.
- Schalock PC, Dunnick CA, Nedorost S, et al. American Contact Dermatitis Society Core Allergen Series: 2020 update. Dermatitis. 2020;31:279-282. doi:10.1097/DER.0000000000000621
- Yu J, Atwater AR, Brod B, et al. Pediatric baseline patch test series: Pediatric Contact Dermatitis Workgroup. Dermatitis. 2018;29:206-212. doi:10.1097/DER.0000000000000385
- Bußmann C, Novak N. Systemic therapy of atopic dermatitis. Allergol Select. 2017;1:1-8. doi:10.5414/ALX01285E
- Sung CT, McGowan MA, Machler BC, et al. Systemic treatments for allergic contact dermatitis. Dermatitis. 2019;30:46-53. doi:10.1097/DER.0000000000000435
- Johnson H, Adler BL, Yu J. Dupilumab for allergic contact dermatitis: an overview of its use and impact on patch testing. Cutis. 2022;109:265-267, E4-E5. doi:10.12788/cutis.0519
- Todberg T, Zachariae C, Krustrup D, et al. The effect of treatment with anti-interleukin-17 in patients with allergic contact dermatitis. Contact Dermatitis. 2018;78:431-432. doi:10.1111/cod.12988
- Ungar B, Pavel AB, Li R, et al. Phase 2 randomized, double-blind study of IL-17 targeting with secukinumab in atopic dermatitis [published online May 16, 2020]. J Allergy Clin Immunol. 2021;147:394-397. doi:10.1016/j.jaci.2020.04.055
- Perche PO, Cook MK, Feldman SR. Abrocitinib: a new FDA-approved drug for moderate-to-severe atopic dermatitis [published online May 19, 2022]. Ann Pharmacother. doi:10.1177/10600280221096713
Practice Points
- Although it previously was thought that atopic dermatitis (AD) and allergic contact dermatitis (ACD) could not coexist due to their polarized immune pathways, current evidence suggests otherwise.
- When both diagnoses are suspected, patch testing should be considered as well as therapeutic strategies that can treat both AD and ACD simultaneously.
Vismodegib for Basal Cell Carcinoma and Beyond: What Dermatologists Need to Know
Basal cell carcinomas (BCCs) are considered the most common cutaneous cancers. Approximately 80% of nonmelanoma skin cancers are BCCs.1,2 Surgical management is the gold standard for early-stage and localized BCCs; it may include simple excision vs Mohs micrographic surgery.3,4 However, if left untreated, these lesions can progress to an advanced stage (locally advanced BCC) or infrequently may spread to distant sites (metastatic BCC). In the advanced stage, the lesions are no longer manageable by surgery or radiation therapy.5,6 Recently, inhibitors targeting the hedgehog (Hh) pathway have shown great promise for these patients. The first drug approved by the US Food and Drug Administration (FDA) for locally advanced and metastatic BCC is vismodegib.7 In this article, we provide a clinical review of vismodegib for the management of BCC, including a discussion of the Hh pathway in BCC, adverse effects of vismodegib, use of vismodegib in adnexal skin tumors, recommended doses for vismodegib therapy in BCC, and management of the side effects of treatment.
Hh Pathway in BCC
In embryonic development, the Hh signaling pathway is crucial across a broad spectrum of species, including humans. Various members of the Hh family have been recognized, all working as secreted regulatory proteins.8 The name of the Hh signaling pathway is derived from a polypeptide ligand called hedgehog found in some fruit flies. Mutations in the gene led to fruit fly larvae that had a spiky hairy pattern of denticles similar to hedgehogs, leading to the name of this molecule.9 The transmembrane protein smoothened (SMO) is the main component of the Hh signaling pathway and initiates a signaling cascade that in turn leads to an increased expression of target genes, such as GLI1. Patched (PTCH), also a transmembrane protein and a cell-surface receptor for the secreted Hh ligand, suppresses the signaling capacity of SMO. Upon binding of the Hh ligand to the PTCH receptor, the suppression of SMO is relieved and a signal is propagated, evoking a cellular response.10 Molecular and genetic studies have reported that genetic alterations in the Hh signaling pathway are almost universally present in all BCCs, leading to an aberrant activation of the pathway and an uncontrolled proliferation of the basal cells. Frequently, these alterations have been shown to cause loss of function of PTCH homologue 1, which usually acts to inhibit the SMO homologue signaling activity.11,12
Because of the potential importance of Hh signaling in other solid malignancies and the failure of topical inhibition of SMO,13 subsequent studies on the development of Hh pathway inhibitors have mostly focused on the systemic approach. A multitude of Hh pathway inhibitors have been developed thus far, such as SANT1-SANT4, GDC-0449, IPI-926, BMS-833923 (XL139), HhAntag-691, and MK-4101.14 Many of these inhibitors have been clinically investigated.13,15,16
Systemic SMO Inhibitor: Vismodegib
Vismodegib was the earliest systemic SMO inhibitor to fulfill early clinical evaluation15,16 and the first drug to receive FDA approval for the management of advanced or metastatic BCC. Vismodegib is a small-molecule SMO inhibitor used for the management of selected locally advanced BCC and metastatic BCC in adults.3,17 Although there is a possibility of recurrence following drug withdrawal, vismodegib constitutes a new therapeutic strategy presenting positive benefits to patients. It may provide superior improvement over sunitinib, which has shown efficacy in a few patients; however, the efficacy and tolerance of sunitinib have been shown to be limited.18,19
Adverse Effects of Vismodegib Therapy
Adverse events with vismodegib use have been reported in 98% of patients (N=491); most of these were mild to moderate.20 However, the frequency of adverse events could prove to be a therapeutic challenge for patients requiring extended treatment. The most frequently reported reversible side effects were muscle spasms (64%), alopecia (62%), weight loss (33%), fatigue (28%), decreased appetite (25%), diarrhea (17%), nausea (16%), dysgeusia (54%), and ageusia (22%).20 In clinical trials, amenorrhea was noticed in 30% (3/10) of females with reproductive potential.2 Apart from alopecia and possibly amenorrhea, these side effects are reversible.17 Alkeraye et al17 reported 3 clinical cases of persistent alopecia following the use of vismodegib. Amenorrhea is a possible side effect of unknown reversibility.7
Vismodegib is a pregnancy category D medication.4 Severe birth defects, including craniofacial abnormalities, retardations in normal growth, open perineum, and absence of digital fusion at a corresponding 20% of the recommended daily dose, were found in rat studies. Embryo-fetal death was noted when rats were exposedto concentrations comparable to the recommended human dose.4
Hepatic events with the use of vismodegib have been reported. The use of vismodegib in randomized controlled trials resulted in elevation of both alanine aminotransferase and aspartate aminotransferase levels compared with placebo.21 Moreover, severe hepatotoxicity with vismodegib has been reported.22-24 A study conducted by Edwards et al25 concluded that the use of vismodegib in patients with severe liver disease must include thorough risk-benefit assessment, with caution in using other concomitant hepatotoxic medications.
Rare adverse events also have been reported in the literature, including vismodegib-induced pancreatitis in a 79-year-old patient treated for locally advanced, recurrent BCC that was cleared following cessation of therapy.26 Additionally, atypical fibroxanthoma was observed in an 83-year-old patient after 30 days of treatment with vismodegib for multiple BCCs.27 The development of other secondary malignancies, such as squamous cell carcinoma, melanoma, keratoacanthomas, and pilomatricomas, during or after the long-term use of vismodegib also have been described.28-35
Use of Vismodegib for Adnexal Skin Tumors
The role of the sonic Hh–PTCH pathway in the pathogenesis of adnexal tumors varies in the literature. Some studies propose the involvement of this pathway in the formation of adnexal tumors such as trichoblastoma, trichoepithelioma, and cylindroma, as in BCC. Various lines of evidence support this involvement. Firstly, in mice, the spontaneous generation of numerous BCCs, trichoblastomas, trichoepitheliomas, and cylindromas has been observed following constitutive activation of the sonic Hh–PTCH pathway.36 Secondly, in trichoepitheliomas, there have been positive results in molecular research into the tumor suppressor gene PTCH homologue 1, PTCH1, whose mutations cause constitutive activation of the sonic Hh–PTCH pathway.37 Thirdly, GLI138 and SOX939 transcription factors associated with the signaling pathway of sonic Hh–PTCH appear to have increased levels in adnexal carcinomas.19 Lepesant et al19 reported a notable clinical response to vismodegib in trichoblastic carcinoma. Baur et al40 reported successful treatment of multiple familial trichoepitheliomas with vismodegib. Nonetheless, more studies are required to assess the efficacy and reliability of vismodegib in the management of adnexal tumors.
Recommended Dose of Vismodegib Therapy
The vismodegib dosage that is approved by the FDA is 150 mg/d until disease progression or the development of intolerable side effects.4 Higher dosing regimens were evaluated with 270 mg/d and 540 mg/d. No added therapeutic benefit was noted with the increase in the dose, and no dose-limiting toxic effects were observed.41
Management of Vismodegib Side Effects
Managing patient expectations is a crucial step in improving dysgeusia. The experience of dysgeusia varies among patients; thus, patients should be instructed to adjust their diets according to their level of dysgeusia, which can be achieved by changing ingredients or dressings used with their diet. This step has been proven to be effective in overcoming vismodegib-related dysgeusia. Also, fluid taste distortion may lead to dehydration and an increase in creatine level. Thus, patients should be encouraged to monitor fluid intake. Moreover, a treatment hiatus of 2 to 8 months results in near-complete improvement of taste distortion.
For muscle spasms, quinine, treatment break for 1 month, gentle exercise of affected areas, or muscle relaxants such as baclofen and temazepam all are effective methods. For vismodegib-related alopecia, managing patient expectations is key; patients should be aware that hair may take 6 to 12 months or even longer to regrow. In addition, shaving less frequently helps improve alopecia.
For gastrointestinal disorders, loperamide with or without codeine phosphate is effective in resolving diarrhea, and metoclopramide is mostly adequate in treating nausea. Another adverse event is weight loss; weight loss of 5% or more of total body weight prompts dietetic referral. If weight loss persists, a treatment break might be needed to regain weight.
Overall, treatment breaks are sufficient to resolve adverse events caused by vismodegib and do not compromise efficacy of treatment. The duration of a treatment break should be considered before initiation. In one clinical trial, a longer treatment break was associated with fewer adverse effects without affecting the efficacy of treatment.42
Conclusion
Vismodegib provides an effective alternative to surgical intervention in the management of BCC. However, patients must be monitored vigilantly, as adverse events are common (>90%).
- Sekulic A, Migden MR, Oro AE, et al. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. N Engl J Med. 2012;366:2171-2179.
- Rogers HW, Weinstock MA, Harris AR, et al. Incidence estimate of nonmelanoma skin cancer in the United States, 2006. Arch Dermatol. 2010;146:283-287.
- Von Hoff DD, LoRusso PM, Rudin CM, et al. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N Engl J Med. 2009;361:1164-1172.
- Cirrone F, Harris CS. Vismodegib and the hedgehog pathway: a new treatment for basal cell carcinoma. Clin Ther. 2012;34:2039-2050.
- Ruiz-Salas V, Alegre M, López-Ferrer A, et al. Vismodegib: a review [article in English, Spanish]. Actas Dermosifiliogr. 2014;105:744-751.
- Rubin AI, Chen EH, Ratner D. Basal-cell carcinoma. N Engl J Med. 2005;353:2262-2269.
- Cusack CA, Nijhawan R, Miller B, et al. Vismodegib for locally advanced basal cell carcinoma in a heart transplant patient. JAMA Dermatol. 2015;151:70-72.
- Aszterbaum M, Rothman A, Johnson RL, et al. Identification of mutations in the human PATCHED gene in sporadic basal cell carcinomas and in patients with the basal cell nevus syndrome. J Invest Dermatol. 1998;110:885-888.
- Abidi A. Hedgehog signaling pathway: a novel target for cancer therapy: vismodegib, a promising therapeutic option in treatment of basal cell carcinomas. Indian J Pharmacol. 2014;46:3-12.
- St-Jacques B, Dassule HR, Karavanova I, et al. Sonic hedgehog signaling is essential for hair development. Curr Biol. 1998;8:1058-1068.
- Gailani MR, Ståhle-Bäckdahl M, Leffell DJ, et al. The role of the human homologue of Drosophila patched in sporadic basal cell carcinomas. Nat Genet. 1996;14:78-81.
- Hall JM, Bell ML, Finger TE. Disruption of sonic hedgehog signaling alters growth and patterning of lingual taste papillae. Dev Biol. 2003;255:263-277.
- Bai CB, Stephen D, Joyner AL. All mouse ventral spinal cord patterning by hedgehog is Gli dependent and involves an activator function of Gli3. Dev Cell. 2004;6:103-115.
- Wang B, Fallon JF, Beachy PA. Hedgehog-regulated processing of Gli3 produces an anterior/posterior repressor gradient in the developing vertebrate limb. Cell. 2000;100:423-434.
- Sekulic A, Mangold AR, Northfelt DW, et al. Advanced basal cell carcinoma of the skin: targeting the hedgehog pathway. Curr Opin Oncol. 2013;25:218-223.
- Ingham PW, Placzek M. Orchestrating ontogenesis: variations on a theme by sonic hedgehog. Nature Rev Genet. 2006;7:841-850.
- Alkeraye S, Maire C, Desmedt E, et al. Persistent alopecia induced by vismodegib. Br J Dermatol. 2015;172:1671-1672.
- Battistella M, Mateus C, Lassau N, et al. Sunitinib efficacy in the treatment of metastatic skin adnexal carcinomas: report of two patients with hidradenocarcinoma and trichoblastic carcinoma. J Eur Acad Dermatol Venereol. 2010;24:199-203.
- Lepesant P, Crinquette M, Alkeraye S, et al. Vismodegib induces significant clinical response in locally advanced trichoblastic carcinoma. Br J Dermatol. 2015;173:1059-1062.
- Basset-Seguin N, Hauschild A, Grob JJ, et al. Vismodegib in patients with advanced basal cell carcinoma (STEVIE): a pre-plannedinterim analysis of an international, open-label trial. Lancet Oncol. 2015;16:729-736.
- Catenacci DV, Junttila MR, Karrison T, et al. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J Clin Oncol. 2015;33:4284-4292.
- Sanchez BE, Hajjafar L. Severe hepatotoxicity in a patient treated with hedgehog inhibitor: first case report. Gastroenterology. 2011;140:S974-S975.
- Ly P, Wolf K, Wilson J. A case of hepatotoxicity associated with vismodegib. JAAD Case Rep. 2018;5:57-59.
- Eiger-Moscovich M, Reich E, Tauber G, et al. Efficacy of vismodegib for the treatment of orbital and advanced periocular basal cell carcinoma. Am J Ophthalmol. 2019;207:62-70.
- Edwards BJ, Raisch DW, Saraykar SS, et al. Hepatotoxicity with vismodegib: an MD Anderson Cancer Center and Research on Adverse Drug Events and Reports Project. Drugs R D. 2017;17:211-218.
- Velter C, Blanc J, Robert C. Acute pancreatitis after vismodegib for basal cell carcinoma: a causal relation? Eur J Cancer. 2019;118:67-69.
- Giorgini C, Barbaccia V, Croci GA, et al. Rapid development of atypical fibroxanthoma during vismodegib treatment. Clin Exp Dermatol. 2019;44:86-88.
- Saintes C, Saint-Jean M, Brocard A, et al. Development of squamous cell carcinoma into basal cell carcinoma under treatment with vismodegib. J Eur Acad Dermatol Venereol. 2015;29:1006-1009.
- Zhu GA, Sundram U, Chang ALS. Two different scenarios of squamous cell carcinoma within advanced basal cell carcinomas: cases illustrating the importance of serial biopsy during vismodegib usage. JAMA Dermatol. 2014;150:970-973.
- Poulalhon N, Dalle S, Balme B, et al. Fast-growing cutaneous squamous cell carcinoma in a patient treated with vismodegib. Dermatology. 2015;230:101-104.
- Orouji A, Goerdt S, Utikal J, et al. Multiple highly and moderately differentiated squamous cell carcinomas of the skin during vismodegib treatment of inoperable basal cell carcinoma. Br J Dermatol. 2014;171:431-433.
- Iarrobino A, Messina JL, Kudchadkar R, et al. Emergence of a squamous cell carcinoma phenotype following treatment of metastatic basal cell carcinoma with vismodegib. J Am Acad Dermatol. 2013;69:E33-E34.
- Giuffrida R, Kashofer K, Dika E, et al. Fast growing melanoma following treatment with vismodegib for locally advanced basal cell carcinomas: report of two cases. Eur J Cancer. 2018;91:177-179.
- Aasi S, Silkiss R, Tang JY, et al. New onset of keratoacanthomas after vismodegib treatment for locally advanced basal cell carcinomas: a report of 2 cases. JAMA Dermatol. 2013;149:242-243.
- Magdaleno-Tapial J, Valenzuela-Oñate C, Ortiz-Salvador JM, et al. Pilomatricomas secondary to treatment with vismodegib. JAAD Case Rep. 2018;5:12-14.
- Nilsson M, Undèn AB, Krause D, et al. Induction of basal cell carcinomas and trichoepitheliomas in mice overexpressing GLI-1. Proc Natl Acad Sci U S A. 2000;97:3438-3443.
- Vorechovský I, Undén AB, Sandstedt B, et al. Trichoepitheliomas contain somatic mutations in the overexpressed PTCH gene: support for a gatekeeper mechanism in skin tumorigenesis. Cancer Res. 1997;57:4677-4681.
- Hatta N, Hirano T, Kimura T, et al. Molecular diagnosis of basal cell carcinoma and other basaloid cell neoplasms of the skin by the quantification of Gli1 transcript levels. J Cutan Pathol. 2005;32:131-136.
- Vidal VP, Ortonne N, Schedl A. SOX9 expression is a general marker of basal cell carcinoma and adnexal-related neoplasms. J Cutan Pathol. 2008;35:373-379.
- Baur V, Papadopoulos T, Kazakov DV, et al. A case of multiple familial trichoepitheliomas responding to treatment with the hedgehog signaling pathway inhibitor vismodegib. Virchows Arch. 2018;473:241-246.
- LoRusso PM, Rudin CM, Reddy JC, et al. Phase I trial of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally advanced or metastatic solid tumors. Clin Cancer Res. 2011;17:2502-2511.
- Fife K, Herd R, Lalondrelle S, et al. Managing adverse events associated with vismodegib in the treatment of basal cell carcinoma. Future Oncol. 2017;13:175-184.
Basal cell carcinomas (BCCs) are considered the most common cutaneous cancers. Approximately 80% of nonmelanoma skin cancers are BCCs.1,2 Surgical management is the gold standard for early-stage and localized BCCs; it may include simple excision vs Mohs micrographic surgery.3,4 However, if left untreated, these lesions can progress to an advanced stage (locally advanced BCC) or infrequently may spread to distant sites (metastatic BCC). In the advanced stage, the lesions are no longer manageable by surgery or radiation therapy.5,6 Recently, inhibitors targeting the hedgehog (Hh) pathway have shown great promise for these patients. The first drug approved by the US Food and Drug Administration (FDA) for locally advanced and metastatic BCC is vismodegib.7 In this article, we provide a clinical review of vismodegib for the management of BCC, including a discussion of the Hh pathway in BCC, adverse effects of vismodegib, use of vismodegib in adnexal skin tumors, recommended doses for vismodegib therapy in BCC, and management of the side effects of treatment.
Hh Pathway in BCC
In embryonic development, the Hh signaling pathway is crucial across a broad spectrum of species, including humans. Various members of the Hh family have been recognized, all working as secreted regulatory proteins.8 The name of the Hh signaling pathway is derived from a polypeptide ligand called hedgehog found in some fruit flies. Mutations in the gene led to fruit fly larvae that had a spiky hairy pattern of denticles similar to hedgehogs, leading to the name of this molecule.9 The transmembrane protein smoothened (SMO) is the main component of the Hh signaling pathway and initiates a signaling cascade that in turn leads to an increased expression of target genes, such as GLI1. Patched (PTCH), also a transmembrane protein and a cell-surface receptor for the secreted Hh ligand, suppresses the signaling capacity of SMO. Upon binding of the Hh ligand to the PTCH receptor, the suppression of SMO is relieved and a signal is propagated, evoking a cellular response.10 Molecular and genetic studies have reported that genetic alterations in the Hh signaling pathway are almost universally present in all BCCs, leading to an aberrant activation of the pathway and an uncontrolled proliferation of the basal cells. Frequently, these alterations have been shown to cause loss of function of PTCH homologue 1, which usually acts to inhibit the SMO homologue signaling activity.11,12
Because of the potential importance of Hh signaling in other solid malignancies and the failure of topical inhibition of SMO,13 subsequent studies on the development of Hh pathway inhibitors have mostly focused on the systemic approach. A multitude of Hh pathway inhibitors have been developed thus far, such as SANT1-SANT4, GDC-0449, IPI-926, BMS-833923 (XL139), HhAntag-691, and MK-4101.14 Many of these inhibitors have been clinically investigated.13,15,16
Systemic SMO Inhibitor: Vismodegib
Vismodegib was the earliest systemic SMO inhibitor to fulfill early clinical evaluation15,16 and the first drug to receive FDA approval for the management of advanced or metastatic BCC. Vismodegib is a small-molecule SMO inhibitor used for the management of selected locally advanced BCC and metastatic BCC in adults.3,17 Although there is a possibility of recurrence following drug withdrawal, vismodegib constitutes a new therapeutic strategy presenting positive benefits to patients. It may provide superior improvement over sunitinib, which has shown efficacy in a few patients; however, the efficacy and tolerance of sunitinib have been shown to be limited.18,19
Adverse Effects of Vismodegib Therapy
Adverse events with vismodegib use have been reported in 98% of patients (N=491); most of these were mild to moderate.20 However, the frequency of adverse events could prove to be a therapeutic challenge for patients requiring extended treatment. The most frequently reported reversible side effects were muscle spasms (64%), alopecia (62%), weight loss (33%), fatigue (28%), decreased appetite (25%), diarrhea (17%), nausea (16%), dysgeusia (54%), and ageusia (22%).20 In clinical trials, amenorrhea was noticed in 30% (3/10) of females with reproductive potential.2 Apart from alopecia and possibly amenorrhea, these side effects are reversible.17 Alkeraye et al17 reported 3 clinical cases of persistent alopecia following the use of vismodegib. Amenorrhea is a possible side effect of unknown reversibility.7
Vismodegib is a pregnancy category D medication.4 Severe birth defects, including craniofacial abnormalities, retardations in normal growth, open perineum, and absence of digital fusion at a corresponding 20% of the recommended daily dose, were found in rat studies. Embryo-fetal death was noted when rats were exposedto concentrations comparable to the recommended human dose.4
Hepatic events with the use of vismodegib have been reported. The use of vismodegib in randomized controlled trials resulted in elevation of both alanine aminotransferase and aspartate aminotransferase levels compared with placebo.21 Moreover, severe hepatotoxicity with vismodegib has been reported.22-24 A study conducted by Edwards et al25 concluded that the use of vismodegib in patients with severe liver disease must include thorough risk-benefit assessment, with caution in using other concomitant hepatotoxic medications.
Rare adverse events also have been reported in the literature, including vismodegib-induced pancreatitis in a 79-year-old patient treated for locally advanced, recurrent BCC that was cleared following cessation of therapy.26 Additionally, atypical fibroxanthoma was observed in an 83-year-old patient after 30 days of treatment with vismodegib for multiple BCCs.27 The development of other secondary malignancies, such as squamous cell carcinoma, melanoma, keratoacanthomas, and pilomatricomas, during or after the long-term use of vismodegib also have been described.28-35
Use of Vismodegib for Adnexal Skin Tumors
The role of the sonic Hh–PTCH pathway in the pathogenesis of adnexal tumors varies in the literature. Some studies propose the involvement of this pathway in the formation of adnexal tumors such as trichoblastoma, trichoepithelioma, and cylindroma, as in BCC. Various lines of evidence support this involvement. Firstly, in mice, the spontaneous generation of numerous BCCs, trichoblastomas, trichoepitheliomas, and cylindromas has been observed following constitutive activation of the sonic Hh–PTCH pathway.36 Secondly, in trichoepitheliomas, there have been positive results in molecular research into the tumor suppressor gene PTCH homologue 1, PTCH1, whose mutations cause constitutive activation of the sonic Hh–PTCH pathway.37 Thirdly, GLI138 and SOX939 transcription factors associated with the signaling pathway of sonic Hh–PTCH appear to have increased levels in adnexal carcinomas.19 Lepesant et al19 reported a notable clinical response to vismodegib in trichoblastic carcinoma. Baur et al40 reported successful treatment of multiple familial trichoepitheliomas with vismodegib. Nonetheless, more studies are required to assess the efficacy and reliability of vismodegib in the management of adnexal tumors.
Recommended Dose of Vismodegib Therapy
The vismodegib dosage that is approved by the FDA is 150 mg/d until disease progression or the development of intolerable side effects.4 Higher dosing regimens were evaluated with 270 mg/d and 540 mg/d. No added therapeutic benefit was noted with the increase in the dose, and no dose-limiting toxic effects were observed.41
Management of Vismodegib Side Effects
Managing patient expectations is a crucial step in improving dysgeusia. The experience of dysgeusia varies among patients; thus, patients should be instructed to adjust their diets according to their level of dysgeusia, which can be achieved by changing ingredients or dressings used with their diet. This step has been proven to be effective in overcoming vismodegib-related dysgeusia. Also, fluid taste distortion may lead to dehydration and an increase in creatine level. Thus, patients should be encouraged to monitor fluid intake. Moreover, a treatment hiatus of 2 to 8 months results in near-complete improvement of taste distortion.
For muscle spasms, quinine, treatment break for 1 month, gentle exercise of affected areas, or muscle relaxants such as baclofen and temazepam all are effective methods. For vismodegib-related alopecia, managing patient expectations is key; patients should be aware that hair may take 6 to 12 months or even longer to regrow. In addition, shaving less frequently helps improve alopecia.
For gastrointestinal disorders, loperamide with or without codeine phosphate is effective in resolving diarrhea, and metoclopramide is mostly adequate in treating nausea. Another adverse event is weight loss; weight loss of 5% or more of total body weight prompts dietetic referral. If weight loss persists, a treatment break might be needed to regain weight.
Overall, treatment breaks are sufficient to resolve adverse events caused by vismodegib and do not compromise efficacy of treatment. The duration of a treatment break should be considered before initiation. In one clinical trial, a longer treatment break was associated with fewer adverse effects without affecting the efficacy of treatment.42
Conclusion
Vismodegib provides an effective alternative to surgical intervention in the management of BCC. However, patients must be monitored vigilantly, as adverse events are common (>90%).
Basal cell carcinomas (BCCs) are considered the most common cutaneous cancers. Approximately 80% of nonmelanoma skin cancers are BCCs.1,2 Surgical management is the gold standard for early-stage and localized BCCs; it may include simple excision vs Mohs micrographic surgery.3,4 However, if left untreated, these lesions can progress to an advanced stage (locally advanced BCC) or infrequently may spread to distant sites (metastatic BCC). In the advanced stage, the lesions are no longer manageable by surgery or radiation therapy.5,6 Recently, inhibitors targeting the hedgehog (Hh) pathway have shown great promise for these patients. The first drug approved by the US Food and Drug Administration (FDA) for locally advanced and metastatic BCC is vismodegib.7 In this article, we provide a clinical review of vismodegib for the management of BCC, including a discussion of the Hh pathway in BCC, adverse effects of vismodegib, use of vismodegib in adnexal skin tumors, recommended doses for vismodegib therapy in BCC, and management of the side effects of treatment.
Hh Pathway in BCC
In embryonic development, the Hh signaling pathway is crucial across a broad spectrum of species, including humans. Various members of the Hh family have been recognized, all working as secreted regulatory proteins.8 The name of the Hh signaling pathway is derived from a polypeptide ligand called hedgehog found in some fruit flies. Mutations in the gene led to fruit fly larvae that had a spiky hairy pattern of denticles similar to hedgehogs, leading to the name of this molecule.9 The transmembrane protein smoothened (SMO) is the main component of the Hh signaling pathway and initiates a signaling cascade that in turn leads to an increased expression of target genes, such as GLI1. Patched (PTCH), also a transmembrane protein and a cell-surface receptor for the secreted Hh ligand, suppresses the signaling capacity of SMO. Upon binding of the Hh ligand to the PTCH receptor, the suppression of SMO is relieved and a signal is propagated, evoking a cellular response.10 Molecular and genetic studies have reported that genetic alterations in the Hh signaling pathway are almost universally present in all BCCs, leading to an aberrant activation of the pathway and an uncontrolled proliferation of the basal cells. Frequently, these alterations have been shown to cause loss of function of PTCH homologue 1, which usually acts to inhibit the SMO homologue signaling activity.11,12
Because of the potential importance of Hh signaling in other solid malignancies and the failure of topical inhibition of SMO,13 subsequent studies on the development of Hh pathway inhibitors have mostly focused on the systemic approach. A multitude of Hh pathway inhibitors have been developed thus far, such as SANT1-SANT4, GDC-0449, IPI-926, BMS-833923 (XL139), HhAntag-691, and MK-4101.14 Many of these inhibitors have been clinically investigated.13,15,16
Systemic SMO Inhibitor: Vismodegib
Vismodegib was the earliest systemic SMO inhibitor to fulfill early clinical evaluation15,16 and the first drug to receive FDA approval for the management of advanced or metastatic BCC. Vismodegib is a small-molecule SMO inhibitor used for the management of selected locally advanced BCC and metastatic BCC in adults.3,17 Although there is a possibility of recurrence following drug withdrawal, vismodegib constitutes a new therapeutic strategy presenting positive benefits to patients. It may provide superior improvement over sunitinib, which has shown efficacy in a few patients; however, the efficacy and tolerance of sunitinib have been shown to be limited.18,19
Adverse Effects of Vismodegib Therapy
Adverse events with vismodegib use have been reported in 98% of patients (N=491); most of these were mild to moderate.20 However, the frequency of adverse events could prove to be a therapeutic challenge for patients requiring extended treatment. The most frequently reported reversible side effects were muscle spasms (64%), alopecia (62%), weight loss (33%), fatigue (28%), decreased appetite (25%), diarrhea (17%), nausea (16%), dysgeusia (54%), and ageusia (22%).20 In clinical trials, amenorrhea was noticed in 30% (3/10) of females with reproductive potential.2 Apart from alopecia and possibly amenorrhea, these side effects are reversible.17 Alkeraye et al17 reported 3 clinical cases of persistent alopecia following the use of vismodegib. Amenorrhea is a possible side effect of unknown reversibility.7
Vismodegib is a pregnancy category D medication.4 Severe birth defects, including craniofacial abnormalities, retardations in normal growth, open perineum, and absence of digital fusion at a corresponding 20% of the recommended daily dose, were found in rat studies. Embryo-fetal death was noted when rats were exposedto concentrations comparable to the recommended human dose.4
Hepatic events with the use of vismodegib have been reported. The use of vismodegib in randomized controlled trials resulted in elevation of both alanine aminotransferase and aspartate aminotransferase levels compared with placebo.21 Moreover, severe hepatotoxicity with vismodegib has been reported.22-24 A study conducted by Edwards et al25 concluded that the use of vismodegib in patients with severe liver disease must include thorough risk-benefit assessment, with caution in using other concomitant hepatotoxic medications.
Rare adverse events also have been reported in the literature, including vismodegib-induced pancreatitis in a 79-year-old patient treated for locally advanced, recurrent BCC that was cleared following cessation of therapy.26 Additionally, atypical fibroxanthoma was observed in an 83-year-old patient after 30 days of treatment with vismodegib for multiple BCCs.27 The development of other secondary malignancies, such as squamous cell carcinoma, melanoma, keratoacanthomas, and pilomatricomas, during or after the long-term use of vismodegib also have been described.28-35
Use of Vismodegib for Adnexal Skin Tumors
The role of the sonic Hh–PTCH pathway in the pathogenesis of adnexal tumors varies in the literature. Some studies propose the involvement of this pathway in the formation of adnexal tumors such as trichoblastoma, trichoepithelioma, and cylindroma, as in BCC. Various lines of evidence support this involvement. Firstly, in mice, the spontaneous generation of numerous BCCs, trichoblastomas, trichoepitheliomas, and cylindromas has been observed following constitutive activation of the sonic Hh–PTCH pathway.36 Secondly, in trichoepitheliomas, there have been positive results in molecular research into the tumor suppressor gene PTCH homologue 1, PTCH1, whose mutations cause constitutive activation of the sonic Hh–PTCH pathway.37 Thirdly, GLI138 and SOX939 transcription factors associated with the signaling pathway of sonic Hh–PTCH appear to have increased levels in adnexal carcinomas.19 Lepesant et al19 reported a notable clinical response to vismodegib in trichoblastic carcinoma. Baur et al40 reported successful treatment of multiple familial trichoepitheliomas with vismodegib. Nonetheless, more studies are required to assess the efficacy and reliability of vismodegib in the management of adnexal tumors.
Recommended Dose of Vismodegib Therapy
The vismodegib dosage that is approved by the FDA is 150 mg/d until disease progression or the development of intolerable side effects.4 Higher dosing regimens were evaluated with 270 mg/d and 540 mg/d. No added therapeutic benefit was noted with the increase in the dose, and no dose-limiting toxic effects were observed.41
Management of Vismodegib Side Effects
Managing patient expectations is a crucial step in improving dysgeusia. The experience of dysgeusia varies among patients; thus, patients should be instructed to adjust their diets according to their level of dysgeusia, which can be achieved by changing ingredients or dressings used with their diet. This step has been proven to be effective in overcoming vismodegib-related dysgeusia. Also, fluid taste distortion may lead to dehydration and an increase in creatine level. Thus, patients should be encouraged to monitor fluid intake. Moreover, a treatment hiatus of 2 to 8 months results in near-complete improvement of taste distortion.
For muscle spasms, quinine, treatment break for 1 month, gentle exercise of affected areas, or muscle relaxants such as baclofen and temazepam all are effective methods. For vismodegib-related alopecia, managing patient expectations is key; patients should be aware that hair may take 6 to 12 months or even longer to regrow. In addition, shaving less frequently helps improve alopecia.
For gastrointestinal disorders, loperamide with or without codeine phosphate is effective in resolving diarrhea, and metoclopramide is mostly adequate in treating nausea. Another adverse event is weight loss; weight loss of 5% or more of total body weight prompts dietetic referral. If weight loss persists, a treatment break might be needed to regain weight.
Overall, treatment breaks are sufficient to resolve adverse events caused by vismodegib and do not compromise efficacy of treatment. The duration of a treatment break should be considered before initiation. In one clinical trial, a longer treatment break was associated with fewer adverse effects without affecting the efficacy of treatment.42
Conclusion
Vismodegib provides an effective alternative to surgical intervention in the management of BCC. However, patients must be monitored vigilantly, as adverse events are common (>90%).
- Sekulic A, Migden MR, Oro AE, et al. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. N Engl J Med. 2012;366:2171-2179.
- Rogers HW, Weinstock MA, Harris AR, et al. Incidence estimate of nonmelanoma skin cancer in the United States, 2006. Arch Dermatol. 2010;146:283-287.
- Von Hoff DD, LoRusso PM, Rudin CM, et al. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N Engl J Med. 2009;361:1164-1172.
- Cirrone F, Harris CS. Vismodegib and the hedgehog pathway: a new treatment for basal cell carcinoma. Clin Ther. 2012;34:2039-2050.
- Ruiz-Salas V, Alegre M, López-Ferrer A, et al. Vismodegib: a review [article in English, Spanish]. Actas Dermosifiliogr. 2014;105:744-751.
- Rubin AI, Chen EH, Ratner D. Basal-cell carcinoma. N Engl J Med. 2005;353:2262-2269.
- Cusack CA, Nijhawan R, Miller B, et al. Vismodegib for locally advanced basal cell carcinoma in a heart transplant patient. JAMA Dermatol. 2015;151:70-72.
- Aszterbaum M, Rothman A, Johnson RL, et al. Identification of mutations in the human PATCHED gene in sporadic basal cell carcinomas and in patients with the basal cell nevus syndrome. J Invest Dermatol. 1998;110:885-888.
- Abidi A. Hedgehog signaling pathway: a novel target for cancer therapy: vismodegib, a promising therapeutic option in treatment of basal cell carcinomas. Indian J Pharmacol. 2014;46:3-12.
- St-Jacques B, Dassule HR, Karavanova I, et al. Sonic hedgehog signaling is essential for hair development. Curr Biol. 1998;8:1058-1068.
- Gailani MR, Ståhle-Bäckdahl M, Leffell DJ, et al. The role of the human homologue of Drosophila patched in sporadic basal cell carcinomas. Nat Genet. 1996;14:78-81.
- Hall JM, Bell ML, Finger TE. Disruption of sonic hedgehog signaling alters growth and patterning of lingual taste papillae. Dev Biol. 2003;255:263-277.
- Bai CB, Stephen D, Joyner AL. All mouse ventral spinal cord patterning by hedgehog is Gli dependent and involves an activator function of Gli3. Dev Cell. 2004;6:103-115.
- Wang B, Fallon JF, Beachy PA. Hedgehog-regulated processing of Gli3 produces an anterior/posterior repressor gradient in the developing vertebrate limb. Cell. 2000;100:423-434.
- Sekulic A, Mangold AR, Northfelt DW, et al. Advanced basal cell carcinoma of the skin: targeting the hedgehog pathway. Curr Opin Oncol. 2013;25:218-223.
- Ingham PW, Placzek M. Orchestrating ontogenesis: variations on a theme by sonic hedgehog. Nature Rev Genet. 2006;7:841-850.
- Alkeraye S, Maire C, Desmedt E, et al. Persistent alopecia induced by vismodegib. Br J Dermatol. 2015;172:1671-1672.
- Battistella M, Mateus C, Lassau N, et al. Sunitinib efficacy in the treatment of metastatic skin adnexal carcinomas: report of two patients with hidradenocarcinoma and trichoblastic carcinoma. J Eur Acad Dermatol Venereol. 2010;24:199-203.
- Lepesant P, Crinquette M, Alkeraye S, et al. Vismodegib induces significant clinical response in locally advanced trichoblastic carcinoma. Br J Dermatol. 2015;173:1059-1062.
- Basset-Seguin N, Hauschild A, Grob JJ, et al. Vismodegib in patients with advanced basal cell carcinoma (STEVIE): a pre-plannedinterim analysis of an international, open-label trial. Lancet Oncol. 2015;16:729-736.
- Catenacci DV, Junttila MR, Karrison T, et al. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J Clin Oncol. 2015;33:4284-4292.
- Sanchez BE, Hajjafar L. Severe hepatotoxicity in a patient treated with hedgehog inhibitor: first case report. Gastroenterology. 2011;140:S974-S975.
- Ly P, Wolf K, Wilson J. A case of hepatotoxicity associated with vismodegib. JAAD Case Rep. 2018;5:57-59.
- Eiger-Moscovich M, Reich E, Tauber G, et al. Efficacy of vismodegib for the treatment of orbital and advanced periocular basal cell carcinoma. Am J Ophthalmol. 2019;207:62-70.
- Edwards BJ, Raisch DW, Saraykar SS, et al. Hepatotoxicity with vismodegib: an MD Anderson Cancer Center and Research on Adverse Drug Events and Reports Project. Drugs R D. 2017;17:211-218.
- Velter C, Blanc J, Robert C. Acute pancreatitis after vismodegib for basal cell carcinoma: a causal relation? Eur J Cancer. 2019;118:67-69.
- Giorgini C, Barbaccia V, Croci GA, et al. Rapid development of atypical fibroxanthoma during vismodegib treatment. Clin Exp Dermatol. 2019;44:86-88.
- Saintes C, Saint-Jean M, Brocard A, et al. Development of squamous cell carcinoma into basal cell carcinoma under treatment with vismodegib. J Eur Acad Dermatol Venereol. 2015;29:1006-1009.
- Zhu GA, Sundram U, Chang ALS. Two different scenarios of squamous cell carcinoma within advanced basal cell carcinomas: cases illustrating the importance of serial biopsy during vismodegib usage. JAMA Dermatol. 2014;150:970-973.
- Poulalhon N, Dalle S, Balme B, et al. Fast-growing cutaneous squamous cell carcinoma in a patient treated with vismodegib. Dermatology. 2015;230:101-104.
- Orouji A, Goerdt S, Utikal J, et al. Multiple highly and moderately differentiated squamous cell carcinomas of the skin during vismodegib treatment of inoperable basal cell carcinoma. Br J Dermatol. 2014;171:431-433.
- Iarrobino A, Messina JL, Kudchadkar R, et al. Emergence of a squamous cell carcinoma phenotype following treatment of metastatic basal cell carcinoma with vismodegib. J Am Acad Dermatol. 2013;69:E33-E34.
- Giuffrida R, Kashofer K, Dika E, et al. Fast growing melanoma following treatment with vismodegib for locally advanced basal cell carcinomas: report of two cases. Eur J Cancer. 2018;91:177-179.
- Aasi S, Silkiss R, Tang JY, et al. New onset of keratoacanthomas after vismodegib treatment for locally advanced basal cell carcinomas: a report of 2 cases. JAMA Dermatol. 2013;149:242-243.
- Magdaleno-Tapial J, Valenzuela-Oñate C, Ortiz-Salvador JM, et al. Pilomatricomas secondary to treatment with vismodegib. JAAD Case Rep. 2018;5:12-14.
- Nilsson M, Undèn AB, Krause D, et al. Induction of basal cell carcinomas and trichoepitheliomas in mice overexpressing GLI-1. Proc Natl Acad Sci U S A. 2000;97:3438-3443.
- Vorechovský I, Undén AB, Sandstedt B, et al. Trichoepitheliomas contain somatic mutations in the overexpressed PTCH gene: support for a gatekeeper mechanism in skin tumorigenesis. Cancer Res. 1997;57:4677-4681.
- Hatta N, Hirano T, Kimura T, et al. Molecular diagnosis of basal cell carcinoma and other basaloid cell neoplasms of the skin by the quantification of Gli1 transcript levels. J Cutan Pathol. 2005;32:131-136.
- Vidal VP, Ortonne N, Schedl A. SOX9 expression is a general marker of basal cell carcinoma and adnexal-related neoplasms. J Cutan Pathol. 2008;35:373-379.
- Baur V, Papadopoulos T, Kazakov DV, et al. A case of multiple familial trichoepitheliomas responding to treatment with the hedgehog signaling pathway inhibitor vismodegib. Virchows Arch. 2018;473:241-246.
- LoRusso PM, Rudin CM, Reddy JC, et al. Phase I trial of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally advanced or metastatic solid tumors. Clin Cancer Res. 2011;17:2502-2511.
- Fife K, Herd R, Lalondrelle S, et al. Managing adverse events associated with vismodegib in the treatment of basal cell carcinoma. Future Oncol. 2017;13:175-184.
- Sekulic A, Migden MR, Oro AE, et al. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. N Engl J Med. 2012;366:2171-2179.
- Rogers HW, Weinstock MA, Harris AR, et al. Incidence estimate of nonmelanoma skin cancer in the United States, 2006. Arch Dermatol. 2010;146:283-287.
- Von Hoff DD, LoRusso PM, Rudin CM, et al. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N Engl J Med. 2009;361:1164-1172.
- Cirrone F, Harris CS. Vismodegib and the hedgehog pathway: a new treatment for basal cell carcinoma. Clin Ther. 2012;34:2039-2050.
- Ruiz-Salas V, Alegre M, López-Ferrer A, et al. Vismodegib: a review [article in English, Spanish]. Actas Dermosifiliogr. 2014;105:744-751.
- Rubin AI, Chen EH, Ratner D. Basal-cell carcinoma. N Engl J Med. 2005;353:2262-2269.
- Cusack CA, Nijhawan R, Miller B, et al. Vismodegib for locally advanced basal cell carcinoma in a heart transplant patient. JAMA Dermatol. 2015;151:70-72.
- Aszterbaum M, Rothman A, Johnson RL, et al. Identification of mutations in the human PATCHED gene in sporadic basal cell carcinomas and in patients with the basal cell nevus syndrome. J Invest Dermatol. 1998;110:885-888.
- Abidi A. Hedgehog signaling pathway: a novel target for cancer therapy: vismodegib, a promising therapeutic option in treatment of basal cell carcinomas. Indian J Pharmacol. 2014;46:3-12.
- St-Jacques B, Dassule HR, Karavanova I, et al. Sonic hedgehog signaling is essential for hair development. Curr Biol. 1998;8:1058-1068.
- Gailani MR, Ståhle-Bäckdahl M, Leffell DJ, et al. The role of the human homologue of Drosophila patched in sporadic basal cell carcinomas. Nat Genet. 1996;14:78-81.
- Hall JM, Bell ML, Finger TE. Disruption of sonic hedgehog signaling alters growth and patterning of lingual taste papillae. Dev Biol. 2003;255:263-277.
- Bai CB, Stephen D, Joyner AL. All mouse ventral spinal cord patterning by hedgehog is Gli dependent and involves an activator function of Gli3. Dev Cell. 2004;6:103-115.
- Wang B, Fallon JF, Beachy PA. Hedgehog-regulated processing of Gli3 produces an anterior/posterior repressor gradient in the developing vertebrate limb. Cell. 2000;100:423-434.
- Sekulic A, Mangold AR, Northfelt DW, et al. Advanced basal cell carcinoma of the skin: targeting the hedgehog pathway. Curr Opin Oncol. 2013;25:218-223.
- Ingham PW, Placzek M. Orchestrating ontogenesis: variations on a theme by sonic hedgehog. Nature Rev Genet. 2006;7:841-850.
- Alkeraye S, Maire C, Desmedt E, et al. Persistent alopecia induced by vismodegib. Br J Dermatol. 2015;172:1671-1672.
- Battistella M, Mateus C, Lassau N, et al. Sunitinib efficacy in the treatment of metastatic skin adnexal carcinomas: report of two patients with hidradenocarcinoma and trichoblastic carcinoma. J Eur Acad Dermatol Venereol. 2010;24:199-203.
- Lepesant P, Crinquette M, Alkeraye S, et al. Vismodegib induces significant clinical response in locally advanced trichoblastic carcinoma. Br J Dermatol. 2015;173:1059-1062.
- Basset-Seguin N, Hauschild A, Grob JJ, et al. Vismodegib in patients with advanced basal cell carcinoma (STEVIE): a pre-plannedinterim analysis of an international, open-label trial. Lancet Oncol. 2015;16:729-736.
- Catenacci DV, Junttila MR, Karrison T, et al. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J Clin Oncol. 2015;33:4284-4292.
- Sanchez BE, Hajjafar L. Severe hepatotoxicity in a patient treated with hedgehog inhibitor: first case report. Gastroenterology. 2011;140:S974-S975.
- Ly P, Wolf K, Wilson J. A case of hepatotoxicity associated with vismodegib. JAAD Case Rep. 2018;5:57-59.
- Eiger-Moscovich M, Reich E, Tauber G, et al. Efficacy of vismodegib for the treatment of orbital and advanced periocular basal cell carcinoma. Am J Ophthalmol. 2019;207:62-70.
- Edwards BJ, Raisch DW, Saraykar SS, et al. Hepatotoxicity with vismodegib: an MD Anderson Cancer Center and Research on Adverse Drug Events and Reports Project. Drugs R D. 2017;17:211-218.
- Velter C, Blanc J, Robert C. Acute pancreatitis after vismodegib for basal cell carcinoma: a causal relation? Eur J Cancer. 2019;118:67-69.
- Giorgini C, Barbaccia V, Croci GA, et al. Rapid development of atypical fibroxanthoma during vismodegib treatment. Clin Exp Dermatol. 2019;44:86-88.
- Saintes C, Saint-Jean M, Brocard A, et al. Development of squamous cell carcinoma into basal cell carcinoma under treatment with vismodegib. J Eur Acad Dermatol Venereol. 2015;29:1006-1009.
- Zhu GA, Sundram U, Chang ALS. Two different scenarios of squamous cell carcinoma within advanced basal cell carcinomas: cases illustrating the importance of serial biopsy during vismodegib usage. JAMA Dermatol. 2014;150:970-973.
- Poulalhon N, Dalle S, Balme B, et al. Fast-growing cutaneous squamous cell carcinoma in a patient treated with vismodegib. Dermatology. 2015;230:101-104.
- Orouji A, Goerdt S, Utikal J, et al. Multiple highly and moderately differentiated squamous cell carcinomas of the skin during vismodegib treatment of inoperable basal cell carcinoma. Br J Dermatol. 2014;171:431-433.
- Iarrobino A, Messina JL, Kudchadkar R, et al. Emergence of a squamous cell carcinoma phenotype following treatment of metastatic basal cell carcinoma with vismodegib. J Am Acad Dermatol. 2013;69:E33-E34.
- Giuffrida R, Kashofer K, Dika E, et al. Fast growing melanoma following treatment with vismodegib for locally advanced basal cell carcinomas: report of two cases. Eur J Cancer. 2018;91:177-179.
- Aasi S, Silkiss R, Tang JY, et al. New onset of keratoacanthomas after vismodegib treatment for locally advanced basal cell carcinomas: a report of 2 cases. JAMA Dermatol. 2013;149:242-243.
- Magdaleno-Tapial J, Valenzuela-Oñate C, Ortiz-Salvador JM, et al. Pilomatricomas secondary to treatment with vismodegib. JAAD Case Rep. 2018;5:12-14.
- Nilsson M, Undèn AB, Krause D, et al. Induction of basal cell carcinomas and trichoepitheliomas in mice overexpressing GLI-1. Proc Natl Acad Sci U S A. 2000;97:3438-3443.
- Vorechovský I, Undén AB, Sandstedt B, et al. Trichoepitheliomas contain somatic mutations in the overexpressed PTCH gene: support for a gatekeeper mechanism in skin tumorigenesis. Cancer Res. 1997;57:4677-4681.
- Hatta N, Hirano T, Kimura T, et al. Molecular diagnosis of basal cell carcinoma and other basaloid cell neoplasms of the skin by the quantification of Gli1 transcript levels. J Cutan Pathol. 2005;32:131-136.
- Vidal VP, Ortonne N, Schedl A. SOX9 expression is a general marker of basal cell carcinoma and adnexal-related neoplasms. J Cutan Pathol. 2008;35:373-379.
- Baur V, Papadopoulos T, Kazakov DV, et al. A case of multiple familial trichoepitheliomas responding to treatment with the hedgehog signaling pathway inhibitor vismodegib. Virchows Arch. 2018;473:241-246.
- LoRusso PM, Rudin CM, Reddy JC, et al. Phase I trial of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally advanced or metastatic solid tumors. Clin Cancer Res. 2011;17:2502-2511.
- Fife K, Herd R, Lalondrelle S, et al. Managing adverse events associated with vismodegib in the treatment of basal cell carcinoma. Future Oncol. 2017;13:175-184.
Practice Points
- The recommended dosage of vismodegib is 150 mg/d until unendurable side effects develop or disease progression occurs.
- The efficacy of vismodegib in the management of locally advanced basal cell carcinoma (BCC) and metastatic BCC is promising. Thus, it is now considered an effective substitute to surgical therapy.
- Patients using vismodegib must be closely monitored, as it is commonly associated with adverse events.
Surgical Deroofing for Hidradenitis Suppurativa
Practice Gap
Hidradenitis suppurativa (HS) is a chronic inflammatory skin condition characterized by inflammatory nodules, abscesses, sinus tracts, fistulae, and scarring, mainly in intertriginous areas. The extent of disease—classified using the Hurley staging system (stages I–III)—helps guide treatment, which includes medical management and surgical intervention in later stages.
First-line treatment of HS includes topical or systemic medications, or both. Surgical therapy typically is reserved for refractory HS in moderate to severe disease (Hurley stages II and III) and is combined with pharmacotherapy. Specifically, clinical management guidelines issued by an expert committee of the United States and Canadian Hidradenitis Suppurativa Foundations recommend excision or deroofing for recurrent nodules and tunnels.1
Surgical options for HS that are available to the outpatient dermatologist include incision and drainage, electrosurgery, CO2 laser evaporation, excision, and deroofing (also known as unroofing).2 Deroofing is a fairly novel therapy; many dermatologists are unfamiliar with the procedure. A PubMed search of articles indexed for MEDLINE related to HS prior to 2010 revealed only 1 article containing the word deroofing and only 4 articles containing unroofing.
The pathophysiology of HS has important implications for successful treatment. Inflammation of the follicular pilosebaceous unit along with follicular occlusion create challenges with treatment.3 It is postulated that a defect in the glassy membrane of the infra-infundibular wall predisposes the pilosebaceous follicle to lose its structural integrality as pressure builds from plugging of the duct,4 which can result in the clinical hallmarks of HS including tunneling tracts, bridging nodules, abscesses, and fistulae that form with lateral expansion of the plugged follicle.
Leaking of the contents of these plugged follicles into surrounding tissue produces an inflammatory response in characteristic HS lesions. Because debris within the lesions moves laterally instead of being able to burst to the surface, the lesions have difficulty fully healing. Unroofing the lesions and removing built-up debris allows them to heal more expediently and quiets the underlying immune response by removing the stimulus.4
Herein, we describe the benefits, risks, and surgical process of deroofing for HS.
Technique and Tools
Deroofing is performed under local anesthesia, stepwise as follows:
1. Identify sinus tracts and infiltrate the area with lidocaine (Figure, A).
2. Use a blunt probe to define the borders of the area to be unroofed and to evaluate for any communicating sinus tracts (Figure, B).
3. Remove the roof of underlying abscesses and tracts, using a probe as a guide (Figure, C).
4. Enter through the skin or sinus opening using electrocautery or with a scalpel or scissors; perform blunt dissection.
5. Reflect back the entirety of skin overlying the probed areas and remove the skin to expose the base of the lesion (Figure, D).
6. Explore the exposed base and walls of the lesion with the probe again to assess for hidden tracts; take care not to create false tracts.
7. Debride the surgical wound using curettage or rough gauze grattage to remove remaining inflammatory debris or biofilm. To achieve hemostasis, apply aluminum chloride or ferric chloride. Coat the wound with petroleum jelly and gauze and allow it to heal by secondary intention.
8. Educate the patient on wound care—once-daily gentle cleansing with soap and water, followed by application of a moist dressing—which is similar to wound healing by secondary intention from other causes.2,4
Practice Implications
A deroofing procedure has many benefits compared to other surgical modalities for the treatment of HS. Deroofing requires only a probe, curette, and electrocautery device, making the procedure more cost-effective than excision, which requires a full tray of equipment and sutures. Furthermore, margins do not need to be taken with deroofing, and no undermining or closure is needed, which saves time during the operation and minimizes the risk for complications, including dehiscence and formation of new sinus tracts.4 No specialized equipment, such as a CO2 laser, is required, which makes deroofing accessible to every clinical dermatologist in any demographic or geographic setting.
Evidence of Benefit—Saylor and colleagues5 found that deroofing carries a 12.5% complication rate, which includes postoperative bleeding, hypergranulation tissue, and rarely wound infection. This rate is significantly lower than the 26% complication rate associated with local excision, which includes wound dehiscence, infection, and contracture (P<.001). Deroofing also was found to have an HS recurrence rate of 14.5%, which is significantly less than the 30% recurrence rate seen with local excision (P=.015). Saylor et al5 also concluded that incision and drainage was recommended only for immediate relief of HS because of its 100% recurrence rate.
van der Zee2 reported on 88 lesions from 44 patients that were treated by surgical deroofing, resulting in an average defect of 3.0 cm in length and a mean healing time of 14 days. The typical outcome was cosmetically acceptable scarring; this finding was supported by a postoperative survey (>1 year), to which 37 of 44 patients responded and assigned an average satisfaction score of 8 (of a possible 10) and a recommendation rate of 90%.2
Procedural Coding—Specific Current Procedural Terminology codes (11450-11471) from the International Classification of Diseases, Tenth Revision, exist for HS deroofing procedures; the applicable code for a given case depends on the final length of the surgical defect. Documentation to support these codes is similar to the note for an excision procedure, taking care to include location, depth, and length of the excision; healing by secondary intention; and the diagnosis of HS.
Final Thoughts
Deroofing is a surgical option that can be beneficial to patients with HS. It is a relatively simple procedure available to any dermatologist, regardless of setting. We encourage dermatologists to consider deroofing, even in patients with Hurley stage II lesions, because it can yield cosmetically acceptable and definitive results, given the variety of therapies available for HS. Deroofing also can be superior to standard excision, especially because of the potential complications with standard excision and quicker operative time with deroofing. As more providers become familiar with the deroofing procedure for HS, further studies can be undertaken to add to the paucity of data about deroofing and how it compares to other surgical treatments.
- Alikhan A, Sayed C, Alavi A, et al. North American clinical management guidelines for hidradenitis suppurativa: a publication from the United States and Canadian Hidradenitis Suppurativa Foundations: part I: diagnosis, evaluation, and the use of complementary and procedural management. J Am Acad Dermatol. 2019;81:76-90. doi:10.1016/j.jaad.2019.02.067
- van der Zee HH, Prens EP, Boer J. Deroofing: a tissue-saving surgical technique for the treatment of mild to moderate hidradenitis suppurativa lesions. J Am Acad Dermatol. 2010;63:475-480. doi:10.1016/j.jaad.2009.12.018
- Napolitano M, Megna M, Timoshchuk EA, et al. Hidradenitis suppurativa: from pathogenesis to diagnosis and treatment. Clin Cosmet Investig Dermatol. 2017;10:105-115. doi:10.2147/CCID.S111019
- Danby FW. Commentary: unroofing for hidradenitis suppurativa, why and how. J Am Acad Dermatol. 2010;63:481.e1-481.e3. doi:10.1016/j.jaad.2010.01.033
- Saylor DK, Brownstone ND, Naik HB. Office-based surgical intervention for hidradenitis suppurativa (HS): a focused review for dermatologists. Dermatol Ther (Heidelb). 2020;10:529-549. doi:10.1007/s13555-020-00391-x
Practice Gap
Hidradenitis suppurativa (HS) is a chronic inflammatory skin condition characterized by inflammatory nodules, abscesses, sinus tracts, fistulae, and scarring, mainly in intertriginous areas. The extent of disease—classified using the Hurley staging system (stages I–III)—helps guide treatment, which includes medical management and surgical intervention in later stages.
First-line treatment of HS includes topical or systemic medications, or both. Surgical therapy typically is reserved for refractory HS in moderate to severe disease (Hurley stages II and III) and is combined with pharmacotherapy. Specifically, clinical management guidelines issued by an expert committee of the United States and Canadian Hidradenitis Suppurativa Foundations recommend excision or deroofing for recurrent nodules and tunnels.1
Surgical options for HS that are available to the outpatient dermatologist include incision and drainage, electrosurgery, CO2 laser evaporation, excision, and deroofing (also known as unroofing).2 Deroofing is a fairly novel therapy; many dermatologists are unfamiliar with the procedure. A PubMed search of articles indexed for MEDLINE related to HS prior to 2010 revealed only 1 article containing the word deroofing and only 4 articles containing unroofing.
The pathophysiology of HS has important implications for successful treatment. Inflammation of the follicular pilosebaceous unit along with follicular occlusion create challenges with treatment.3 It is postulated that a defect in the glassy membrane of the infra-infundibular wall predisposes the pilosebaceous follicle to lose its structural integrality as pressure builds from plugging of the duct,4 which can result in the clinical hallmarks of HS including tunneling tracts, bridging nodules, abscesses, and fistulae that form with lateral expansion of the plugged follicle.
Leaking of the contents of these plugged follicles into surrounding tissue produces an inflammatory response in characteristic HS lesions. Because debris within the lesions moves laterally instead of being able to burst to the surface, the lesions have difficulty fully healing. Unroofing the lesions and removing built-up debris allows them to heal more expediently and quiets the underlying immune response by removing the stimulus.4
Herein, we describe the benefits, risks, and surgical process of deroofing for HS.
Technique and Tools
Deroofing is performed under local anesthesia, stepwise as follows:
1. Identify sinus tracts and infiltrate the area with lidocaine (Figure, A).
2. Use a blunt probe to define the borders of the area to be unroofed and to evaluate for any communicating sinus tracts (Figure, B).
3. Remove the roof of underlying abscesses and tracts, using a probe as a guide (Figure, C).
4. Enter through the skin or sinus opening using electrocautery or with a scalpel or scissors; perform blunt dissection.
5. Reflect back the entirety of skin overlying the probed areas and remove the skin to expose the base of the lesion (Figure, D).
6. Explore the exposed base and walls of the lesion with the probe again to assess for hidden tracts; take care not to create false tracts.
7. Debride the surgical wound using curettage or rough gauze grattage to remove remaining inflammatory debris or biofilm. To achieve hemostasis, apply aluminum chloride or ferric chloride. Coat the wound with petroleum jelly and gauze and allow it to heal by secondary intention.
8. Educate the patient on wound care—once-daily gentle cleansing with soap and water, followed by application of a moist dressing—which is similar to wound healing by secondary intention from other causes.2,4
Practice Implications
A deroofing procedure has many benefits compared to other surgical modalities for the treatment of HS. Deroofing requires only a probe, curette, and electrocautery device, making the procedure more cost-effective than excision, which requires a full tray of equipment and sutures. Furthermore, margins do not need to be taken with deroofing, and no undermining or closure is needed, which saves time during the operation and minimizes the risk for complications, including dehiscence and formation of new sinus tracts.4 No specialized equipment, such as a CO2 laser, is required, which makes deroofing accessible to every clinical dermatologist in any demographic or geographic setting.
Evidence of Benefit—Saylor and colleagues5 found that deroofing carries a 12.5% complication rate, which includes postoperative bleeding, hypergranulation tissue, and rarely wound infection. This rate is significantly lower than the 26% complication rate associated with local excision, which includes wound dehiscence, infection, and contracture (P<.001). Deroofing also was found to have an HS recurrence rate of 14.5%, which is significantly less than the 30% recurrence rate seen with local excision (P=.015). Saylor et al5 also concluded that incision and drainage was recommended only for immediate relief of HS because of its 100% recurrence rate.
van der Zee2 reported on 88 lesions from 44 patients that were treated by surgical deroofing, resulting in an average defect of 3.0 cm in length and a mean healing time of 14 days. The typical outcome was cosmetically acceptable scarring; this finding was supported by a postoperative survey (>1 year), to which 37 of 44 patients responded and assigned an average satisfaction score of 8 (of a possible 10) and a recommendation rate of 90%.2
Procedural Coding—Specific Current Procedural Terminology codes (11450-11471) from the International Classification of Diseases, Tenth Revision, exist for HS deroofing procedures; the applicable code for a given case depends on the final length of the surgical defect. Documentation to support these codes is similar to the note for an excision procedure, taking care to include location, depth, and length of the excision; healing by secondary intention; and the diagnosis of HS.
Final Thoughts
Deroofing is a surgical option that can be beneficial to patients with HS. It is a relatively simple procedure available to any dermatologist, regardless of setting. We encourage dermatologists to consider deroofing, even in patients with Hurley stage II lesions, because it can yield cosmetically acceptable and definitive results, given the variety of therapies available for HS. Deroofing also can be superior to standard excision, especially because of the potential complications with standard excision and quicker operative time with deroofing. As more providers become familiar with the deroofing procedure for HS, further studies can be undertaken to add to the paucity of data about deroofing and how it compares to other surgical treatments.
Practice Gap
Hidradenitis suppurativa (HS) is a chronic inflammatory skin condition characterized by inflammatory nodules, abscesses, sinus tracts, fistulae, and scarring, mainly in intertriginous areas. The extent of disease—classified using the Hurley staging system (stages I–III)—helps guide treatment, which includes medical management and surgical intervention in later stages.
First-line treatment of HS includes topical or systemic medications, or both. Surgical therapy typically is reserved for refractory HS in moderate to severe disease (Hurley stages II and III) and is combined with pharmacotherapy. Specifically, clinical management guidelines issued by an expert committee of the United States and Canadian Hidradenitis Suppurativa Foundations recommend excision or deroofing for recurrent nodules and tunnels.1
Surgical options for HS that are available to the outpatient dermatologist include incision and drainage, electrosurgery, CO2 laser evaporation, excision, and deroofing (also known as unroofing).2 Deroofing is a fairly novel therapy; many dermatologists are unfamiliar with the procedure. A PubMed search of articles indexed for MEDLINE related to HS prior to 2010 revealed only 1 article containing the word deroofing and only 4 articles containing unroofing.
The pathophysiology of HS has important implications for successful treatment. Inflammation of the follicular pilosebaceous unit along with follicular occlusion create challenges with treatment.3 It is postulated that a defect in the glassy membrane of the infra-infundibular wall predisposes the pilosebaceous follicle to lose its structural integrality as pressure builds from plugging of the duct,4 which can result in the clinical hallmarks of HS including tunneling tracts, bridging nodules, abscesses, and fistulae that form with lateral expansion of the plugged follicle.
Leaking of the contents of these plugged follicles into surrounding tissue produces an inflammatory response in characteristic HS lesions. Because debris within the lesions moves laterally instead of being able to burst to the surface, the lesions have difficulty fully healing. Unroofing the lesions and removing built-up debris allows them to heal more expediently and quiets the underlying immune response by removing the stimulus.4
Herein, we describe the benefits, risks, and surgical process of deroofing for HS.
Technique and Tools
Deroofing is performed under local anesthesia, stepwise as follows:
1. Identify sinus tracts and infiltrate the area with lidocaine (Figure, A).
2. Use a blunt probe to define the borders of the area to be unroofed and to evaluate for any communicating sinus tracts (Figure, B).
3. Remove the roof of underlying abscesses and tracts, using a probe as a guide (Figure, C).
4. Enter through the skin or sinus opening using electrocautery or with a scalpel or scissors; perform blunt dissection.
5. Reflect back the entirety of skin overlying the probed areas and remove the skin to expose the base of the lesion (Figure, D).
6. Explore the exposed base and walls of the lesion with the probe again to assess for hidden tracts; take care not to create false tracts.
7. Debride the surgical wound using curettage or rough gauze grattage to remove remaining inflammatory debris or biofilm. To achieve hemostasis, apply aluminum chloride or ferric chloride. Coat the wound with petroleum jelly and gauze and allow it to heal by secondary intention.
8. Educate the patient on wound care—once-daily gentle cleansing with soap and water, followed by application of a moist dressing—which is similar to wound healing by secondary intention from other causes.2,4
Practice Implications
A deroofing procedure has many benefits compared to other surgical modalities for the treatment of HS. Deroofing requires only a probe, curette, and electrocautery device, making the procedure more cost-effective than excision, which requires a full tray of equipment and sutures. Furthermore, margins do not need to be taken with deroofing, and no undermining or closure is needed, which saves time during the operation and minimizes the risk for complications, including dehiscence and formation of new sinus tracts.4 No specialized equipment, such as a CO2 laser, is required, which makes deroofing accessible to every clinical dermatologist in any demographic or geographic setting.
Evidence of Benefit—Saylor and colleagues5 found that deroofing carries a 12.5% complication rate, which includes postoperative bleeding, hypergranulation tissue, and rarely wound infection. This rate is significantly lower than the 26% complication rate associated with local excision, which includes wound dehiscence, infection, and contracture (P<.001). Deroofing also was found to have an HS recurrence rate of 14.5%, which is significantly less than the 30% recurrence rate seen with local excision (P=.015). Saylor et al5 also concluded that incision and drainage was recommended only for immediate relief of HS because of its 100% recurrence rate.
van der Zee2 reported on 88 lesions from 44 patients that were treated by surgical deroofing, resulting in an average defect of 3.0 cm in length and a mean healing time of 14 days. The typical outcome was cosmetically acceptable scarring; this finding was supported by a postoperative survey (>1 year), to which 37 of 44 patients responded and assigned an average satisfaction score of 8 (of a possible 10) and a recommendation rate of 90%.2
Procedural Coding—Specific Current Procedural Terminology codes (11450-11471) from the International Classification of Diseases, Tenth Revision, exist for HS deroofing procedures; the applicable code for a given case depends on the final length of the surgical defect. Documentation to support these codes is similar to the note for an excision procedure, taking care to include location, depth, and length of the excision; healing by secondary intention; and the diagnosis of HS.
Final Thoughts
Deroofing is a surgical option that can be beneficial to patients with HS. It is a relatively simple procedure available to any dermatologist, regardless of setting. We encourage dermatologists to consider deroofing, even in patients with Hurley stage II lesions, because it can yield cosmetically acceptable and definitive results, given the variety of therapies available for HS. Deroofing also can be superior to standard excision, especially because of the potential complications with standard excision and quicker operative time with deroofing. As more providers become familiar with the deroofing procedure for HS, further studies can be undertaken to add to the paucity of data about deroofing and how it compares to other surgical treatments.
- Alikhan A, Sayed C, Alavi A, et al. North American clinical management guidelines for hidradenitis suppurativa: a publication from the United States and Canadian Hidradenitis Suppurativa Foundations: part I: diagnosis, evaluation, and the use of complementary and procedural management. J Am Acad Dermatol. 2019;81:76-90. doi:10.1016/j.jaad.2019.02.067
- van der Zee HH, Prens EP, Boer J. Deroofing: a tissue-saving surgical technique for the treatment of mild to moderate hidradenitis suppurativa lesions. J Am Acad Dermatol. 2010;63:475-480. doi:10.1016/j.jaad.2009.12.018
- Napolitano M, Megna M, Timoshchuk EA, et al. Hidradenitis suppurativa: from pathogenesis to diagnosis and treatment. Clin Cosmet Investig Dermatol. 2017;10:105-115. doi:10.2147/CCID.S111019
- Danby FW. Commentary: unroofing for hidradenitis suppurativa, why and how. J Am Acad Dermatol. 2010;63:481.e1-481.e3. doi:10.1016/j.jaad.2010.01.033
- Saylor DK, Brownstone ND, Naik HB. Office-based surgical intervention for hidradenitis suppurativa (HS): a focused review for dermatologists. Dermatol Ther (Heidelb). 2020;10:529-549. doi:10.1007/s13555-020-00391-x
- Alikhan A, Sayed C, Alavi A, et al. North American clinical management guidelines for hidradenitis suppurativa: a publication from the United States and Canadian Hidradenitis Suppurativa Foundations: part I: diagnosis, evaluation, and the use of complementary and procedural management. J Am Acad Dermatol. 2019;81:76-90. doi:10.1016/j.jaad.2019.02.067
- van der Zee HH, Prens EP, Boer J. Deroofing: a tissue-saving surgical technique for the treatment of mild to moderate hidradenitis suppurativa lesions. J Am Acad Dermatol. 2010;63:475-480. doi:10.1016/j.jaad.2009.12.018
- Napolitano M, Megna M, Timoshchuk EA, et al. Hidradenitis suppurativa: from pathogenesis to diagnosis and treatment. Clin Cosmet Investig Dermatol. 2017;10:105-115. doi:10.2147/CCID.S111019
- Danby FW. Commentary: unroofing for hidradenitis suppurativa, why and how. J Am Acad Dermatol. 2010;63:481.e1-481.e3. doi:10.1016/j.jaad.2010.01.033
- Saylor DK, Brownstone ND, Naik HB. Office-based surgical intervention for hidradenitis suppurativa (HS): a focused review for dermatologists. Dermatol Ther (Heidelb). 2020;10:529-549. doi:10.1007/s13555-020-00391-x
COX-2, TNF inhibitor combo appear to have limited role in reducing axSpA spinal damage progression
GHENT, BELGIUM – A strong numerical signal suggests the addition of a selective cyclooxygenase-2 (COX-2) inhibitor to a tumor necrosis factor (TNF) inhibitor can reduce spinal radiographic progression in patients with active radiographic axial spondyloarthritis (axSpA) over 2 years, although results are not statistically significant.
Lead researcher and rheumatologist, Fabian Proft, MD, based at Charité University Medicine, Berlin, presented the findings of the study at the 13th International Congress on Spondyloarthritides.
Only 97 patients completed the study, and its follow-up period lasted 2 years, which is a relatively short period of time in which to determine the effects of an intervention that might affect structural progression of the spine, Dr. Proft said.
“Based on these data, I won’t treat all my patients with celecoxib,” he told this news organization. However, he added that, “If I have a patient with residual symptoms under biological DMARDs [disease-modifying antirheumatic drugs], and I feel they are at high risk of radiographic spinal progression and they still have symptoms, then I would add in an NSAID – and for that I’d choose a selective COX-2 inhibitor based on radiographic spinal progression data.”
Walter P. Maksymowych, MD, rheumatologist from the University of Alberta, Calgary, commented on the study findings in an interview. “This is an important clinical question because we want to know whether we should be adding an anti-inflammatory in patients who are on biologic therapies. There’s been a long debate and investigation into whether anti-inflammatories might prevent new bone formation and thereby prevent disease progression.”
He went on by acknowledging that there was no statistically significant difference in the primary endpoint (change in modified Stoke Ankylosing Spondylitis Spinal Score [mSASSS]) between the groups, but added that, “there was a sizable numerical difference, and I think this leaves the community somewhat hanging dry without a definitive answer. However, I do have concerns about whether there was an adequate sample size to address the study question.”
To add or not to add a selective COX-2 inhibitor to TNF inhibitor in axSpA treatment
The study aimed to investigate the effect of a selective COX-2 inhibitor when added to anti-TNF therapy with golimumab (Simponi), compared with golimumab therapy alone, on the progression of spinal structural damage over 2 years in patients with active radiographic axSpA.
“To date, we don’t have many treatments with evidence of reducing spinal radiographic progression in axSpA,” Dr. Proft said. “There was one study showing an effect of celecoxib, but another with diclofenac that failed to show any effect. As a result, there was a hypothesis that perhaps there was a selective COX-2 inhibitor effect.”
To investigate this further, Dr. Proft selected patients with high radiographic axSpA disease activity (Bath Ankylosing Spondylitis Disease Activity Index [BASDAI] ≥ 4) and with existing structural changes – both recognized risk factors for further progression. Participants had to have either an elevated C-reactive protein (CRP) > 5 mg/L and/or ≥ 1 syndesmophyte at screening, as well as a history of inadequate response to at least two DMARDs. Other patient risk factors for radiographic spinal progression included male gender and smoking. Duration of axSpA was unlimited.
Three radiographic readers were blinded for all clinical data and chronology. The primary endpoint was the change in mSASSS, while secondary endpoints were the presence of new syndesmophytes and clinical outcomes including activity, function, mobility, and health-related quality of life, as well as safety assessments.
Patients were treated with only golimumab (50 mg subcutaneous every 4 weeks) for the first 12 weeks and then only those patients with a good clinical response (n = 109) went into phase two of the study, at which point they were randomized 1:1 to golimumab monotherapy (control, 50 mg subcutaneous every 4 weeks), or golimumab (50 mg subcutaneous every 4 weeks) plus celecoxib (400 mg once daily) for 2 years. Radiographs were taken at baseline (week 0) and after 2 years. A total of 45 patients completed the combination therapy and 52 completed the monotherapy.
No statistical significance but a numerical difference found
“The primary outcome, which was change in mSASSS score, clearly shows a numerical difference between the combination arm at 1.1 and the monotherapy arm at 1.7 points, showing more structural progression in the monotherapy arm, compared to the combination arm,” Dr. Proft reported. However, he stressed that this difference did not reach statistical significance.
New syndesmophytes occurred in 25% with monotherapy and 11.1% with combination treatment. Again, this difference did not reach statistical significance.
“This might be due to sample size but also to the length of follow-up because a longer follow-up [given structural changes occur relatively slowly] might have shown a greater difference,” Dr. Proft pointed out.
Clinical data, according to Ankylosing Spondylitis Disease Activity Score with CRP and BASDAI, showed that both groups responded very well to therapy, and there were no differences seen between the two groups in terms of clinical parameters.
“It is important when we add a drug – and we know that NSAIDs can have safety concerns – that we do not see any statistically significant serious adverse events between patient groups,” Dr. Proft noted.
There were no significant differences in adverse events between monotherapy and combination therapy. There were 162 infections in the combination arm and 150 in the monotherapy arm. Combination therapy led to seven serious adverse events, and monotherapy occurred with five adverse events.
Dr. Proft added that four patients discontinued in the combination arm, compared with only one in the monotherapy arm, with a variety of different reasons for the discontinuations.
The study was supported by a grant from the German Ministry of Education and Research, and golimumab was provided free of charge by Merck Sharp & Dohme. Dr. Proft reported serving on speakers bureaus for Amgen, AbbVie, Bristol-Myers Squibb, Celgene, Janssen, Merck Sharp & Dohme, Novartis, Pfizer, Roche, and UCB; serving as a consultant to Novartis; and receiving grant or research support from Novartis, UCB, and Lilly. Dr. Maksymowych declared having no relevant conflicts of interest.
GHENT, BELGIUM – A strong numerical signal suggests the addition of a selective cyclooxygenase-2 (COX-2) inhibitor to a tumor necrosis factor (TNF) inhibitor can reduce spinal radiographic progression in patients with active radiographic axial spondyloarthritis (axSpA) over 2 years, although results are not statistically significant.
Lead researcher and rheumatologist, Fabian Proft, MD, based at Charité University Medicine, Berlin, presented the findings of the study at the 13th International Congress on Spondyloarthritides.
Only 97 patients completed the study, and its follow-up period lasted 2 years, which is a relatively short period of time in which to determine the effects of an intervention that might affect structural progression of the spine, Dr. Proft said.
“Based on these data, I won’t treat all my patients with celecoxib,” he told this news organization. However, he added that, “If I have a patient with residual symptoms under biological DMARDs [disease-modifying antirheumatic drugs], and I feel they are at high risk of radiographic spinal progression and they still have symptoms, then I would add in an NSAID – and for that I’d choose a selective COX-2 inhibitor based on radiographic spinal progression data.”
Walter P. Maksymowych, MD, rheumatologist from the University of Alberta, Calgary, commented on the study findings in an interview. “This is an important clinical question because we want to know whether we should be adding an anti-inflammatory in patients who are on biologic therapies. There’s been a long debate and investigation into whether anti-inflammatories might prevent new bone formation and thereby prevent disease progression.”
He went on by acknowledging that there was no statistically significant difference in the primary endpoint (change in modified Stoke Ankylosing Spondylitis Spinal Score [mSASSS]) between the groups, but added that, “there was a sizable numerical difference, and I think this leaves the community somewhat hanging dry without a definitive answer. However, I do have concerns about whether there was an adequate sample size to address the study question.”
To add or not to add a selective COX-2 inhibitor to TNF inhibitor in axSpA treatment
The study aimed to investigate the effect of a selective COX-2 inhibitor when added to anti-TNF therapy with golimumab (Simponi), compared with golimumab therapy alone, on the progression of spinal structural damage over 2 years in patients with active radiographic axSpA.
“To date, we don’t have many treatments with evidence of reducing spinal radiographic progression in axSpA,” Dr. Proft said. “There was one study showing an effect of celecoxib, but another with diclofenac that failed to show any effect. As a result, there was a hypothesis that perhaps there was a selective COX-2 inhibitor effect.”
To investigate this further, Dr. Proft selected patients with high radiographic axSpA disease activity (Bath Ankylosing Spondylitis Disease Activity Index [BASDAI] ≥ 4) and with existing structural changes – both recognized risk factors for further progression. Participants had to have either an elevated C-reactive protein (CRP) > 5 mg/L and/or ≥ 1 syndesmophyte at screening, as well as a history of inadequate response to at least two DMARDs. Other patient risk factors for radiographic spinal progression included male gender and smoking. Duration of axSpA was unlimited.
Three radiographic readers were blinded for all clinical data and chronology. The primary endpoint was the change in mSASSS, while secondary endpoints were the presence of new syndesmophytes and clinical outcomes including activity, function, mobility, and health-related quality of life, as well as safety assessments.
Patients were treated with only golimumab (50 mg subcutaneous every 4 weeks) for the first 12 weeks and then only those patients with a good clinical response (n = 109) went into phase two of the study, at which point they were randomized 1:1 to golimumab monotherapy (control, 50 mg subcutaneous every 4 weeks), or golimumab (50 mg subcutaneous every 4 weeks) plus celecoxib (400 mg once daily) for 2 years. Radiographs were taken at baseline (week 0) and after 2 years. A total of 45 patients completed the combination therapy and 52 completed the monotherapy.
No statistical significance but a numerical difference found
“The primary outcome, which was change in mSASSS score, clearly shows a numerical difference between the combination arm at 1.1 and the monotherapy arm at 1.7 points, showing more structural progression in the monotherapy arm, compared to the combination arm,” Dr. Proft reported. However, he stressed that this difference did not reach statistical significance.
New syndesmophytes occurred in 25% with monotherapy and 11.1% with combination treatment. Again, this difference did not reach statistical significance.
“This might be due to sample size but also to the length of follow-up because a longer follow-up [given structural changes occur relatively slowly] might have shown a greater difference,” Dr. Proft pointed out.
Clinical data, according to Ankylosing Spondylitis Disease Activity Score with CRP and BASDAI, showed that both groups responded very well to therapy, and there were no differences seen between the two groups in terms of clinical parameters.
“It is important when we add a drug – and we know that NSAIDs can have safety concerns – that we do not see any statistically significant serious adverse events between patient groups,” Dr. Proft noted.
There were no significant differences in adverse events between monotherapy and combination therapy. There were 162 infections in the combination arm and 150 in the monotherapy arm. Combination therapy led to seven serious adverse events, and monotherapy occurred with five adverse events.
Dr. Proft added that four patients discontinued in the combination arm, compared with only one in the monotherapy arm, with a variety of different reasons for the discontinuations.
The study was supported by a grant from the German Ministry of Education and Research, and golimumab was provided free of charge by Merck Sharp & Dohme. Dr. Proft reported serving on speakers bureaus for Amgen, AbbVie, Bristol-Myers Squibb, Celgene, Janssen, Merck Sharp & Dohme, Novartis, Pfizer, Roche, and UCB; serving as a consultant to Novartis; and receiving grant or research support from Novartis, UCB, and Lilly. Dr. Maksymowych declared having no relevant conflicts of interest.
GHENT, BELGIUM – A strong numerical signal suggests the addition of a selective cyclooxygenase-2 (COX-2) inhibitor to a tumor necrosis factor (TNF) inhibitor can reduce spinal radiographic progression in patients with active radiographic axial spondyloarthritis (axSpA) over 2 years, although results are not statistically significant.
Lead researcher and rheumatologist, Fabian Proft, MD, based at Charité University Medicine, Berlin, presented the findings of the study at the 13th International Congress on Spondyloarthritides.
Only 97 patients completed the study, and its follow-up period lasted 2 years, which is a relatively short period of time in which to determine the effects of an intervention that might affect structural progression of the spine, Dr. Proft said.
“Based on these data, I won’t treat all my patients with celecoxib,” he told this news organization. However, he added that, “If I have a patient with residual symptoms under biological DMARDs [disease-modifying antirheumatic drugs], and I feel they are at high risk of radiographic spinal progression and they still have symptoms, then I would add in an NSAID – and for that I’d choose a selective COX-2 inhibitor based on radiographic spinal progression data.”
Walter P. Maksymowych, MD, rheumatologist from the University of Alberta, Calgary, commented on the study findings in an interview. “This is an important clinical question because we want to know whether we should be adding an anti-inflammatory in patients who are on biologic therapies. There’s been a long debate and investigation into whether anti-inflammatories might prevent new bone formation and thereby prevent disease progression.”
He went on by acknowledging that there was no statistically significant difference in the primary endpoint (change in modified Stoke Ankylosing Spondylitis Spinal Score [mSASSS]) between the groups, but added that, “there was a sizable numerical difference, and I think this leaves the community somewhat hanging dry without a definitive answer. However, I do have concerns about whether there was an adequate sample size to address the study question.”
To add or not to add a selective COX-2 inhibitor to TNF inhibitor in axSpA treatment
The study aimed to investigate the effect of a selective COX-2 inhibitor when added to anti-TNF therapy with golimumab (Simponi), compared with golimumab therapy alone, on the progression of spinal structural damage over 2 years in patients with active radiographic axSpA.
“To date, we don’t have many treatments with evidence of reducing spinal radiographic progression in axSpA,” Dr. Proft said. “There was one study showing an effect of celecoxib, but another with diclofenac that failed to show any effect. As a result, there was a hypothesis that perhaps there was a selective COX-2 inhibitor effect.”
To investigate this further, Dr. Proft selected patients with high radiographic axSpA disease activity (Bath Ankylosing Spondylitis Disease Activity Index [BASDAI] ≥ 4) and with existing structural changes – both recognized risk factors for further progression. Participants had to have either an elevated C-reactive protein (CRP) > 5 mg/L and/or ≥ 1 syndesmophyte at screening, as well as a history of inadequate response to at least two DMARDs. Other patient risk factors for radiographic spinal progression included male gender and smoking. Duration of axSpA was unlimited.
Three radiographic readers were blinded for all clinical data and chronology. The primary endpoint was the change in mSASSS, while secondary endpoints were the presence of new syndesmophytes and clinical outcomes including activity, function, mobility, and health-related quality of life, as well as safety assessments.
Patients were treated with only golimumab (50 mg subcutaneous every 4 weeks) for the first 12 weeks and then only those patients with a good clinical response (n = 109) went into phase two of the study, at which point they were randomized 1:1 to golimumab monotherapy (control, 50 mg subcutaneous every 4 weeks), or golimumab (50 mg subcutaneous every 4 weeks) plus celecoxib (400 mg once daily) for 2 years. Radiographs were taken at baseline (week 0) and after 2 years. A total of 45 patients completed the combination therapy and 52 completed the monotherapy.
No statistical significance but a numerical difference found
“The primary outcome, which was change in mSASSS score, clearly shows a numerical difference between the combination arm at 1.1 and the monotherapy arm at 1.7 points, showing more structural progression in the monotherapy arm, compared to the combination arm,” Dr. Proft reported. However, he stressed that this difference did not reach statistical significance.
New syndesmophytes occurred in 25% with monotherapy and 11.1% with combination treatment. Again, this difference did not reach statistical significance.
“This might be due to sample size but also to the length of follow-up because a longer follow-up [given structural changes occur relatively slowly] might have shown a greater difference,” Dr. Proft pointed out.
Clinical data, according to Ankylosing Spondylitis Disease Activity Score with CRP and BASDAI, showed that both groups responded very well to therapy, and there were no differences seen between the two groups in terms of clinical parameters.
“It is important when we add a drug – and we know that NSAIDs can have safety concerns – that we do not see any statistically significant serious adverse events between patient groups,” Dr. Proft noted.
There were no significant differences in adverse events between monotherapy and combination therapy. There were 162 infections in the combination arm and 150 in the monotherapy arm. Combination therapy led to seven serious adverse events, and monotherapy occurred with five adverse events.
Dr. Proft added that four patients discontinued in the combination arm, compared with only one in the monotherapy arm, with a variety of different reasons for the discontinuations.
The study was supported by a grant from the German Ministry of Education and Research, and golimumab was provided free of charge by Merck Sharp & Dohme. Dr. Proft reported serving on speakers bureaus for Amgen, AbbVie, Bristol-Myers Squibb, Celgene, Janssen, Merck Sharp & Dohme, Novartis, Pfizer, Roche, and UCB; serving as a consultant to Novartis; and receiving grant or research support from Novartis, UCB, and Lilly. Dr. Maksymowych declared having no relevant conflicts of interest.
AT THE 2022 SPA CONGRESS
Polypodium leucotomos found to reverse AK skin damage
MILAN – Application of topical or both treated over 12 months, in a randomized, blinded study presented at the annual congress of the European Academy of Dermatology and Venereology.
At 12 months, the percentage of patients with a normal or almost normal honeycomb pattern when evaluated blindly with reflectance confocal microscopy (RCM) was about twice as great in either of the two groups that received PLE relative to those treated with topical photoprotection alone, according to Giovanni Pellacani, MD, PhD, chair of dermatology, University of Sapienza, Rome.
“In patients with severe actinic keratosis, the 12-month use of a PLE-based topical or oral photoprotection is associated with positive clinical and anatomical outcomes,” Dr. Pellacani said.
PLE, which is already commonly used in sun protection products, is derived from a South American species of fern and has been proposed for a broad array of dermatologic diseases. According to Dr. Pellacani, in vivo studies associating PLE with immune photoprotection make this agent particularly promising for severe AKs.
In this study involving two clinical research centers in Italy, 131 patients with photoaging and at least three AKs were randomized to one of three treatment arms. The control arm received topical photoprotection with an SPF of 100 or higher applied twice daily to all sun-exposed areas. The two treatment arms received the same topical photoprotection plus either a PLE-containing topical cream alone or a PLE-containing topical cream plus PLE in an oral form (240 mg) once daily
Patients were evaluated at 3 months, 6 months, and 1 year with several measures, including the Actinic Keratosis Area Score Index (AKASI) and the AK Field Assessment Scale Area (AK-FAS). They were also assessed with RCM. All clinical assessments and RCM evaluations, which assessed seven different parameters, such as honeycomb pattern, mottled pigmentation, and reticulated collagen, were performed by dermatologists blinded to the treatment assignment.
Complete data were available for 116 patients who completed all three evaluations over the 12 months of follow-up. On RCM, 50% of those receiving the oral and topical forms of PLE and 45% of those receiving topical PLE had normalization of the honeycomb pattern. These responses were significantly greater (P = .04 for both) than the 26% with normalization in the control group.
Although there were no significant differences in any of the other parameters evaluated by RCM, the improvement in the honeycomb pattern was accompanied by a 7% improvement in the AKASI score in patients taking PLE, either topically or orally and topically, while there was a 6% worsening (P < .001) among controls.
The AK-FAS score improved at 12 months by 26% in the group on oral/topical PLE and by 4% in the group on topical PLE. The score worsened by 13% among controls.
Over the course of the study, patients were permitted to take an appropriate therapy, such as imiquimod, cryotherapy, or 5-flourouracil if there was worsening of the AK-FAS score or if new lesions appeared.
On this measure, 38% of controls and 11% of those randomized to topical PLE had progressive disease versus only 2% of those randomized to take both topical and oral PLE, Dr. Pellacani reported.
The lower rate of new lesions or a start of a new drug over the course of the study in the group receiving both the topical and the oral formulations of PLE relative to those receiving topical PLE alone did not reach statistical significance, but Dr. Pellacani concluded that the addition of PLE to topical photoprotection without PLE seemed to provide a potentially clinically meaningful advantage.
Larger studies and longer term studies are needed, according to Dr. Pellacani, who noted that the substantial body of clinical studies associating PLE with benefit in a variety of dermatologic disorders has been weakened by the absence of well-designed studies that are adequately powered to guide clinical use.
Salvador González, MD, PhD, a dermatology specialist at Alcalá University, Madrid, also believes that PLE deserves further evaluation not just for photoprotection but for reinvigorating damaged skin due to its antioxidant and anti-inflammatory properties. He was the senior author of a 2020 paper in Photochemical and Photobiological Sciences that summarized the potential benefits of PLE in preventing damage related to sun exposure.
Among its mechanism, PLE generates reactive oxygen species (ROS) and prevents depletion of Langerhans cells induced by ultraviolet (UV) light, Dr. González explained in an interview. “At the cellular level, PLE activates tumor suppression p53, inhibits UV-induced COX-2 expression, reduces inflammation, and preventions immunosuppression,” he continued. In addition, he said PLE also prevents UV-A-induced common deletions related to mitochondrial damage and MMP1 expression induced by various UV wavelengths.
“These molecular and cellular effects may translate into long-term inhibition of carcinogenesis including actinic keratosis,” he said, noting that all of these findings “justify the work by Pellacani and collaborators.”
Dr. Pellacani reports no potential conflicts of interest. Dr. González has a financial relationship with Cantabria Laboratories.
MILAN – Application of topical or both treated over 12 months, in a randomized, blinded study presented at the annual congress of the European Academy of Dermatology and Venereology.
At 12 months, the percentage of patients with a normal or almost normal honeycomb pattern when evaluated blindly with reflectance confocal microscopy (RCM) was about twice as great in either of the two groups that received PLE relative to those treated with topical photoprotection alone, according to Giovanni Pellacani, MD, PhD, chair of dermatology, University of Sapienza, Rome.
“In patients with severe actinic keratosis, the 12-month use of a PLE-based topical or oral photoprotection is associated with positive clinical and anatomical outcomes,” Dr. Pellacani said.
PLE, which is already commonly used in sun protection products, is derived from a South American species of fern and has been proposed for a broad array of dermatologic diseases. According to Dr. Pellacani, in vivo studies associating PLE with immune photoprotection make this agent particularly promising for severe AKs.
In this study involving two clinical research centers in Italy, 131 patients with photoaging and at least three AKs were randomized to one of three treatment arms. The control arm received topical photoprotection with an SPF of 100 or higher applied twice daily to all sun-exposed areas. The two treatment arms received the same topical photoprotection plus either a PLE-containing topical cream alone or a PLE-containing topical cream plus PLE in an oral form (240 mg) once daily
Patients were evaluated at 3 months, 6 months, and 1 year with several measures, including the Actinic Keratosis Area Score Index (AKASI) and the AK Field Assessment Scale Area (AK-FAS). They were also assessed with RCM. All clinical assessments and RCM evaluations, which assessed seven different parameters, such as honeycomb pattern, mottled pigmentation, and reticulated collagen, were performed by dermatologists blinded to the treatment assignment.
Complete data were available for 116 patients who completed all three evaluations over the 12 months of follow-up. On RCM, 50% of those receiving the oral and topical forms of PLE and 45% of those receiving topical PLE had normalization of the honeycomb pattern. These responses were significantly greater (P = .04 for both) than the 26% with normalization in the control group.
Although there were no significant differences in any of the other parameters evaluated by RCM, the improvement in the honeycomb pattern was accompanied by a 7% improvement in the AKASI score in patients taking PLE, either topically or orally and topically, while there was a 6% worsening (P < .001) among controls.
The AK-FAS score improved at 12 months by 26% in the group on oral/topical PLE and by 4% in the group on topical PLE. The score worsened by 13% among controls.
Over the course of the study, patients were permitted to take an appropriate therapy, such as imiquimod, cryotherapy, or 5-flourouracil if there was worsening of the AK-FAS score or if new lesions appeared.
On this measure, 38% of controls and 11% of those randomized to topical PLE had progressive disease versus only 2% of those randomized to take both topical and oral PLE, Dr. Pellacani reported.
The lower rate of new lesions or a start of a new drug over the course of the study in the group receiving both the topical and the oral formulations of PLE relative to those receiving topical PLE alone did not reach statistical significance, but Dr. Pellacani concluded that the addition of PLE to topical photoprotection without PLE seemed to provide a potentially clinically meaningful advantage.
Larger studies and longer term studies are needed, according to Dr. Pellacani, who noted that the substantial body of clinical studies associating PLE with benefit in a variety of dermatologic disorders has been weakened by the absence of well-designed studies that are adequately powered to guide clinical use.
Salvador González, MD, PhD, a dermatology specialist at Alcalá University, Madrid, also believes that PLE deserves further evaluation not just for photoprotection but for reinvigorating damaged skin due to its antioxidant and anti-inflammatory properties. He was the senior author of a 2020 paper in Photochemical and Photobiological Sciences that summarized the potential benefits of PLE in preventing damage related to sun exposure.
Among its mechanism, PLE generates reactive oxygen species (ROS) and prevents depletion of Langerhans cells induced by ultraviolet (UV) light, Dr. González explained in an interview. “At the cellular level, PLE activates tumor suppression p53, inhibits UV-induced COX-2 expression, reduces inflammation, and preventions immunosuppression,” he continued. In addition, he said PLE also prevents UV-A-induced common deletions related to mitochondrial damage and MMP1 expression induced by various UV wavelengths.
“These molecular and cellular effects may translate into long-term inhibition of carcinogenesis including actinic keratosis,” he said, noting that all of these findings “justify the work by Pellacani and collaborators.”
Dr. Pellacani reports no potential conflicts of interest. Dr. González has a financial relationship with Cantabria Laboratories.
MILAN – Application of topical or both treated over 12 months, in a randomized, blinded study presented at the annual congress of the European Academy of Dermatology and Venereology.
At 12 months, the percentage of patients with a normal or almost normal honeycomb pattern when evaluated blindly with reflectance confocal microscopy (RCM) was about twice as great in either of the two groups that received PLE relative to those treated with topical photoprotection alone, according to Giovanni Pellacani, MD, PhD, chair of dermatology, University of Sapienza, Rome.
“In patients with severe actinic keratosis, the 12-month use of a PLE-based topical or oral photoprotection is associated with positive clinical and anatomical outcomes,” Dr. Pellacani said.
PLE, which is already commonly used in sun protection products, is derived from a South American species of fern and has been proposed for a broad array of dermatologic diseases. According to Dr. Pellacani, in vivo studies associating PLE with immune photoprotection make this agent particularly promising for severe AKs.
In this study involving two clinical research centers in Italy, 131 patients with photoaging and at least three AKs were randomized to one of three treatment arms. The control arm received topical photoprotection with an SPF of 100 or higher applied twice daily to all sun-exposed areas. The two treatment arms received the same topical photoprotection plus either a PLE-containing topical cream alone or a PLE-containing topical cream plus PLE in an oral form (240 mg) once daily
Patients were evaluated at 3 months, 6 months, and 1 year with several measures, including the Actinic Keratosis Area Score Index (AKASI) and the AK Field Assessment Scale Area (AK-FAS). They were also assessed with RCM. All clinical assessments and RCM evaluations, which assessed seven different parameters, such as honeycomb pattern, mottled pigmentation, and reticulated collagen, were performed by dermatologists blinded to the treatment assignment.
Complete data were available for 116 patients who completed all three evaluations over the 12 months of follow-up. On RCM, 50% of those receiving the oral and topical forms of PLE and 45% of those receiving topical PLE had normalization of the honeycomb pattern. These responses were significantly greater (P = .04 for both) than the 26% with normalization in the control group.
Although there were no significant differences in any of the other parameters evaluated by RCM, the improvement in the honeycomb pattern was accompanied by a 7% improvement in the AKASI score in patients taking PLE, either topically or orally and topically, while there was a 6% worsening (P < .001) among controls.
The AK-FAS score improved at 12 months by 26% in the group on oral/topical PLE and by 4% in the group on topical PLE. The score worsened by 13% among controls.
Over the course of the study, patients were permitted to take an appropriate therapy, such as imiquimod, cryotherapy, or 5-flourouracil if there was worsening of the AK-FAS score or if new lesions appeared.
On this measure, 38% of controls and 11% of those randomized to topical PLE had progressive disease versus only 2% of those randomized to take both topical and oral PLE, Dr. Pellacani reported.
The lower rate of new lesions or a start of a new drug over the course of the study in the group receiving both the topical and the oral formulations of PLE relative to those receiving topical PLE alone did not reach statistical significance, but Dr. Pellacani concluded that the addition of PLE to topical photoprotection without PLE seemed to provide a potentially clinically meaningful advantage.
Larger studies and longer term studies are needed, according to Dr. Pellacani, who noted that the substantial body of clinical studies associating PLE with benefit in a variety of dermatologic disorders has been weakened by the absence of well-designed studies that are adequately powered to guide clinical use.
Salvador González, MD, PhD, a dermatology specialist at Alcalá University, Madrid, also believes that PLE deserves further evaluation not just for photoprotection but for reinvigorating damaged skin due to its antioxidant and anti-inflammatory properties. He was the senior author of a 2020 paper in Photochemical and Photobiological Sciences that summarized the potential benefits of PLE in preventing damage related to sun exposure.
Among its mechanism, PLE generates reactive oxygen species (ROS) and prevents depletion of Langerhans cells induced by ultraviolet (UV) light, Dr. González explained in an interview. “At the cellular level, PLE activates tumor suppression p53, inhibits UV-induced COX-2 expression, reduces inflammation, and preventions immunosuppression,” he continued. In addition, he said PLE also prevents UV-A-induced common deletions related to mitochondrial damage and MMP1 expression induced by various UV wavelengths.
“These molecular and cellular effects may translate into long-term inhibition of carcinogenesis including actinic keratosis,” he said, noting that all of these findings “justify the work by Pellacani and collaborators.”
Dr. Pellacani reports no potential conflicts of interest. Dr. González has a financial relationship with Cantabria Laboratories.
AT THE EADV CONGRESS
No invasive strategy benefit at 5 years in ISCHEMIA-CKD extension study
A trip to the cath lab for possible revascularization after a positive stress test, compared with a wait-and-see approach backed by optimal medications, did not improve 5-year survival for patients with advanced chronic kidney disease (CKD) in the ISCHEMIA-CKD trial’s extension study, ISCHEMIA-CKD EXTEND.
The long-term results, from the same 777 patients followed for an average of 2.2 years in the main trial, are consistent with the overall findings of no survival advantage with an initially invasive strategy, compared with one that is initially conservative. The finding applies to patients like those in the trial who had moderate to severe ischemia at stress testing and whose CKD put them in an especially high-risk and little-studied coronary artery disease (CAD) category.
Indeed, in a reflection of that high-risk status, 5-year all-cause mortality reached about 40% and cardiovascular (CV) mortality approached 30%, with no significant differences between patients in the invasive- and conservative-strategy groups.
Those numbers arguably put CKD’s effect on CAD survival in about the same league as an ejection fraction (EF) of 35% or less. For context, all-cause mortality over 3-4 years was about 32% in the REVIVED-BCIS2 trial of such patients with ischemic reduced-EF cardiomyopathy, whether or not they were revascularized, observed Sripal Bangalore, MD, MHA.
Yet in ISCHEMIA-CKD EXTEND, “you’re seeing in a group of patients, with largely preserved EF but advanced CKD, a mortality rate close to 40% at 5 years,” said Dr. Bangalore of New York University.
Although the study doesn’t show benefit from the initially invasive approach in CKD patients with stable CAD, for those with acute coronary syndromes (ACS), it seems to suggest that “at least the invasive strategy is safe,” Dr. Bangalore said during a press conference preceding his presentation of the study Aug. 29 at the annual congress of the European Society of Cardiology, held in Barcelona.
REVIVED-BCIS2 was also presented at the ESC sessions on Aug. 27, as reported by this news organization.
ISCHEMIA-CKD EXTEND “is a large trial and a very well-done trial. The results are robust, and they should influence clinical practice,” Deepak L. Bhatt, MD, MPH, Brigham and Women’s Hospital Heart & Vascular Center, Boston, said as the invited discussant after Dr. Bangalore’s presentation.
“The main message here, really, is don’t just go looking for ischemia, at least with the modalities used in this trial, in your CKD patients as a routine practice, and then try to stomp out that ischemia with revascularization,” Dr. Bhatt said. “The right thing to do in these high-risk patients is to focus on lifestyle modification and intensive medical therapy.”
A caveat, he said, is that the trial’s results don’t apply to the types of patients excluded from it, including those with recent ACS and those who are highly symptomatic or have an EF of less than 35%.
“Those CKD patients likely benefit from an invasive strategy with anatomically appropriate revascularization,” whether percutaneous coronary intervention (PCI) or coronary bypass surgery, Dr. Bhatt said.
At a median follow-up of 5 years in the extension study, the rates of death from any cause were 40.6% for patients in the invasive-strategy group and 37.4% for those in the conservative-strategy group. That yielded a hazard ratio of 1.12 (95% confidence interval, 0.89-1.41; P = .32) after adjustment for age, sex, diabetes status, EF, dialysis status, and – for patients not on dialysis – baseline estimated glomerular filtration rate.
The rates of CV death were 29% for patients managed invasively and 27% for those initially managed conservatively, for a similarly adjusted HR of 1.04 (95% CI, 0.80-1.37; P = .75).
In subgroup analyses, Dr. Bangalore reported, there were no significant differences in all-cause or CV mortality by diabetes status, by severity of baseline ischemia, or by whether the patient had recently experienced new or more frequent angina at study entry, was on guideline-directed medical therapy at baseline, or was on dialysis.
Among the contributions of ISCHEMIA-CKD and its 5-year extension study, Dr. Bangalore told this news organization, is that the relative safety of revascularization they showed may help to counter “renalism,” that is, the aversion to invasive intervention in patients with advanced CKD in clinical practice.
For example, if a patient with advanced CKD presents with an acute myocardial infarction, “people are hesitant to take them to the cath lab,” Dr. Bangalore said. But “if you follow protocols, if you follow strategies to minimize the risk, you can safely go ahead and do it.”
But in patients with stable CAD, as the ISCHEMIA-CKD studies show, “routinely revascularizing them may not have significant benefits.”
ISCHEMIC-CKD and its extension study were funded by the National Heart, Lung, and Blood Institute. Dr. Bangalore discloses receiving research grants from NHLBI and serving as a consultant for Abbott Vascular, Biotronik, Boston Scientific, Amgen, Pfizer, Merck, and Reata. Dr. Bhatt has disclosed grants and/or personal fees from many companies; personal fees from WebMD and other publications or organizations; and having other relationships with Medscape Cardiology and other publications or organizations.
A version of this article first appeared on Medscape.com.
A trip to the cath lab for possible revascularization after a positive stress test, compared with a wait-and-see approach backed by optimal medications, did not improve 5-year survival for patients with advanced chronic kidney disease (CKD) in the ISCHEMIA-CKD trial’s extension study, ISCHEMIA-CKD EXTEND.
The long-term results, from the same 777 patients followed for an average of 2.2 years in the main trial, are consistent with the overall findings of no survival advantage with an initially invasive strategy, compared with one that is initially conservative. The finding applies to patients like those in the trial who had moderate to severe ischemia at stress testing and whose CKD put them in an especially high-risk and little-studied coronary artery disease (CAD) category.
Indeed, in a reflection of that high-risk status, 5-year all-cause mortality reached about 40% and cardiovascular (CV) mortality approached 30%, with no significant differences between patients in the invasive- and conservative-strategy groups.
Those numbers arguably put CKD’s effect on CAD survival in about the same league as an ejection fraction (EF) of 35% or less. For context, all-cause mortality over 3-4 years was about 32% in the REVIVED-BCIS2 trial of such patients with ischemic reduced-EF cardiomyopathy, whether or not they were revascularized, observed Sripal Bangalore, MD, MHA.
Yet in ISCHEMIA-CKD EXTEND, “you’re seeing in a group of patients, with largely preserved EF but advanced CKD, a mortality rate close to 40% at 5 years,” said Dr. Bangalore of New York University.
Although the study doesn’t show benefit from the initially invasive approach in CKD patients with stable CAD, for those with acute coronary syndromes (ACS), it seems to suggest that “at least the invasive strategy is safe,” Dr. Bangalore said during a press conference preceding his presentation of the study Aug. 29 at the annual congress of the European Society of Cardiology, held in Barcelona.
REVIVED-BCIS2 was also presented at the ESC sessions on Aug. 27, as reported by this news organization.
ISCHEMIA-CKD EXTEND “is a large trial and a very well-done trial. The results are robust, and they should influence clinical practice,” Deepak L. Bhatt, MD, MPH, Brigham and Women’s Hospital Heart & Vascular Center, Boston, said as the invited discussant after Dr. Bangalore’s presentation.
“The main message here, really, is don’t just go looking for ischemia, at least with the modalities used in this trial, in your CKD patients as a routine practice, and then try to stomp out that ischemia with revascularization,” Dr. Bhatt said. “The right thing to do in these high-risk patients is to focus on lifestyle modification and intensive medical therapy.”
A caveat, he said, is that the trial’s results don’t apply to the types of patients excluded from it, including those with recent ACS and those who are highly symptomatic or have an EF of less than 35%.
“Those CKD patients likely benefit from an invasive strategy with anatomically appropriate revascularization,” whether percutaneous coronary intervention (PCI) or coronary bypass surgery, Dr. Bhatt said.
At a median follow-up of 5 years in the extension study, the rates of death from any cause were 40.6% for patients in the invasive-strategy group and 37.4% for those in the conservative-strategy group. That yielded a hazard ratio of 1.12 (95% confidence interval, 0.89-1.41; P = .32) after adjustment for age, sex, diabetes status, EF, dialysis status, and – for patients not on dialysis – baseline estimated glomerular filtration rate.
The rates of CV death were 29% for patients managed invasively and 27% for those initially managed conservatively, for a similarly adjusted HR of 1.04 (95% CI, 0.80-1.37; P = .75).
In subgroup analyses, Dr. Bangalore reported, there were no significant differences in all-cause or CV mortality by diabetes status, by severity of baseline ischemia, or by whether the patient had recently experienced new or more frequent angina at study entry, was on guideline-directed medical therapy at baseline, or was on dialysis.
Among the contributions of ISCHEMIA-CKD and its 5-year extension study, Dr. Bangalore told this news organization, is that the relative safety of revascularization they showed may help to counter “renalism,” that is, the aversion to invasive intervention in patients with advanced CKD in clinical practice.
For example, if a patient with advanced CKD presents with an acute myocardial infarction, “people are hesitant to take them to the cath lab,” Dr. Bangalore said. But “if you follow protocols, if you follow strategies to minimize the risk, you can safely go ahead and do it.”
But in patients with stable CAD, as the ISCHEMIA-CKD studies show, “routinely revascularizing them may not have significant benefits.”
ISCHEMIC-CKD and its extension study were funded by the National Heart, Lung, and Blood Institute. Dr. Bangalore discloses receiving research grants from NHLBI and serving as a consultant for Abbott Vascular, Biotronik, Boston Scientific, Amgen, Pfizer, Merck, and Reata. Dr. Bhatt has disclosed grants and/or personal fees from many companies; personal fees from WebMD and other publications or organizations; and having other relationships with Medscape Cardiology and other publications or organizations.
A version of this article first appeared on Medscape.com.
A trip to the cath lab for possible revascularization after a positive stress test, compared with a wait-and-see approach backed by optimal medications, did not improve 5-year survival for patients with advanced chronic kidney disease (CKD) in the ISCHEMIA-CKD trial’s extension study, ISCHEMIA-CKD EXTEND.
The long-term results, from the same 777 patients followed for an average of 2.2 years in the main trial, are consistent with the overall findings of no survival advantage with an initially invasive strategy, compared with one that is initially conservative. The finding applies to patients like those in the trial who had moderate to severe ischemia at stress testing and whose CKD put them in an especially high-risk and little-studied coronary artery disease (CAD) category.
Indeed, in a reflection of that high-risk status, 5-year all-cause mortality reached about 40% and cardiovascular (CV) mortality approached 30%, with no significant differences between patients in the invasive- and conservative-strategy groups.
Those numbers arguably put CKD’s effect on CAD survival in about the same league as an ejection fraction (EF) of 35% or less. For context, all-cause mortality over 3-4 years was about 32% in the REVIVED-BCIS2 trial of such patients with ischemic reduced-EF cardiomyopathy, whether or not they were revascularized, observed Sripal Bangalore, MD, MHA.
Yet in ISCHEMIA-CKD EXTEND, “you’re seeing in a group of patients, with largely preserved EF but advanced CKD, a mortality rate close to 40% at 5 years,” said Dr. Bangalore of New York University.
Although the study doesn’t show benefit from the initially invasive approach in CKD patients with stable CAD, for those with acute coronary syndromes (ACS), it seems to suggest that “at least the invasive strategy is safe,” Dr. Bangalore said during a press conference preceding his presentation of the study Aug. 29 at the annual congress of the European Society of Cardiology, held in Barcelona.
REVIVED-BCIS2 was also presented at the ESC sessions on Aug. 27, as reported by this news organization.
ISCHEMIA-CKD EXTEND “is a large trial and a very well-done trial. The results are robust, and they should influence clinical practice,” Deepak L. Bhatt, MD, MPH, Brigham and Women’s Hospital Heart & Vascular Center, Boston, said as the invited discussant after Dr. Bangalore’s presentation.
“The main message here, really, is don’t just go looking for ischemia, at least with the modalities used in this trial, in your CKD patients as a routine practice, and then try to stomp out that ischemia with revascularization,” Dr. Bhatt said. “The right thing to do in these high-risk patients is to focus on lifestyle modification and intensive medical therapy.”
A caveat, he said, is that the trial’s results don’t apply to the types of patients excluded from it, including those with recent ACS and those who are highly symptomatic or have an EF of less than 35%.
“Those CKD patients likely benefit from an invasive strategy with anatomically appropriate revascularization,” whether percutaneous coronary intervention (PCI) or coronary bypass surgery, Dr. Bhatt said.
At a median follow-up of 5 years in the extension study, the rates of death from any cause were 40.6% for patients in the invasive-strategy group and 37.4% for those in the conservative-strategy group. That yielded a hazard ratio of 1.12 (95% confidence interval, 0.89-1.41; P = .32) after adjustment for age, sex, diabetes status, EF, dialysis status, and – for patients not on dialysis – baseline estimated glomerular filtration rate.
The rates of CV death were 29% for patients managed invasively and 27% for those initially managed conservatively, for a similarly adjusted HR of 1.04 (95% CI, 0.80-1.37; P = .75).
In subgroup analyses, Dr. Bangalore reported, there were no significant differences in all-cause or CV mortality by diabetes status, by severity of baseline ischemia, or by whether the patient had recently experienced new or more frequent angina at study entry, was on guideline-directed medical therapy at baseline, or was on dialysis.
Among the contributions of ISCHEMIA-CKD and its 5-year extension study, Dr. Bangalore told this news organization, is that the relative safety of revascularization they showed may help to counter “renalism,” that is, the aversion to invasive intervention in patients with advanced CKD in clinical practice.
For example, if a patient with advanced CKD presents with an acute myocardial infarction, “people are hesitant to take them to the cath lab,” Dr. Bangalore said. But “if you follow protocols, if you follow strategies to minimize the risk, you can safely go ahead and do it.”
But in patients with stable CAD, as the ISCHEMIA-CKD studies show, “routinely revascularizing them may not have significant benefits.”
ISCHEMIC-CKD and its extension study were funded by the National Heart, Lung, and Blood Institute. Dr. Bangalore discloses receiving research grants from NHLBI and serving as a consultant for Abbott Vascular, Biotronik, Boston Scientific, Amgen, Pfizer, Merck, and Reata. Dr. Bhatt has disclosed grants and/or personal fees from many companies; personal fees from WebMD and other publications or organizations; and having other relationships with Medscape Cardiology and other publications or organizations.
A version of this article first appeared on Medscape.com.
FROM ESC CONGRESS 2022
ACC/AHA issue chest pain data standards update to 2021 guideline
The American College of Cardiology/American Heart Association have issued a set of data standards for chest pain and acute myocardial infarction to accompany the 2021 guidelines for evaluation and diagnosis of chest pain.
In October 2021, the AHA/ACC issued a joint clinical practice guideline encouraging clinicians to use standardized risk assessments, clinical pathways, and tools to evaluate and communicate with patients who present with chest pain, as reported by this news organization.
The writing group underscored the need to reach a consensus for the definitions of chest pain. The new document standardizes related data elements for consistent reporting on chest pain syndromes.
“This is an appendix to the guidelines and a planned effort to try to harmonize and bring uniformity to the language applied,” writing committee chair H.V. “Skip” Anderson, MD, with UT Health Science Center, Houston, told this news organization.
“You want heart attack to mean the same thing in Miami Beach as in Western Pennsylvania, as in Oregon and Washington and every place in between,” Dr. Anderson explained. “You want everybody to be using the same language, so that’s what these data standards are meant to do.”
In the document, data elements are grouped into three broad categories: chest pain, myocardial injury, and MI.
“We deliberately followed the plans contained in the new guideline and focused on potentially serious cardiovascular causes of chest pain as might be encountered in emergency departments,” the writing group notes in the document.
The terms “typical” and “atypical” as descriptors of chest pain or anginal syndromes are not used in the new document, in line with the 2021 guidance to abandon these terms.
Instead, the new document divides chest pain syndromes into three categories: “cardiac,” “possible cardiac,” and “noncardiac” – again, in keeping with the chest pain guideline.
The document also includes data elements for risk stratification scoring according to several common risk scoring algorithms and for procedure-related myocardial injury and procedure-related MI.
Each year, chest pain sends more than 7 million adults to the emergency department in the United States. Although noncardiac causes of chest pain make up a large majority of these cases, there are several life-threatening causes of chest pain that must be identified and treated promptly.
Distinguishing between serious and nonserious causes of chest pain is an urgent imperative, the writing group says.
Overall, they say this new clinical lexicon and set of data standards should be “broadly applicable” in various settings, including clinical trials and observational studies, patient care, electronic health records (EHRs), quality and performance improvement initiatives, registries, and public reporting programs.
The 2022 ACC/AHA Key Data Elements and Definitions for Chest Pain and Acute Myocardial Infarction was simultaneously published online in the Journal of the American College of Cardiology and Circulation: Cardiovascular Quality and Outcomes.
It was developed in collaboration with the American College of Emergency Physicians and the Society for Cardiac Angiography and Interventions and endorsed by the Society for Academic Emergency Medicine.
Dr. Anderson noted that “almost all of the guidelines that come out now, certainly in the last few years, have been followed after a certain interval by a set of data standards applicable to the guidelines.”
“It would be really great if it could actually be attached as an appendix, but the nature of the development of these things is such that there will always be a bit of a time lag between the writing group that develops the guidelines and the work group that develops the data standards; you can’t really have them working in parallel at the same time,” Dr. Anderson said in an interview.
This research had no commercial funding. The authors have no relevant disclosures.
A version of this article first appeared on Medscape.com.
The American College of Cardiology/American Heart Association have issued a set of data standards for chest pain and acute myocardial infarction to accompany the 2021 guidelines for evaluation and diagnosis of chest pain.
In October 2021, the AHA/ACC issued a joint clinical practice guideline encouraging clinicians to use standardized risk assessments, clinical pathways, and tools to evaluate and communicate with patients who present with chest pain, as reported by this news organization.
The writing group underscored the need to reach a consensus for the definitions of chest pain. The new document standardizes related data elements for consistent reporting on chest pain syndromes.
“This is an appendix to the guidelines and a planned effort to try to harmonize and bring uniformity to the language applied,” writing committee chair H.V. “Skip” Anderson, MD, with UT Health Science Center, Houston, told this news organization.
“You want heart attack to mean the same thing in Miami Beach as in Western Pennsylvania, as in Oregon and Washington and every place in between,” Dr. Anderson explained. “You want everybody to be using the same language, so that’s what these data standards are meant to do.”
In the document, data elements are grouped into three broad categories: chest pain, myocardial injury, and MI.
“We deliberately followed the plans contained in the new guideline and focused on potentially serious cardiovascular causes of chest pain as might be encountered in emergency departments,” the writing group notes in the document.
The terms “typical” and “atypical” as descriptors of chest pain or anginal syndromes are not used in the new document, in line with the 2021 guidance to abandon these terms.
Instead, the new document divides chest pain syndromes into three categories: “cardiac,” “possible cardiac,” and “noncardiac” – again, in keeping with the chest pain guideline.
The document also includes data elements for risk stratification scoring according to several common risk scoring algorithms and for procedure-related myocardial injury and procedure-related MI.
Each year, chest pain sends more than 7 million adults to the emergency department in the United States. Although noncardiac causes of chest pain make up a large majority of these cases, there are several life-threatening causes of chest pain that must be identified and treated promptly.
Distinguishing between serious and nonserious causes of chest pain is an urgent imperative, the writing group says.
Overall, they say this new clinical lexicon and set of data standards should be “broadly applicable” in various settings, including clinical trials and observational studies, patient care, electronic health records (EHRs), quality and performance improvement initiatives, registries, and public reporting programs.
The 2022 ACC/AHA Key Data Elements and Definitions for Chest Pain and Acute Myocardial Infarction was simultaneously published online in the Journal of the American College of Cardiology and Circulation: Cardiovascular Quality and Outcomes.
It was developed in collaboration with the American College of Emergency Physicians and the Society for Cardiac Angiography and Interventions and endorsed by the Society for Academic Emergency Medicine.
Dr. Anderson noted that “almost all of the guidelines that come out now, certainly in the last few years, have been followed after a certain interval by a set of data standards applicable to the guidelines.”
“It would be really great if it could actually be attached as an appendix, but the nature of the development of these things is such that there will always be a bit of a time lag between the writing group that develops the guidelines and the work group that develops the data standards; you can’t really have them working in parallel at the same time,” Dr. Anderson said in an interview.
This research had no commercial funding. The authors have no relevant disclosures.
A version of this article first appeared on Medscape.com.
The American College of Cardiology/American Heart Association have issued a set of data standards for chest pain and acute myocardial infarction to accompany the 2021 guidelines for evaluation and diagnosis of chest pain.
In October 2021, the AHA/ACC issued a joint clinical practice guideline encouraging clinicians to use standardized risk assessments, clinical pathways, and tools to evaluate and communicate with patients who present with chest pain, as reported by this news organization.
The writing group underscored the need to reach a consensus for the definitions of chest pain. The new document standardizes related data elements for consistent reporting on chest pain syndromes.
“This is an appendix to the guidelines and a planned effort to try to harmonize and bring uniformity to the language applied,” writing committee chair H.V. “Skip” Anderson, MD, with UT Health Science Center, Houston, told this news organization.
“You want heart attack to mean the same thing in Miami Beach as in Western Pennsylvania, as in Oregon and Washington and every place in between,” Dr. Anderson explained. “You want everybody to be using the same language, so that’s what these data standards are meant to do.”
In the document, data elements are grouped into three broad categories: chest pain, myocardial injury, and MI.
“We deliberately followed the plans contained in the new guideline and focused on potentially serious cardiovascular causes of chest pain as might be encountered in emergency departments,” the writing group notes in the document.
The terms “typical” and “atypical” as descriptors of chest pain or anginal syndromes are not used in the new document, in line with the 2021 guidance to abandon these terms.
Instead, the new document divides chest pain syndromes into three categories: “cardiac,” “possible cardiac,” and “noncardiac” – again, in keeping with the chest pain guideline.
The document also includes data elements for risk stratification scoring according to several common risk scoring algorithms and for procedure-related myocardial injury and procedure-related MI.
Each year, chest pain sends more than 7 million adults to the emergency department in the United States. Although noncardiac causes of chest pain make up a large majority of these cases, there are several life-threatening causes of chest pain that must be identified and treated promptly.
Distinguishing between serious and nonserious causes of chest pain is an urgent imperative, the writing group says.
Overall, they say this new clinical lexicon and set of data standards should be “broadly applicable” in various settings, including clinical trials and observational studies, patient care, electronic health records (EHRs), quality and performance improvement initiatives, registries, and public reporting programs.
The 2022 ACC/AHA Key Data Elements and Definitions for Chest Pain and Acute Myocardial Infarction was simultaneously published online in the Journal of the American College of Cardiology and Circulation: Cardiovascular Quality and Outcomes.
It was developed in collaboration with the American College of Emergency Physicians and the Society for Cardiac Angiography and Interventions and endorsed by the Society for Academic Emergency Medicine.
Dr. Anderson noted that “almost all of the guidelines that come out now, certainly in the last few years, have been followed after a certain interval by a set of data standards applicable to the guidelines.”
“It would be really great if it could actually be attached as an appendix, but the nature of the development of these things is such that there will always be a bit of a time lag between the writing group that develops the guidelines and the work group that develops the data standards; you can’t really have them working in parallel at the same time,” Dr. Anderson said in an interview.
This research had no commercial funding. The authors have no relevant disclosures.
A version of this article first appeared on Medscape.com.