LayerRx Mapping ID
245
Slot System
Featured Buckets
Featured Buckets Admin

Squamous Cell Carcinoma Arising in Chronic Inflammatory Dermatoses

Article Type
Changed
Tue, 01/09/2024 - 12:48
Display Headline
Squamous Cell Carcinoma Arising in Chronic Inflammatory Dermatoses

As many as one-quarter of human cancers are related to chronic inflammation, chronic infection, or both.1 Extrinsic inflammation leads to generation of proinflammatory cytokines that in turn recruit other inflammatory cells, which is thought to generate a positive amplification loop.2 Intrinsic stimuli from proto-oncogenes and mutations in tumor suppressor genes lead to transformed cancer cells that also secrete proinflammatory cytokines, thus propagating the cycle.

Numerous factors have been observed in association with tumor growth, progression, invasion, and metastasis.3 One factor for the development of squamous cell carcinoma (SCC) may be chronic inflammatory dermatoses. To date, reviews of chronic inflammation–associated malignancy have focused on solid organ cancers. We sought to provide an up-to-date review of SCC arising within chronic dermatoses, with an emphasis on the anatomic location of dermatoses involved in the transformation of cancer cells, the lag time from onset of dermatosis to diagnosis of SCC, and the distinctive mechanisms thought to be involved in the tumorigenesis in particular dermatoses.

Discoid Lupus Erythematosus

Discoid lupus erythematosus (DLE) is a chronic cutaneous lupus erythematosus variant with a female to male predominance of 3:1,4 and DLE lesions are prone to malignant transformation. Retrospective cohort studies have attempted to characterize who is at risk for SCC and how SCCs behave depending on their location. Cohorts from China,5 India,6 and Japan7 have noted a higher rate of SCC within DLE lesions in men (female to male ratios of 1:2.2, 1:1.6, and 1:2, respectively) and shorter lag times for SCC onset within DLE lesions of the lips (13, 5, and 10 years, respectively) compared to SCC arising in DLE elsewhere (19.2, 11.2, and 26 years, respectively). Studies have noted that DLE lesions of the lips may be prone to more rapid SCC tumorigenesis compared to DLE on cutaneous sites. One study reported SCC in DLE recurrence, metastasis, and death rates of 29%, 16.1%, and 19.4%, respectively,5 which exceeds reported rates in non-DLE SCCs (20%, 0.5% to 6%, and 1%, respectively).5,8

Because SCC arising within DLE is most common on the lips (Figure 1), it has been hypothesized that the high rate of transformation of DLE lesions on the lips may be due to constant exposure to irritation and tobacco, which may accelerate carcinogenesis.5 It also has been hypothesized that atrophic discoid lesions have lost sun protection and are more prone to mutagenic UV radiation,9 as SCCs arising in DLE lesions virtually always display prominent solar elastosis6; however, SCC has been observed to arise in non–sun-exposed DLE lesions in both White and Black patients.10

Invasive squamous cell carcinoma arising within a labial discoid lupus erythematosus lesion. This patient’s lesions were present for approximately 6 years prior to presentation for carcinoma.
Photograph courtesy of Andrea Murina, MD.
FIGURE 1. Invasive squamous cell carcinoma arising within a labial discoid lupus erythematosus lesion. This patient’s lesions were present for approximately 6 years prior to presentation for carcinoma.

Additionally, use of immunosuppressant medications may accelerate the emergence of malignancy or more aggressive forms of malignancy; however, patients with autoimmune disease have a greater risk for malignancy at baseline,11 thus making it difficult to determine the excess risk from medications. There also may be a role for human papillomavirus (HPV) accelerating SCC development in DLE lesions, as demonstrated in a case of SCC arising in DLE lesions of the ears, with viral staining evident within the tumors.12 However, testing for HPV is not routinely performed in these cases.

Dermatologists need to be aware of the relatively rapid tumorigenesis and aggressive behavior of transformation and aggression seen with SCC arising within orolabial DLE lesions compared to cutaneous lesions, especially those on the lips.

Lichen Planus

Although patients with typical cutaneous lichen planus lesions do not have an increased risk for SCC,13 variants of lichen planus may predispose patients to SCC.

 

 

Oral Lichen Planus—Oral lichen planus (OLP) lesions are prone to malignant transformation. A systematic review of 16 studies evaluating the risk for OLP-associated SCC revealed an overall transformation rate of 1.09%, with a mean lag time of 4.3 years,14 compared to a reference rate of 0.2% for oral SCC.15 A meta-analysis of 19,676 patients with OLP and other oral lichenoid lesions revealed an oral SCC rate of 1.1%, with higher rates of transformation seen in cigarette smokers, alcoholics, and patients with hepatitis C virus infection.16 The ulcerative subtype of OLP appears to present a greater risk for malignant transformation.15 Dermatologists also should be cognizant that treatments for OLP such as topical calcineurin inhibitors may support the development of malignancy within inflammatory lesions.17

Hypertrophic Lichen Planus—The hypertrophic variant of lichen planus (HLP) also is prone to malignant transformation. A 1991 epidemiologic study from Sweden of malignancy arising in lichen planus revealed a disproportionate number of cases arising in verrucous or hypertrophic lesions, with a mean of 12.2 years from onset of the dermatosis to malignancy diagnosis.13 A subsequent 2015 retrospective study of 38 patients revealed that SCC had a propensity for the lower limb, favoring the pretibial region and the calf over the foot and the ankle with a reported lag time of 11 years.18

Although metastatic SCC arising in HLP is rare, 2 cases have been reported. A 24-year-old woman presented with an HLP plaque on the lower leg that developed during childhood and rapidly enlarged 2 months prior to presentation; she eventually died from metastatic disease.19 In another case, a 34-year-old man presented with an HLP lesion of approximately 10 years’ duration. A well-differentiated SCC was excised, and he developed lymph node metastases 5 months later.20

It is important to note that HLP on the legs often is misdiagnosed as SCC, as pseudoepitheliomatous hyperplasia and squamous metaplasia can be difficult to differentiate clinically and histologically.21,22 In the case of multiple eruptive SCCs of the lower leg, clinical correlation is essential to avoid unnecessary and ineffective surgical treatment.

Patients with HLP may exhibit Wickham striae, follicular accentuation, and mucocutaneous lichen planus at other sites, or a correlative initiation of possible culprit medications.23 Because true SCC arising within HLP is relatively rare, its malignant potential is not as clear as those arising within DLE; however, the lower limb appears to be the most common location for SCC within HLP.Nail Lichen Planus—Squamous cell carcinoma arising in nail lichen planus is rare. A report of 2 patients were diagnosed with lichen planus approximately 15 years prior to diagnosis of ungual SCC.24 Given the rarity of this presentation, it is difficult to ascertain the approximate lag time and other risk factors. Furthermore, the role of HPV in these cases was not ruled out. Oncogenic HPV strains have been reported in patients with periungual SCC.25,26

Lichen Sclerosus

Lichen sclerosus (LS) is a chronic inflammatory dermatosis that favors the anogenital area in a female to male ratio of 10:1.27 It is considered a premalignant condition for SCC tumorigenesis and may be a strong predictor of vulvar SCC (Figure 2), as 62% of vulvar SCC cases (N=78) may have adjacent LS.28

Poorly differentiated squamous cell carcinoma arising within vulvar lichen sclerosus. This patient’s dermatosis was present for approximately 7 years prior to presentation for carcinoma.
Photograph courtesy of Laura C. Williams, MD (New Orleans, Louisiana).
FIGURE 2. Poorly differentiated squamous cell carcinoma arising within vulvar lichen sclerosus. This patient’s dermatosis was present for approximately 7 years prior to presentation for carcinoma.

In a Dutch cohort of 3038 women with LS, 2.6% of patients developed vulvar SCC at a median of 3.3 years after LS diagnosis.29 Other studies have estimated a lag time of 4 years until SCC presentation.30 An Italian cohort of 976 women similarly observed that 2.7% of patients developed premalignancy or SCC.31 It was previously estimated that 3% to 5% of patients with LS developed SCC; however, prior studies may have included cases of vulvar intraepithelial neoplasia with low risk for invasive SCC, which might have overestimated true risk of SCC.32 Another confounding factor for elucidating SCC on a background of LS may be the presence of HPV.33 Extragenital LS does not appear to have similar potential for malignant transformation.34

 

 

In a prospective Australian cohort of 507 women with LS (mean age, 55.4 years), remission was induced with potent topical corticosteroids.35 Patients who were adherent to a topical regimen did not develop SCC during follow-up. Those who were nonadherent or partially adherent had a 4.7% risk for SCC.35 In a similar prospective study of 83 women in France, the SCC rate was 9.6% in lesions that were untreated or irregularly treated.36 These studies provide essential evidence that appropriately treating LS can prevent SCC at a later date, though longer-term data are lacking.

The rate of SCC arising in male genital LS may approach 8.4%,37 with a lag time of 17 years from onset of LS to SCC diagnosis.38 Although circumcision often is considered curative for male genital LS, patients have been observed to develop penile SCC at least 5 years after circumcision.39 Male penile SCC in a background of LS may not necessarily be HPV associated.40

Marjolin Ulcer

Chronic ulcers or scars, typically postburn scars, may undergo malignant transformation, with SCC being the most common carcinoma.41 Squamous cell carcinoma in the context of a chronic ulcer or wound is known as a Marjolin ulcer (MU). Up to 2% of burn scars have been observed to undergo malignant transformation.42 Marjolin ulcers tend to behave aggressively once they form, and it has been proposed that removal of scar tissue may be a preventive therapeutic strategy.43 Cohort studies of MU on the lower extremities have observed lag times of 26.444 and 37.945 years, with both studies also noting relatively high rates of local recurrence.

The pathogenesis of MU appears to be multifactorial. Chronic inflammation and scar formation have been implicated. Chronic inflammation and irritation of lesions at natural creases are thought to increase mitotic activity,41 and local accumulation of toxin may promote mutagenesis.46 Scar formation may create a locally immunoprivileged site, allowing for developing tumors to evade the immune system47 and become even more aggressive as the tumor accumulates.48 Scar formation also may prevent the ability of immune cells to penetrate the tumor microenvironment and access lymphatic channels.49

Hidradenitis Suppurativa

As many as 3.2% of patients with chronic hidradenitis suppurativa (HS) experience malignant transformation to SCC.50 Early HS displays subclinical lymphedema in affected sites, which can progress to chronic fibrosis, stasis, and accumulation of protein-rich fluid.51 Stasis changes have been associated with altered local inflammatory proteins, such as toll-like receptors, β-defensins, and interleukins.52

A retrospective cohort study of 12 patients revealed a lag time of 28.5 years from HS diagnosis to the manifestation of malignancy.53 After local excision, 7 patients developed recurrence, with 100% mortality. Squamous cell carcinomas were well differentiated and moderately differentiated.53 A 2017 literature review of 62 case reports calculated a mean lag time of 27 years. Despite 85% of SCCs being well differentiated and moderately differentiated, nearly half of patients died within 2 years.54 As seen in other inflammatory conditions, HPV can complicate perineal HS and promote SCC tumorigenesis.55

Squamous cell carcinomas arising within HS lesions are more prevalent in males (6.75:1 ratio),54,56 despite HS being more prevalent in females (2:1 ratio).57 Similar to DLE, SCCs arising in HS are aggressive and are seen more in males, despite both conditions being female predominant. Incidence and mortality rates for primary cutaneous SCC are higher for men vs women58; however, the discordance in aggressive behavior seen more commonly in SCC arising from HS or DLE in male patients has yet to be explained.

 

 

Necrobiosis Lipoidica Diabeticorum

Malignancy arising within necrobiosis lipoidica diabeticorum (NLD) is rare. A review of 14 published cases noted that 13 were SCC and 1 was leiomyosarcoma.59 The lag time was 21.5 years; 31% of cases (N=14) presented with regional lymph node metastasis. Although chronic ulceration is a risk factor for SCC and occurs in as many as one-third of NLD cases, its correlation with ulceration and malignant transformation has not been characterized.

Epidermolysis Bullosa

Recessive dystrophic epidermolysis bullosa (RDEB) is a noninflammatory inherited blistering disease, and patients have an inherently high risk for aggressive SCC.60 Other forms of epidermolysis bullosa can lead to SCC, but the rarer RDEB accounts for 69% of SCC cases, with a median age of 36 years at presentation.61 Although SCCs tend to be well differentiated in RDEB (73.9%),61 they also exhibit highly aggressive behavior.62 In the most severe variant—RDEB-generalized severe—the cumulative risk for SCC-related death in an Australian population was 84.4% at 34 years of age.63

As RDEB is an inherited disorder with potential for malignancy at a young age, the pathogenesis is plausibly different from the previously discussed inflammatory dermatoses. This disease is characterized by a mutation in the collagen VII gene, leading to loss of anchoring fibrils and a basement membrane zone split.64 There also can be inherent fibroblast alterations; RDEB fibroblasts create an environment for tumor growth by supporting malignant-cell adhesion and invasion.65 Mutations in p53,66 local alterations in transforming growth factor β activity,67 and downstream matrix metalloproteinase activity68 have been implicated.

Additionally, keratinocytes may retain the N-terminal noncollagenous (NC1) domain of truncated collagen VII while losing the anchoring NC2 domain in mutated collagen VII RDEB, thereby supporting anchorless keratinocyte survival and higher metastatic potential.69 Retention of this truncated NC1 domain has shown conversion of RDEB keratinocytes to tumor in a xenotransplant mouse model.70 A high level of type VII collagen itself may inherently be protumorigenic for keratinocytes.71

There does not appear to be evidence for HPV involvement in RDEB-associated SCC.72 Squamous cell carcinoma development in RDEB appears to be multifactorial,73 but validated tumor models are lacking. Other than conventional oncologic therapy, future directions in the management of RDEB may include gene-, protein- and cell-targeted therapies.73

Conclusion

Squamous cell carcinomas are known to arise within chronic cutaneous inflammatory dermatoses. Tumorigenesis peaks relatively early in new orolabial DLE, LS, and OLP cases, and can occur over many decades in cutaneous DLE, HLP, HS, NLD, and chronic wounds or scars, summarized in the Table. Frequent SCCs are observed in high-risk subtypes of epidermolysis bullosa. Dermatologists must examine areas affected by these diseases at regular intervals, being mindful of the possibility of SCC development. Furthermore, dermatologists should adopt a lower threshold to biopsy suspicious lesions, especially those that develop within relatively new orolabial DLE, chronic HS, or chronic wound cases, as SCC in these settings is particularly aggressive and displays mortality and metastasis rates that exceed those of common cutaneous SCC.


References
  1. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007;121:2373-2380. doi:10.1002/ijc.23173
  2. Mantovani A, Allavena P, Sica A, et al. Cancer-related inflammation. Nature. 2008;454:436-444. doi:10.1038/nature07205
  3. Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development. Front Immunol. 2011;2:98. doi:10.3389/fimmu.2011.00098
  4. Tebbe B. Clinical course and prognosis of cutaneous lupus erythematosus. Clin Dermatol. 2004;22:121-124. doi:10.1016/j.clindermatol.2003.12.018
  5. Tao J, Zhang X, Guo N, et al. Squamous cell carcinoma complicating discoid lupus erythematosus in Chinese patients: review of the literature, 1964-2010. J Am Acad Dermatol. 2012;66:695-696. doi:10.1016 /j.jaad.2011.09.033
  6. Fernandes MS, Girisha BS, Viswanathan N, et al. Discoid lupus erythematosus with squamous cell carcinoma: a case report and review of the literature in Indian patients. Lupus. 2015;24:1562-1566. doi:10.1177/0961203315599245
  7. Makita E, Akasaka E, Sakuraba Y, et al. Squamous cell carcinoma on the lip arising from discoid lupus erythematosus: a case report and review of Japanese patients. Eur J Dermatol. 2016;26:395-396. doi:10.1684/ejd.2016.2780
  8. Clayman GL, Lee JJ, Holsinger FC, et al. Mortality risk from squamous cell skin cancer. J Clin Oncol. 2005;23:759-765. doi:10.1200/JCO.2005.02.155
  9. Arvanitidou I-E, Nikitakis NG, Georgaki M, et al. Multiple primary squamous cell carcinomas of the lower lip and tongue arising in discoid lupus erythematosus: a case report. Oral Surg Oral Med Oral Pathol Oral Radiol. 2018;125:e22-e30. doi:10.1016/j.oooo.2017.08.012
  10. Alsanafi S, Werth VP. Squamous cell carcinomas arising in discoid lupus erythematosus scars: unusual occurrence in an African-American and in a sun-protected area. J Clin Rheumatol. 2011;17:35-36. doi:10.1097/RHU.0b013e3182051928
  11. Goobie GC, Bernatsky S, Ramsey-Goldman R, et al. Malignancies in systemic lupus erythematosus: a 2015 update. Curr Opin Rheumatol. 2015;27:454-460. doi:10.1097/BOR.0000000000000202
  12. Simpson JK, Medina-Flores R, Deng J-S. Squamous cell carcinoma arising in discoid lupus erythematosus lesions of the ears infected with human papillomavirus. Cutis. 2010;86:195-198.
  13. Sigurgeirsson B, Lindelöf B. Lichen planus and malignancy. an epidemiologic study of 2071 patients and a review of the literature. Arch Dermatol. 1991;127:1684-1688. doi:10.1001/archderm.127.11.1684
  14. Fitzpatrick SG, Hirsch SA, Gordon SC. The malignant transformation of oral lichen planus and oral lichenoid lesions: a systematic review. J Am Dent Assoc. 2014;145:45-56. doi:10.14219/jada.2013.10
  15. Laniosz V, Torgerson RR, Ramos-Rodriguez AJ, et al. Incidence of squamous cell carcinoma in oral lichen planus: a 25-year population-based study. Int J Dermatol. 2019;58:296-301. doi:10.1111/ijd.14215
  16. Aghbari SMH, Abushouk AI, Attia A, et al. Malignant transformation of oral lichen planus and oral lichenoid lesions: a meta-analysis of 20095 patient data. Oral Oncol. 2017;68:92-102. doi:10.1016/j.oraloncology.2017.03.012
  17. Morita M, Asoda S, Tsunoda K, et al. The onset risk of carcinoma in patients continuing tacrolimus topical treatment for oral lichen planus: a case report. Odontology. 2017;105:262-266. doi:10.1007/s10266-016-0255-4
  18. Knackstedt TJ, Collins LK, Li Z, et al. Squamous cell carcinoma arising in hypertrophic lichen planus: a review and analysis of 38 cases. Dermatol Surg. 2015;41:1411-1418. doi:10.1097/DSS.0000000000000565
  19. Tong LX, Weinstock MJ, Drews R, et al. Widely metastatic squamous cell carcinoma originating from malignant transformation of hypertrophic lichen planus in a 24-year-old woman: case report and review of the literature. Pediatr Dermatol. 2015;32:e98-e101. doi:10.1111/pde.12549
  20. Ardabili M, Gambichler T, Rotterdam S, et al. Metastatic cutaneous squamous cell carcinoma arising from a previous area of chronic hypertrophic lichen planus. Dermatol Online J. 2003;9:10.
  21. Bowen AR, Burt L, Boucher K, et al. Use of proliferation rate, p53 staining and perforating elastic fibers in distinguishing keratoacanthoma from hypertrophic lichen planus: a pilot study. J Cutan Pathol. 2012;39:243-250. doi:10.1111/j.1600-0560.2011.01834.x
  22. Totonchy MB, Leventhal JS, Ko CJ, et al. Hypertrophic lichen planus and well-differentiated squamous cell carcinoma: a diagnostic conundrum. Dermatol Surg. 2018;44:1466-1470. doi:10.1097/DSS.0000000000001465
  23. Levandoski KA, Nazarian RM, Asgari MM. Hypertrophic lichen planus mimicking squamous cell carcinoma: the importance of clinicopathologic correlation. JAAD Case Rep. 2017;3:151-154. doi: 10.1016/j.jdcr.2017.01.020
  24. Okiyama N, Satoh T, Yokozeki H, et al. Squamous cell carcinoma arising from lichen planus of nail matrix and nail bed. J Am Acad Dermatol. 2005;53:908-909. doi:10.1016/j.jaad.2005.04.052
  25. Riddel C, Rashid R, Thomas V. Ungual and periungual human papillomavirus-associated squamous cell carcinoma: a review. J Am Acad Dermatol. 2011;64:1147-1153. doi:10.1016/j.jaad.2010.02.057
  26. Shimizu A, Kuriyama Y, Hasegawa M, et al. Nail squamous cell carcinoma: a hidden high-risk human papillomavirus reservoir for sexually transmitted infections. J Am Acad Dermatol. 2019;81:1358-1370. doi:10.1016/j.jaad.2019.03.070
  27. Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. J Am Acad Dermatol. 1995;32:393-416. doi:10.1016/0190-9622(95)90060-8
  28. Leibowitch M, Neill S, Pelisse M, et al. The epithelial changes associated with squamous cell carcinoma of the vulva: a review of the clinical, histological and viral findings in 78 women. Br J Obstet Gynaecol. 1990;97:1135-1139. doi:10.1111/j.1471-0528.1990.tb02502.x
  29. Bleeker MCG, Visser PJ, Overbeek LIH, et al. Lichen sclerosus: incidence and risk of vulvar squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev. 2016;25:1224-1230. doi:10.1158/1055-9965.EPI-16-0019
  30. Carlson JA, Ambros R, Malfetano J, et al. Vulvar lichen sclerosus and squamous cell carcinoma: a cohort, case control, and investigational study with historical perspective; implications for chronic inflammation and sclerosis in the development of neoplasia. Hum Pathol. 1998;29:932-948. doi:10.1016/s0046-8177(98)90198-8
  31. Micheletti L, Preti M, Radici G, et al. Vulvar lichen sclerosus and neoplastic transformation: a retrospective study of 976 cases. J Low Genit Tract Dis. 2016;20:180-183. doi:10.1097/LGT.0000000000000186
  32. Cooper SM, Madnani N, Margesson L. Reduced risk of squamous cell carcinoma with adequate treatment of vulvar lichen sclerosus. JAMA Dermatol. 2015;151:1059-1060. doi:10.1001/jamadermatol.2015.0644
  33. Rakislova N, Alemany L, Clavero O, et al; VVAP Study Group. Differentiated vulvar intraepithelial neoplasia-like and lichen sclerosus-like lesions in HPV-associated squamous cell carcinomas of the vulva. Am J Surg Pathol. 2018;42:828-835. doi:10.1097/PAS.0000000000001047
  34. Val I, Almeida G. An overview of lichen sclerosus. Clin Obstet Gynecol. 2005;48:808-817. doi:10.1097/01.grf.0000179635.64663.3d
  35. Lee A, Bradford J, Fischer G. Long-term management of adult vulvar lichen sclerosus: a prospective cohort study of 507 women. JAMA Dermatol. 2015;151:1061-1067. doi:10.1001/jamadermatol.2015.0643
  36. Renaud-Vilmer C, Cavelier-Balloy B, Porcher R, et al. Vulvar lichen sclerosus: effect of long-term topical application of a potent steroid on the course of the disease. Arch Dermatol. 2004;140:709-712. doi:10.1001/archderm.140.6.709
  37. Minhas S, Manseck A, Watya S, et al. Penile cancer—prevention and premalignant conditions. Urology. 2010;76(2 suppl 1):S24-S35. doi:10.1016/j.urology.2010.04.007
  38. Nasca MR, Innocenzi D, Micali G. Penile cancer among patients with genital lichen sclerosus. J Am Acad Dermatol. 1999;41:911-914. doi:10.1016/s0190-9622(99)70245-8
  39. Philippou P, Shabbir M, Ralph DJ, et al. Genital lichen sclerosus/balanitis xerotica obliterans in men with penile carcinoma: a critical analysis. BJU Int. 2013;111:970-976. doi:10.1111/j.1464-410X.2012.11773.x
  40. Velazquez EF, Cubilla AL. Lichen sclerosus in 68 patients with squamous cell carcinoma of the penis: frequent atypias and correlation with special carcinoma variants suggests a precancerous role. Am J Surg Pathol. 2003;27:1448-1453. doi:10.1097/00000478-200311000-00007
  41. Pekarek B, Buck S, Osher L. A comprehensive review on Marjolin’s ulcers: diagnosis and treatment. J Am Col Certif Wound Spec. 2011;3:60-64. doi:10.1016/j.jcws.2012.04.001
  42. Aydogdu E, Yildirim S, Akoz T. Is surgery an effective and adequate treatment in advanced Marjolin’s ulcer? Burns. 2005;31:421-431. doi:10.1016/j.burns.2005.02.008
  43. Xiao H, Deng K, Liu R, et al. A review of 31 cases of Marjolin’s ulcer on scalp: is it necessary to preventively remove the scar? Int Wound J. 2019;16:479-485. doi:10.1111/iwj.13058
  44. Chaturvedi G, Gupta AK, Das S, et al. Marjolin ulcer: an observational epidemiological study from a tertiary care centre in India. Ann Plast Surg. 2019;83:518-522. doi:10.1097/SAP.0000000000001995
  45. Karasoy Yesilada A, Zeynep Sevim K, Özgur Sucu D, et al. Marjolin ulcer: clinical experience with 34 patients over 15 years. J Cutan Med Surg. 2013;17:404-409. doi:10.2310/7750.2013.13016
  46. Bazalin´ski D, Przybek-Mita J, Baran´ska B, et al. Marjolin’s ulcer in chronic wounds - review of available literature. Contemp Oncol (Pozn). 2017;21:197-202. doi:10.5114/wo.2017.70109
  47. Visuthikosol V, Boonpucknavig V, Nitiyanant P. Squamous carcinoma in scars: clinicopathological correlations. Ann Plast Surg. 1986;16:42-48. doi:10.1097/00000637-198601000-00004
  48. Bostwick J 3rd, Pendergrast WJ Jr, Vasconez LO. Marjolin’s ulcer: an immunologically privileged tumor? Plast Reconstr Surg. 1976;57:66-69.
  49. Kerr-Valentic MA, Samimi K, Rohlen BH, et al. Marjolin’s ulcer: modern analysis of an ancient problem. Plast Reconstr Surg. 2009;123:184-191. doi:10.1097/PRS.0b013e3181904d86
  50. Constantinou C, Widom K, Desantis J, et al. Hidradenitis suppurativa complicated by squamous cell carcinoma. Am Surg. 2008;74:1177-1181.
  51. Fabbrocini G, Ruocco E, De Vita V, et al. Squamous cell carcinoma arising in long-standing hidradenitis suppurativa: an overlooked facet of the immunocompromised district. Clin Dermatol. 2017;35:225-227. doi:10.1016/j.clindermatol.2016.10.019
  52. Baroni A, Buommino E, Piccolo V, et al. Alterations of skin innate immunity in lymphedematous limbs: correlations with opportunistic diseases. Clin Dermatol. 2014;32:592-598. doi:10.1016/j.clindermatol.2014.04.006
  53. Kohorst JJ, Shah KK, Hallemeier CL, et al. Squamous cell carcinoma in perineal, perianal, and gluteal hidradenitis suppurativa: experience in 12 patients. Dermatol Surg. 2019;45:519-526. doi:10.1097/DSS.0000000000001713
  54. Huang C, Lai Z, He M, et al. Successful surgical treatment for squamous cell carcinoma arising from hidradenitis suppurativa: a case report and literature review. Medicine (Baltimore). 2017;96:e5857. doi:10.1097/MD.0000000000005857
  55. Lavogiez C, Delaporte E, Darras-Vercambre S, et al. Clinicopathological study of 13 cases of squamous cell carcinoma complicating hidradenitis suppurativa. Dermatology. 2010;220:147-153. doi:10.1159/000269836
  56. Makris G-M, Poulakaki N, Papanota A-M, et al. Vulvar, perianal and perineal cancer after hidradenitis suppurativa: a systematic review and pooled analysis. Dermatol Surg. 2017;43:107-115. doi:10.1097/DSS.0000000000000944
  57. Cosmatos I, Matcho A, Weinstein R, et al. Analysis of patient claims data to determine the prevalence of hidradenitis suppurativa in the United States. J Am Acad Dermatol. 2013;68:412-419. doi:10.1016/j.jaad.2012.07.027
  58. Hollestein LM, de Vries E, Nijsten T. Trends of cutaneous squamous cell carcinoma in the Netherlands: increased incidence rates, but stable relative survival and mortality 1989-2008. Eur J Cancer. 2012;48:2046-2053. doi:10.1016/j.ejca.2012.01.003
  59. Uva L, Freitas J, Soares de Almeida L, et al. Squamous cell carcinoma arising in ulcerated necrobiosis lipoidica diabeticorum. Int Wound J. 2015;12:741-743. doi:10.1111/iwj.12206
  60. McGrath JA, Schofield OM, Mayou BJ, et al. Epidermolysis bullosa complicated by squamous cell carcinoma: report of 10 cases. J Cutan Pathol. 1992;19:116-123. doi:10.1111/j.1600-0560.1992.tb01352.x
  61. Montaudié H, Chiaverini C, Sbidian E, et al. Inherited epidermolysis bullosa and squamous cell carcinoma: a systematic review of 117 cases. Orphanet J Rare Dis. 2016;11:117. doi:10.1186/s13023-016-0489-9.
  62. Fine J-D. Inherited epidermolysis bullosa: past, present, and future. Ann N Y Acad Sci. 2010;1194:213-222. doi:10.1111/j.1749-6632.2010.05463.x
  63. Kim M, Li M, Intong-Wheeler LRA, et al. Epidemiology and outcome of squamous cell carcinoma in epidermolysis bullosa in Australia and New Zealand. Acta Derm Venereol. 2018;98:70-76. doi:10.2340/00015555-2781
  64. Bruckner-Tuderman L, Mitsuhashi Y, Schnyder UW, et al. Anchoring fibrils and type VII collagen are absent from skin in severe recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 1989;93:3-9. doi:10.1111/1523-1747.ep12277331
  65. Ng Y-Z, Pourreyron C, Salas-Alanis JC, et al. Fibroblast-derived dermal matrix drives development of aggressive cutaneous squamous cell carcinoma in patients with recessive dystrophic epidermolysis bullosa. Cancer Res. 2012;72:3522-3534. doi:10.1158/0008-5472.CAN-11-2996
  66. Arbiser JL, Fan C-Y, Su X, et al. Involvement of p53 and p16 tumor suppressor genes in recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2004;123:788-790. doi:10.1111/j.0022-202X.2004.23418.x
  67. Knaup J, Gruber C, Krammer B, et al. TGFbeta-signaling in squamous cell carcinoma occurring in recessive dystrophic epidermolysis bullosa. Anal Cell Pathol (Amst). 2011;34:339-353. doi:10.3233/ACP-2011-0039
  68. Kivisaari AK, Kallajoki M, Mirtti T, et al. Transformation-specific matrix metalloproteinases (MMP)-7 and MMP-13 are expressed by tumour cells in epidermolysis bullosa-associated squamous cell carcinomas. Br J Dermatol. 2008;158:778-785. doi:10.1111/j.1365-2133.2008.08466.x
  69. Rodeck U, Fertala A, Uitto J. Anchorless keratinocyte survival: an emerging pathogenic mechanism for squamous cell carcinoma in recessive dystrophic epidermolysis bullosa. Exp Dermatol. 2007;16:465-467. doi:10.1111/j.1600-0625.2007.00563.x
  70. Ortiz-Urda S, Garcia J, Green CL, et al. Type VII collagen is required for Ras-driven human epidermal tumorigenesis. Science. 2005;307:1773-1776. doi:10.1126/science.1106209
  71. Pourreyron C, Chen M, McGrath JA, et al. High levels of type VII collagen expression in recessive dystrophic epidermolysis bullosa cutaneous squamous cell carcinoma keratinocytes increases PI3K and MAPK signalling, cell migration and invasion. Br J Dermatol. 2014;170:1256-1265. doi:10.1111/bjd.12715
  72. Purdie KJ, Pourreyron C, Fassihi H, et al. No evidence that human papillomavirus is responsible for the aggressive nature of recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2010;130:2853-2855. doi:10.1038/jid.2010.243
  73. South AP, O’Toole EA. Understanding the pathogenesis of recessive dystrophic epidermolysis bullosa squamous cell carcinoma. Dermatol Clin. 2010;28:171-178. doi:10.1016/j.det.2009.10.023
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Tulane University, New Orleans, Louisiana. Dr. Kuraitis also is from Roswell Park Cancer Center, Buffalo, New York.

Dr. Kuraitis is a speaker and consultant for Ortho Dermatologics and a consultant for UCB. Dr. Murina is a speaker for AbbVie, Amgen, Bristol-Myers Squibb, Janssen, Pfizer, and UCB. She also is a consultant for AbbVie, Bristol-Meyers Squibb, Janssen, Novartis, Ortho Dermatologics, and UCB.

Correspondence: Drew Kuraitis, MD, PhD (dkuraiti@tulane.edu).

Issue
Cutis - 113(1)
Publications
Topics
Page Number
29-34
Sections
Author and Disclosure Information

From the Department of Dermatology, Tulane University, New Orleans, Louisiana. Dr. Kuraitis also is from Roswell Park Cancer Center, Buffalo, New York.

Dr. Kuraitis is a speaker and consultant for Ortho Dermatologics and a consultant for UCB. Dr. Murina is a speaker for AbbVie, Amgen, Bristol-Myers Squibb, Janssen, Pfizer, and UCB. She also is a consultant for AbbVie, Bristol-Meyers Squibb, Janssen, Novartis, Ortho Dermatologics, and UCB.

Correspondence: Drew Kuraitis, MD, PhD (dkuraiti@tulane.edu).

Author and Disclosure Information

From the Department of Dermatology, Tulane University, New Orleans, Louisiana. Dr. Kuraitis also is from Roswell Park Cancer Center, Buffalo, New York.

Dr. Kuraitis is a speaker and consultant for Ortho Dermatologics and a consultant for UCB. Dr. Murina is a speaker for AbbVie, Amgen, Bristol-Myers Squibb, Janssen, Pfizer, and UCB. She also is a consultant for AbbVie, Bristol-Meyers Squibb, Janssen, Novartis, Ortho Dermatologics, and UCB.

Correspondence: Drew Kuraitis, MD, PhD (dkuraiti@tulane.edu).

Article PDF
Article PDF

As many as one-quarter of human cancers are related to chronic inflammation, chronic infection, or both.1 Extrinsic inflammation leads to generation of proinflammatory cytokines that in turn recruit other inflammatory cells, which is thought to generate a positive amplification loop.2 Intrinsic stimuli from proto-oncogenes and mutations in tumor suppressor genes lead to transformed cancer cells that also secrete proinflammatory cytokines, thus propagating the cycle.

Numerous factors have been observed in association with tumor growth, progression, invasion, and metastasis.3 One factor for the development of squamous cell carcinoma (SCC) may be chronic inflammatory dermatoses. To date, reviews of chronic inflammation–associated malignancy have focused on solid organ cancers. We sought to provide an up-to-date review of SCC arising within chronic dermatoses, with an emphasis on the anatomic location of dermatoses involved in the transformation of cancer cells, the lag time from onset of dermatosis to diagnosis of SCC, and the distinctive mechanisms thought to be involved in the tumorigenesis in particular dermatoses.

Discoid Lupus Erythematosus

Discoid lupus erythematosus (DLE) is a chronic cutaneous lupus erythematosus variant with a female to male predominance of 3:1,4 and DLE lesions are prone to malignant transformation. Retrospective cohort studies have attempted to characterize who is at risk for SCC and how SCCs behave depending on their location. Cohorts from China,5 India,6 and Japan7 have noted a higher rate of SCC within DLE lesions in men (female to male ratios of 1:2.2, 1:1.6, and 1:2, respectively) and shorter lag times for SCC onset within DLE lesions of the lips (13, 5, and 10 years, respectively) compared to SCC arising in DLE elsewhere (19.2, 11.2, and 26 years, respectively). Studies have noted that DLE lesions of the lips may be prone to more rapid SCC tumorigenesis compared to DLE on cutaneous sites. One study reported SCC in DLE recurrence, metastasis, and death rates of 29%, 16.1%, and 19.4%, respectively,5 which exceeds reported rates in non-DLE SCCs (20%, 0.5% to 6%, and 1%, respectively).5,8

Because SCC arising within DLE is most common on the lips (Figure 1), it has been hypothesized that the high rate of transformation of DLE lesions on the lips may be due to constant exposure to irritation and tobacco, which may accelerate carcinogenesis.5 It also has been hypothesized that atrophic discoid lesions have lost sun protection and are more prone to mutagenic UV radiation,9 as SCCs arising in DLE lesions virtually always display prominent solar elastosis6; however, SCC has been observed to arise in non–sun-exposed DLE lesions in both White and Black patients.10

Invasive squamous cell carcinoma arising within a labial discoid lupus erythematosus lesion. This patient’s lesions were present for approximately 6 years prior to presentation for carcinoma.
Photograph courtesy of Andrea Murina, MD.
FIGURE 1. Invasive squamous cell carcinoma arising within a labial discoid lupus erythematosus lesion. This patient’s lesions were present for approximately 6 years prior to presentation for carcinoma.

Additionally, use of immunosuppressant medications may accelerate the emergence of malignancy or more aggressive forms of malignancy; however, patients with autoimmune disease have a greater risk for malignancy at baseline,11 thus making it difficult to determine the excess risk from medications. There also may be a role for human papillomavirus (HPV) accelerating SCC development in DLE lesions, as demonstrated in a case of SCC arising in DLE lesions of the ears, with viral staining evident within the tumors.12 However, testing for HPV is not routinely performed in these cases.

Dermatologists need to be aware of the relatively rapid tumorigenesis and aggressive behavior of transformation and aggression seen with SCC arising within orolabial DLE lesions compared to cutaneous lesions, especially those on the lips.

Lichen Planus

Although patients with typical cutaneous lichen planus lesions do not have an increased risk for SCC,13 variants of lichen planus may predispose patients to SCC.

 

 

Oral Lichen Planus—Oral lichen planus (OLP) lesions are prone to malignant transformation. A systematic review of 16 studies evaluating the risk for OLP-associated SCC revealed an overall transformation rate of 1.09%, with a mean lag time of 4.3 years,14 compared to a reference rate of 0.2% for oral SCC.15 A meta-analysis of 19,676 patients with OLP and other oral lichenoid lesions revealed an oral SCC rate of 1.1%, with higher rates of transformation seen in cigarette smokers, alcoholics, and patients with hepatitis C virus infection.16 The ulcerative subtype of OLP appears to present a greater risk for malignant transformation.15 Dermatologists also should be cognizant that treatments for OLP such as topical calcineurin inhibitors may support the development of malignancy within inflammatory lesions.17

Hypertrophic Lichen Planus—The hypertrophic variant of lichen planus (HLP) also is prone to malignant transformation. A 1991 epidemiologic study from Sweden of malignancy arising in lichen planus revealed a disproportionate number of cases arising in verrucous or hypertrophic lesions, with a mean of 12.2 years from onset of the dermatosis to malignancy diagnosis.13 A subsequent 2015 retrospective study of 38 patients revealed that SCC had a propensity for the lower limb, favoring the pretibial region and the calf over the foot and the ankle with a reported lag time of 11 years.18

Although metastatic SCC arising in HLP is rare, 2 cases have been reported. A 24-year-old woman presented with an HLP plaque on the lower leg that developed during childhood and rapidly enlarged 2 months prior to presentation; she eventually died from metastatic disease.19 In another case, a 34-year-old man presented with an HLP lesion of approximately 10 years’ duration. A well-differentiated SCC was excised, and he developed lymph node metastases 5 months later.20

It is important to note that HLP on the legs often is misdiagnosed as SCC, as pseudoepitheliomatous hyperplasia and squamous metaplasia can be difficult to differentiate clinically and histologically.21,22 In the case of multiple eruptive SCCs of the lower leg, clinical correlation is essential to avoid unnecessary and ineffective surgical treatment.

Patients with HLP may exhibit Wickham striae, follicular accentuation, and mucocutaneous lichen planus at other sites, or a correlative initiation of possible culprit medications.23 Because true SCC arising within HLP is relatively rare, its malignant potential is not as clear as those arising within DLE; however, the lower limb appears to be the most common location for SCC within HLP.Nail Lichen Planus—Squamous cell carcinoma arising in nail lichen planus is rare. A report of 2 patients were diagnosed with lichen planus approximately 15 years prior to diagnosis of ungual SCC.24 Given the rarity of this presentation, it is difficult to ascertain the approximate lag time and other risk factors. Furthermore, the role of HPV in these cases was not ruled out. Oncogenic HPV strains have been reported in patients with periungual SCC.25,26

Lichen Sclerosus

Lichen sclerosus (LS) is a chronic inflammatory dermatosis that favors the anogenital area in a female to male ratio of 10:1.27 It is considered a premalignant condition for SCC tumorigenesis and may be a strong predictor of vulvar SCC (Figure 2), as 62% of vulvar SCC cases (N=78) may have adjacent LS.28

Poorly differentiated squamous cell carcinoma arising within vulvar lichen sclerosus. This patient’s dermatosis was present for approximately 7 years prior to presentation for carcinoma.
Photograph courtesy of Laura C. Williams, MD (New Orleans, Louisiana).
FIGURE 2. Poorly differentiated squamous cell carcinoma arising within vulvar lichen sclerosus. This patient’s dermatosis was present for approximately 7 years prior to presentation for carcinoma.

In a Dutch cohort of 3038 women with LS, 2.6% of patients developed vulvar SCC at a median of 3.3 years after LS diagnosis.29 Other studies have estimated a lag time of 4 years until SCC presentation.30 An Italian cohort of 976 women similarly observed that 2.7% of patients developed premalignancy or SCC.31 It was previously estimated that 3% to 5% of patients with LS developed SCC; however, prior studies may have included cases of vulvar intraepithelial neoplasia with low risk for invasive SCC, which might have overestimated true risk of SCC.32 Another confounding factor for elucidating SCC on a background of LS may be the presence of HPV.33 Extragenital LS does not appear to have similar potential for malignant transformation.34

 

 

In a prospective Australian cohort of 507 women with LS (mean age, 55.4 years), remission was induced with potent topical corticosteroids.35 Patients who were adherent to a topical regimen did not develop SCC during follow-up. Those who were nonadherent or partially adherent had a 4.7% risk for SCC.35 In a similar prospective study of 83 women in France, the SCC rate was 9.6% in lesions that were untreated or irregularly treated.36 These studies provide essential evidence that appropriately treating LS can prevent SCC at a later date, though longer-term data are lacking.

The rate of SCC arising in male genital LS may approach 8.4%,37 with a lag time of 17 years from onset of LS to SCC diagnosis.38 Although circumcision often is considered curative for male genital LS, patients have been observed to develop penile SCC at least 5 years after circumcision.39 Male penile SCC in a background of LS may not necessarily be HPV associated.40

Marjolin Ulcer

Chronic ulcers or scars, typically postburn scars, may undergo malignant transformation, with SCC being the most common carcinoma.41 Squamous cell carcinoma in the context of a chronic ulcer or wound is known as a Marjolin ulcer (MU). Up to 2% of burn scars have been observed to undergo malignant transformation.42 Marjolin ulcers tend to behave aggressively once they form, and it has been proposed that removal of scar tissue may be a preventive therapeutic strategy.43 Cohort studies of MU on the lower extremities have observed lag times of 26.444 and 37.945 years, with both studies also noting relatively high rates of local recurrence.

The pathogenesis of MU appears to be multifactorial. Chronic inflammation and scar formation have been implicated. Chronic inflammation and irritation of lesions at natural creases are thought to increase mitotic activity,41 and local accumulation of toxin may promote mutagenesis.46 Scar formation may create a locally immunoprivileged site, allowing for developing tumors to evade the immune system47 and become even more aggressive as the tumor accumulates.48 Scar formation also may prevent the ability of immune cells to penetrate the tumor microenvironment and access lymphatic channels.49

Hidradenitis Suppurativa

As many as 3.2% of patients with chronic hidradenitis suppurativa (HS) experience malignant transformation to SCC.50 Early HS displays subclinical lymphedema in affected sites, which can progress to chronic fibrosis, stasis, and accumulation of protein-rich fluid.51 Stasis changes have been associated with altered local inflammatory proteins, such as toll-like receptors, β-defensins, and interleukins.52

A retrospective cohort study of 12 patients revealed a lag time of 28.5 years from HS diagnosis to the manifestation of malignancy.53 After local excision, 7 patients developed recurrence, with 100% mortality. Squamous cell carcinomas were well differentiated and moderately differentiated.53 A 2017 literature review of 62 case reports calculated a mean lag time of 27 years. Despite 85% of SCCs being well differentiated and moderately differentiated, nearly half of patients died within 2 years.54 As seen in other inflammatory conditions, HPV can complicate perineal HS and promote SCC tumorigenesis.55

Squamous cell carcinomas arising within HS lesions are more prevalent in males (6.75:1 ratio),54,56 despite HS being more prevalent in females (2:1 ratio).57 Similar to DLE, SCCs arising in HS are aggressive and are seen more in males, despite both conditions being female predominant. Incidence and mortality rates for primary cutaneous SCC are higher for men vs women58; however, the discordance in aggressive behavior seen more commonly in SCC arising from HS or DLE in male patients has yet to be explained.

 

 

Necrobiosis Lipoidica Diabeticorum

Malignancy arising within necrobiosis lipoidica diabeticorum (NLD) is rare. A review of 14 published cases noted that 13 were SCC and 1 was leiomyosarcoma.59 The lag time was 21.5 years; 31% of cases (N=14) presented with regional lymph node metastasis. Although chronic ulceration is a risk factor for SCC and occurs in as many as one-third of NLD cases, its correlation with ulceration and malignant transformation has not been characterized.

Epidermolysis Bullosa

Recessive dystrophic epidermolysis bullosa (RDEB) is a noninflammatory inherited blistering disease, and patients have an inherently high risk for aggressive SCC.60 Other forms of epidermolysis bullosa can lead to SCC, but the rarer RDEB accounts for 69% of SCC cases, with a median age of 36 years at presentation.61 Although SCCs tend to be well differentiated in RDEB (73.9%),61 they also exhibit highly aggressive behavior.62 In the most severe variant—RDEB-generalized severe—the cumulative risk for SCC-related death in an Australian population was 84.4% at 34 years of age.63

As RDEB is an inherited disorder with potential for malignancy at a young age, the pathogenesis is plausibly different from the previously discussed inflammatory dermatoses. This disease is characterized by a mutation in the collagen VII gene, leading to loss of anchoring fibrils and a basement membrane zone split.64 There also can be inherent fibroblast alterations; RDEB fibroblasts create an environment for tumor growth by supporting malignant-cell adhesion and invasion.65 Mutations in p53,66 local alterations in transforming growth factor β activity,67 and downstream matrix metalloproteinase activity68 have been implicated.

Additionally, keratinocytes may retain the N-terminal noncollagenous (NC1) domain of truncated collagen VII while losing the anchoring NC2 domain in mutated collagen VII RDEB, thereby supporting anchorless keratinocyte survival and higher metastatic potential.69 Retention of this truncated NC1 domain has shown conversion of RDEB keratinocytes to tumor in a xenotransplant mouse model.70 A high level of type VII collagen itself may inherently be protumorigenic for keratinocytes.71

There does not appear to be evidence for HPV involvement in RDEB-associated SCC.72 Squamous cell carcinoma development in RDEB appears to be multifactorial,73 but validated tumor models are lacking. Other than conventional oncologic therapy, future directions in the management of RDEB may include gene-, protein- and cell-targeted therapies.73

Conclusion

Squamous cell carcinomas are known to arise within chronic cutaneous inflammatory dermatoses. Tumorigenesis peaks relatively early in new orolabial DLE, LS, and OLP cases, and can occur over many decades in cutaneous DLE, HLP, HS, NLD, and chronic wounds or scars, summarized in the Table. Frequent SCCs are observed in high-risk subtypes of epidermolysis bullosa. Dermatologists must examine areas affected by these diseases at regular intervals, being mindful of the possibility of SCC development. Furthermore, dermatologists should adopt a lower threshold to biopsy suspicious lesions, especially those that develop within relatively new orolabial DLE, chronic HS, or chronic wound cases, as SCC in these settings is particularly aggressive and displays mortality and metastasis rates that exceed those of common cutaneous SCC.


As many as one-quarter of human cancers are related to chronic inflammation, chronic infection, or both.1 Extrinsic inflammation leads to generation of proinflammatory cytokines that in turn recruit other inflammatory cells, which is thought to generate a positive amplification loop.2 Intrinsic stimuli from proto-oncogenes and mutations in tumor suppressor genes lead to transformed cancer cells that also secrete proinflammatory cytokines, thus propagating the cycle.

Numerous factors have been observed in association with tumor growth, progression, invasion, and metastasis.3 One factor for the development of squamous cell carcinoma (SCC) may be chronic inflammatory dermatoses. To date, reviews of chronic inflammation–associated malignancy have focused on solid organ cancers. We sought to provide an up-to-date review of SCC arising within chronic dermatoses, with an emphasis on the anatomic location of dermatoses involved in the transformation of cancer cells, the lag time from onset of dermatosis to diagnosis of SCC, and the distinctive mechanisms thought to be involved in the tumorigenesis in particular dermatoses.

Discoid Lupus Erythematosus

Discoid lupus erythematosus (DLE) is a chronic cutaneous lupus erythematosus variant with a female to male predominance of 3:1,4 and DLE lesions are prone to malignant transformation. Retrospective cohort studies have attempted to characterize who is at risk for SCC and how SCCs behave depending on their location. Cohorts from China,5 India,6 and Japan7 have noted a higher rate of SCC within DLE lesions in men (female to male ratios of 1:2.2, 1:1.6, and 1:2, respectively) and shorter lag times for SCC onset within DLE lesions of the lips (13, 5, and 10 years, respectively) compared to SCC arising in DLE elsewhere (19.2, 11.2, and 26 years, respectively). Studies have noted that DLE lesions of the lips may be prone to more rapid SCC tumorigenesis compared to DLE on cutaneous sites. One study reported SCC in DLE recurrence, metastasis, and death rates of 29%, 16.1%, and 19.4%, respectively,5 which exceeds reported rates in non-DLE SCCs (20%, 0.5% to 6%, and 1%, respectively).5,8

Because SCC arising within DLE is most common on the lips (Figure 1), it has been hypothesized that the high rate of transformation of DLE lesions on the lips may be due to constant exposure to irritation and tobacco, which may accelerate carcinogenesis.5 It also has been hypothesized that atrophic discoid lesions have lost sun protection and are more prone to mutagenic UV radiation,9 as SCCs arising in DLE lesions virtually always display prominent solar elastosis6; however, SCC has been observed to arise in non–sun-exposed DLE lesions in both White and Black patients.10

Invasive squamous cell carcinoma arising within a labial discoid lupus erythematosus lesion. This patient’s lesions were present for approximately 6 years prior to presentation for carcinoma.
Photograph courtesy of Andrea Murina, MD.
FIGURE 1. Invasive squamous cell carcinoma arising within a labial discoid lupus erythematosus lesion. This patient’s lesions were present for approximately 6 years prior to presentation for carcinoma.

Additionally, use of immunosuppressant medications may accelerate the emergence of malignancy or more aggressive forms of malignancy; however, patients with autoimmune disease have a greater risk for malignancy at baseline,11 thus making it difficult to determine the excess risk from medications. There also may be a role for human papillomavirus (HPV) accelerating SCC development in DLE lesions, as demonstrated in a case of SCC arising in DLE lesions of the ears, with viral staining evident within the tumors.12 However, testing for HPV is not routinely performed in these cases.

Dermatologists need to be aware of the relatively rapid tumorigenesis and aggressive behavior of transformation and aggression seen with SCC arising within orolabial DLE lesions compared to cutaneous lesions, especially those on the lips.

Lichen Planus

Although patients with typical cutaneous lichen planus lesions do not have an increased risk for SCC,13 variants of lichen planus may predispose patients to SCC.

 

 

Oral Lichen Planus—Oral lichen planus (OLP) lesions are prone to malignant transformation. A systematic review of 16 studies evaluating the risk for OLP-associated SCC revealed an overall transformation rate of 1.09%, with a mean lag time of 4.3 years,14 compared to a reference rate of 0.2% for oral SCC.15 A meta-analysis of 19,676 patients with OLP and other oral lichenoid lesions revealed an oral SCC rate of 1.1%, with higher rates of transformation seen in cigarette smokers, alcoholics, and patients with hepatitis C virus infection.16 The ulcerative subtype of OLP appears to present a greater risk for malignant transformation.15 Dermatologists also should be cognizant that treatments for OLP such as topical calcineurin inhibitors may support the development of malignancy within inflammatory lesions.17

Hypertrophic Lichen Planus—The hypertrophic variant of lichen planus (HLP) also is prone to malignant transformation. A 1991 epidemiologic study from Sweden of malignancy arising in lichen planus revealed a disproportionate number of cases arising in verrucous or hypertrophic lesions, with a mean of 12.2 years from onset of the dermatosis to malignancy diagnosis.13 A subsequent 2015 retrospective study of 38 patients revealed that SCC had a propensity for the lower limb, favoring the pretibial region and the calf over the foot and the ankle with a reported lag time of 11 years.18

Although metastatic SCC arising in HLP is rare, 2 cases have been reported. A 24-year-old woman presented with an HLP plaque on the lower leg that developed during childhood and rapidly enlarged 2 months prior to presentation; she eventually died from metastatic disease.19 In another case, a 34-year-old man presented with an HLP lesion of approximately 10 years’ duration. A well-differentiated SCC was excised, and he developed lymph node metastases 5 months later.20

It is important to note that HLP on the legs often is misdiagnosed as SCC, as pseudoepitheliomatous hyperplasia and squamous metaplasia can be difficult to differentiate clinically and histologically.21,22 In the case of multiple eruptive SCCs of the lower leg, clinical correlation is essential to avoid unnecessary and ineffective surgical treatment.

Patients with HLP may exhibit Wickham striae, follicular accentuation, and mucocutaneous lichen planus at other sites, or a correlative initiation of possible culprit medications.23 Because true SCC arising within HLP is relatively rare, its malignant potential is not as clear as those arising within DLE; however, the lower limb appears to be the most common location for SCC within HLP.Nail Lichen Planus—Squamous cell carcinoma arising in nail lichen planus is rare. A report of 2 patients were diagnosed with lichen planus approximately 15 years prior to diagnosis of ungual SCC.24 Given the rarity of this presentation, it is difficult to ascertain the approximate lag time and other risk factors. Furthermore, the role of HPV in these cases was not ruled out. Oncogenic HPV strains have been reported in patients with periungual SCC.25,26

Lichen Sclerosus

Lichen sclerosus (LS) is a chronic inflammatory dermatosis that favors the anogenital area in a female to male ratio of 10:1.27 It is considered a premalignant condition for SCC tumorigenesis and may be a strong predictor of vulvar SCC (Figure 2), as 62% of vulvar SCC cases (N=78) may have adjacent LS.28

Poorly differentiated squamous cell carcinoma arising within vulvar lichen sclerosus. This patient’s dermatosis was present for approximately 7 years prior to presentation for carcinoma.
Photograph courtesy of Laura C. Williams, MD (New Orleans, Louisiana).
FIGURE 2. Poorly differentiated squamous cell carcinoma arising within vulvar lichen sclerosus. This patient’s dermatosis was present for approximately 7 years prior to presentation for carcinoma.

In a Dutch cohort of 3038 women with LS, 2.6% of patients developed vulvar SCC at a median of 3.3 years after LS diagnosis.29 Other studies have estimated a lag time of 4 years until SCC presentation.30 An Italian cohort of 976 women similarly observed that 2.7% of patients developed premalignancy or SCC.31 It was previously estimated that 3% to 5% of patients with LS developed SCC; however, prior studies may have included cases of vulvar intraepithelial neoplasia with low risk for invasive SCC, which might have overestimated true risk of SCC.32 Another confounding factor for elucidating SCC on a background of LS may be the presence of HPV.33 Extragenital LS does not appear to have similar potential for malignant transformation.34

 

 

In a prospective Australian cohort of 507 women with LS (mean age, 55.4 years), remission was induced with potent topical corticosteroids.35 Patients who were adherent to a topical regimen did not develop SCC during follow-up. Those who were nonadherent or partially adherent had a 4.7% risk for SCC.35 In a similar prospective study of 83 women in France, the SCC rate was 9.6% in lesions that were untreated or irregularly treated.36 These studies provide essential evidence that appropriately treating LS can prevent SCC at a later date, though longer-term data are lacking.

The rate of SCC arising in male genital LS may approach 8.4%,37 with a lag time of 17 years from onset of LS to SCC diagnosis.38 Although circumcision often is considered curative for male genital LS, patients have been observed to develop penile SCC at least 5 years after circumcision.39 Male penile SCC in a background of LS may not necessarily be HPV associated.40

Marjolin Ulcer

Chronic ulcers or scars, typically postburn scars, may undergo malignant transformation, with SCC being the most common carcinoma.41 Squamous cell carcinoma in the context of a chronic ulcer or wound is known as a Marjolin ulcer (MU). Up to 2% of burn scars have been observed to undergo malignant transformation.42 Marjolin ulcers tend to behave aggressively once they form, and it has been proposed that removal of scar tissue may be a preventive therapeutic strategy.43 Cohort studies of MU on the lower extremities have observed lag times of 26.444 and 37.945 years, with both studies also noting relatively high rates of local recurrence.

The pathogenesis of MU appears to be multifactorial. Chronic inflammation and scar formation have been implicated. Chronic inflammation and irritation of lesions at natural creases are thought to increase mitotic activity,41 and local accumulation of toxin may promote mutagenesis.46 Scar formation may create a locally immunoprivileged site, allowing for developing tumors to evade the immune system47 and become even more aggressive as the tumor accumulates.48 Scar formation also may prevent the ability of immune cells to penetrate the tumor microenvironment and access lymphatic channels.49

Hidradenitis Suppurativa

As many as 3.2% of patients with chronic hidradenitis suppurativa (HS) experience malignant transformation to SCC.50 Early HS displays subclinical lymphedema in affected sites, which can progress to chronic fibrosis, stasis, and accumulation of protein-rich fluid.51 Stasis changes have been associated with altered local inflammatory proteins, such as toll-like receptors, β-defensins, and interleukins.52

A retrospective cohort study of 12 patients revealed a lag time of 28.5 years from HS diagnosis to the manifestation of malignancy.53 After local excision, 7 patients developed recurrence, with 100% mortality. Squamous cell carcinomas were well differentiated and moderately differentiated.53 A 2017 literature review of 62 case reports calculated a mean lag time of 27 years. Despite 85% of SCCs being well differentiated and moderately differentiated, nearly half of patients died within 2 years.54 As seen in other inflammatory conditions, HPV can complicate perineal HS and promote SCC tumorigenesis.55

Squamous cell carcinomas arising within HS lesions are more prevalent in males (6.75:1 ratio),54,56 despite HS being more prevalent in females (2:1 ratio).57 Similar to DLE, SCCs arising in HS are aggressive and are seen more in males, despite both conditions being female predominant. Incidence and mortality rates for primary cutaneous SCC are higher for men vs women58; however, the discordance in aggressive behavior seen more commonly in SCC arising from HS or DLE in male patients has yet to be explained.

 

 

Necrobiosis Lipoidica Diabeticorum

Malignancy arising within necrobiosis lipoidica diabeticorum (NLD) is rare. A review of 14 published cases noted that 13 were SCC and 1 was leiomyosarcoma.59 The lag time was 21.5 years; 31% of cases (N=14) presented with regional lymph node metastasis. Although chronic ulceration is a risk factor for SCC and occurs in as many as one-third of NLD cases, its correlation with ulceration and malignant transformation has not been characterized.

Epidermolysis Bullosa

Recessive dystrophic epidermolysis bullosa (RDEB) is a noninflammatory inherited blistering disease, and patients have an inherently high risk for aggressive SCC.60 Other forms of epidermolysis bullosa can lead to SCC, but the rarer RDEB accounts for 69% of SCC cases, with a median age of 36 years at presentation.61 Although SCCs tend to be well differentiated in RDEB (73.9%),61 they also exhibit highly aggressive behavior.62 In the most severe variant—RDEB-generalized severe—the cumulative risk for SCC-related death in an Australian population was 84.4% at 34 years of age.63

As RDEB is an inherited disorder with potential for malignancy at a young age, the pathogenesis is plausibly different from the previously discussed inflammatory dermatoses. This disease is characterized by a mutation in the collagen VII gene, leading to loss of anchoring fibrils and a basement membrane zone split.64 There also can be inherent fibroblast alterations; RDEB fibroblasts create an environment for tumor growth by supporting malignant-cell adhesion and invasion.65 Mutations in p53,66 local alterations in transforming growth factor β activity,67 and downstream matrix metalloproteinase activity68 have been implicated.

Additionally, keratinocytes may retain the N-terminal noncollagenous (NC1) domain of truncated collagen VII while losing the anchoring NC2 domain in mutated collagen VII RDEB, thereby supporting anchorless keratinocyte survival and higher metastatic potential.69 Retention of this truncated NC1 domain has shown conversion of RDEB keratinocytes to tumor in a xenotransplant mouse model.70 A high level of type VII collagen itself may inherently be protumorigenic for keratinocytes.71

There does not appear to be evidence for HPV involvement in RDEB-associated SCC.72 Squamous cell carcinoma development in RDEB appears to be multifactorial,73 but validated tumor models are lacking. Other than conventional oncologic therapy, future directions in the management of RDEB may include gene-, protein- and cell-targeted therapies.73

Conclusion

Squamous cell carcinomas are known to arise within chronic cutaneous inflammatory dermatoses. Tumorigenesis peaks relatively early in new orolabial DLE, LS, and OLP cases, and can occur over many decades in cutaneous DLE, HLP, HS, NLD, and chronic wounds or scars, summarized in the Table. Frequent SCCs are observed in high-risk subtypes of epidermolysis bullosa. Dermatologists must examine areas affected by these diseases at regular intervals, being mindful of the possibility of SCC development. Furthermore, dermatologists should adopt a lower threshold to biopsy suspicious lesions, especially those that develop within relatively new orolabial DLE, chronic HS, or chronic wound cases, as SCC in these settings is particularly aggressive and displays mortality and metastasis rates that exceed those of common cutaneous SCC.


References
  1. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007;121:2373-2380. doi:10.1002/ijc.23173
  2. Mantovani A, Allavena P, Sica A, et al. Cancer-related inflammation. Nature. 2008;454:436-444. doi:10.1038/nature07205
  3. Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development. Front Immunol. 2011;2:98. doi:10.3389/fimmu.2011.00098
  4. Tebbe B. Clinical course and prognosis of cutaneous lupus erythematosus. Clin Dermatol. 2004;22:121-124. doi:10.1016/j.clindermatol.2003.12.018
  5. Tao J, Zhang X, Guo N, et al. Squamous cell carcinoma complicating discoid lupus erythematosus in Chinese patients: review of the literature, 1964-2010. J Am Acad Dermatol. 2012;66:695-696. doi:10.1016 /j.jaad.2011.09.033
  6. Fernandes MS, Girisha BS, Viswanathan N, et al. Discoid lupus erythematosus with squamous cell carcinoma: a case report and review of the literature in Indian patients. Lupus. 2015;24:1562-1566. doi:10.1177/0961203315599245
  7. Makita E, Akasaka E, Sakuraba Y, et al. Squamous cell carcinoma on the lip arising from discoid lupus erythematosus: a case report and review of Japanese patients. Eur J Dermatol. 2016;26:395-396. doi:10.1684/ejd.2016.2780
  8. Clayman GL, Lee JJ, Holsinger FC, et al. Mortality risk from squamous cell skin cancer. J Clin Oncol. 2005;23:759-765. doi:10.1200/JCO.2005.02.155
  9. Arvanitidou I-E, Nikitakis NG, Georgaki M, et al. Multiple primary squamous cell carcinomas of the lower lip and tongue arising in discoid lupus erythematosus: a case report. Oral Surg Oral Med Oral Pathol Oral Radiol. 2018;125:e22-e30. doi:10.1016/j.oooo.2017.08.012
  10. Alsanafi S, Werth VP. Squamous cell carcinomas arising in discoid lupus erythematosus scars: unusual occurrence in an African-American and in a sun-protected area. J Clin Rheumatol. 2011;17:35-36. doi:10.1097/RHU.0b013e3182051928
  11. Goobie GC, Bernatsky S, Ramsey-Goldman R, et al. Malignancies in systemic lupus erythematosus: a 2015 update. Curr Opin Rheumatol. 2015;27:454-460. doi:10.1097/BOR.0000000000000202
  12. Simpson JK, Medina-Flores R, Deng J-S. Squamous cell carcinoma arising in discoid lupus erythematosus lesions of the ears infected with human papillomavirus. Cutis. 2010;86:195-198.
  13. Sigurgeirsson B, Lindelöf B. Lichen planus and malignancy. an epidemiologic study of 2071 patients and a review of the literature. Arch Dermatol. 1991;127:1684-1688. doi:10.1001/archderm.127.11.1684
  14. Fitzpatrick SG, Hirsch SA, Gordon SC. The malignant transformation of oral lichen planus and oral lichenoid lesions: a systematic review. J Am Dent Assoc. 2014;145:45-56. doi:10.14219/jada.2013.10
  15. Laniosz V, Torgerson RR, Ramos-Rodriguez AJ, et al. Incidence of squamous cell carcinoma in oral lichen planus: a 25-year population-based study. Int J Dermatol. 2019;58:296-301. doi:10.1111/ijd.14215
  16. Aghbari SMH, Abushouk AI, Attia A, et al. Malignant transformation of oral lichen planus and oral lichenoid lesions: a meta-analysis of 20095 patient data. Oral Oncol. 2017;68:92-102. doi:10.1016/j.oraloncology.2017.03.012
  17. Morita M, Asoda S, Tsunoda K, et al. The onset risk of carcinoma in patients continuing tacrolimus topical treatment for oral lichen planus: a case report. Odontology. 2017;105:262-266. doi:10.1007/s10266-016-0255-4
  18. Knackstedt TJ, Collins LK, Li Z, et al. Squamous cell carcinoma arising in hypertrophic lichen planus: a review and analysis of 38 cases. Dermatol Surg. 2015;41:1411-1418. doi:10.1097/DSS.0000000000000565
  19. Tong LX, Weinstock MJ, Drews R, et al. Widely metastatic squamous cell carcinoma originating from malignant transformation of hypertrophic lichen planus in a 24-year-old woman: case report and review of the literature. Pediatr Dermatol. 2015;32:e98-e101. doi:10.1111/pde.12549
  20. Ardabili M, Gambichler T, Rotterdam S, et al. Metastatic cutaneous squamous cell carcinoma arising from a previous area of chronic hypertrophic lichen planus. Dermatol Online J. 2003;9:10.
  21. Bowen AR, Burt L, Boucher K, et al. Use of proliferation rate, p53 staining and perforating elastic fibers in distinguishing keratoacanthoma from hypertrophic lichen planus: a pilot study. J Cutan Pathol. 2012;39:243-250. doi:10.1111/j.1600-0560.2011.01834.x
  22. Totonchy MB, Leventhal JS, Ko CJ, et al. Hypertrophic lichen planus and well-differentiated squamous cell carcinoma: a diagnostic conundrum. Dermatol Surg. 2018;44:1466-1470. doi:10.1097/DSS.0000000000001465
  23. Levandoski KA, Nazarian RM, Asgari MM. Hypertrophic lichen planus mimicking squamous cell carcinoma: the importance of clinicopathologic correlation. JAAD Case Rep. 2017;3:151-154. doi: 10.1016/j.jdcr.2017.01.020
  24. Okiyama N, Satoh T, Yokozeki H, et al. Squamous cell carcinoma arising from lichen planus of nail matrix and nail bed. J Am Acad Dermatol. 2005;53:908-909. doi:10.1016/j.jaad.2005.04.052
  25. Riddel C, Rashid R, Thomas V. Ungual and periungual human papillomavirus-associated squamous cell carcinoma: a review. J Am Acad Dermatol. 2011;64:1147-1153. doi:10.1016/j.jaad.2010.02.057
  26. Shimizu A, Kuriyama Y, Hasegawa M, et al. Nail squamous cell carcinoma: a hidden high-risk human papillomavirus reservoir for sexually transmitted infections. J Am Acad Dermatol. 2019;81:1358-1370. doi:10.1016/j.jaad.2019.03.070
  27. Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. J Am Acad Dermatol. 1995;32:393-416. doi:10.1016/0190-9622(95)90060-8
  28. Leibowitch M, Neill S, Pelisse M, et al. The epithelial changes associated with squamous cell carcinoma of the vulva: a review of the clinical, histological and viral findings in 78 women. Br J Obstet Gynaecol. 1990;97:1135-1139. doi:10.1111/j.1471-0528.1990.tb02502.x
  29. Bleeker MCG, Visser PJ, Overbeek LIH, et al. Lichen sclerosus: incidence and risk of vulvar squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev. 2016;25:1224-1230. doi:10.1158/1055-9965.EPI-16-0019
  30. Carlson JA, Ambros R, Malfetano J, et al. Vulvar lichen sclerosus and squamous cell carcinoma: a cohort, case control, and investigational study with historical perspective; implications for chronic inflammation and sclerosis in the development of neoplasia. Hum Pathol. 1998;29:932-948. doi:10.1016/s0046-8177(98)90198-8
  31. Micheletti L, Preti M, Radici G, et al. Vulvar lichen sclerosus and neoplastic transformation: a retrospective study of 976 cases. J Low Genit Tract Dis. 2016;20:180-183. doi:10.1097/LGT.0000000000000186
  32. Cooper SM, Madnani N, Margesson L. Reduced risk of squamous cell carcinoma with adequate treatment of vulvar lichen sclerosus. JAMA Dermatol. 2015;151:1059-1060. doi:10.1001/jamadermatol.2015.0644
  33. Rakislova N, Alemany L, Clavero O, et al; VVAP Study Group. Differentiated vulvar intraepithelial neoplasia-like and lichen sclerosus-like lesions in HPV-associated squamous cell carcinomas of the vulva. Am J Surg Pathol. 2018;42:828-835. doi:10.1097/PAS.0000000000001047
  34. Val I, Almeida G. An overview of lichen sclerosus. Clin Obstet Gynecol. 2005;48:808-817. doi:10.1097/01.grf.0000179635.64663.3d
  35. Lee A, Bradford J, Fischer G. Long-term management of adult vulvar lichen sclerosus: a prospective cohort study of 507 women. JAMA Dermatol. 2015;151:1061-1067. doi:10.1001/jamadermatol.2015.0643
  36. Renaud-Vilmer C, Cavelier-Balloy B, Porcher R, et al. Vulvar lichen sclerosus: effect of long-term topical application of a potent steroid on the course of the disease. Arch Dermatol. 2004;140:709-712. doi:10.1001/archderm.140.6.709
  37. Minhas S, Manseck A, Watya S, et al. Penile cancer—prevention and premalignant conditions. Urology. 2010;76(2 suppl 1):S24-S35. doi:10.1016/j.urology.2010.04.007
  38. Nasca MR, Innocenzi D, Micali G. Penile cancer among patients with genital lichen sclerosus. J Am Acad Dermatol. 1999;41:911-914. doi:10.1016/s0190-9622(99)70245-8
  39. Philippou P, Shabbir M, Ralph DJ, et al. Genital lichen sclerosus/balanitis xerotica obliterans in men with penile carcinoma: a critical analysis. BJU Int. 2013;111:970-976. doi:10.1111/j.1464-410X.2012.11773.x
  40. Velazquez EF, Cubilla AL. Lichen sclerosus in 68 patients with squamous cell carcinoma of the penis: frequent atypias and correlation with special carcinoma variants suggests a precancerous role. Am J Surg Pathol. 2003;27:1448-1453. doi:10.1097/00000478-200311000-00007
  41. Pekarek B, Buck S, Osher L. A comprehensive review on Marjolin’s ulcers: diagnosis and treatment. J Am Col Certif Wound Spec. 2011;3:60-64. doi:10.1016/j.jcws.2012.04.001
  42. Aydogdu E, Yildirim S, Akoz T. Is surgery an effective and adequate treatment in advanced Marjolin’s ulcer? Burns. 2005;31:421-431. doi:10.1016/j.burns.2005.02.008
  43. Xiao H, Deng K, Liu R, et al. A review of 31 cases of Marjolin’s ulcer on scalp: is it necessary to preventively remove the scar? Int Wound J. 2019;16:479-485. doi:10.1111/iwj.13058
  44. Chaturvedi G, Gupta AK, Das S, et al. Marjolin ulcer: an observational epidemiological study from a tertiary care centre in India. Ann Plast Surg. 2019;83:518-522. doi:10.1097/SAP.0000000000001995
  45. Karasoy Yesilada A, Zeynep Sevim K, Özgur Sucu D, et al. Marjolin ulcer: clinical experience with 34 patients over 15 years. J Cutan Med Surg. 2013;17:404-409. doi:10.2310/7750.2013.13016
  46. Bazalin´ski D, Przybek-Mita J, Baran´ska B, et al. Marjolin’s ulcer in chronic wounds - review of available literature. Contemp Oncol (Pozn). 2017;21:197-202. doi:10.5114/wo.2017.70109
  47. Visuthikosol V, Boonpucknavig V, Nitiyanant P. Squamous carcinoma in scars: clinicopathological correlations. Ann Plast Surg. 1986;16:42-48. doi:10.1097/00000637-198601000-00004
  48. Bostwick J 3rd, Pendergrast WJ Jr, Vasconez LO. Marjolin’s ulcer: an immunologically privileged tumor? Plast Reconstr Surg. 1976;57:66-69.
  49. Kerr-Valentic MA, Samimi K, Rohlen BH, et al. Marjolin’s ulcer: modern analysis of an ancient problem. Plast Reconstr Surg. 2009;123:184-191. doi:10.1097/PRS.0b013e3181904d86
  50. Constantinou C, Widom K, Desantis J, et al. Hidradenitis suppurativa complicated by squamous cell carcinoma. Am Surg. 2008;74:1177-1181.
  51. Fabbrocini G, Ruocco E, De Vita V, et al. Squamous cell carcinoma arising in long-standing hidradenitis suppurativa: an overlooked facet of the immunocompromised district. Clin Dermatol. 2017;35:225-227. doi:10.1016/j.clindermatol.2016.10.019
  52. Baroni A, Buommino E, Piccolo V, et al. Alterations of skin innate immunity in lymphedematous limbs: correlations with opportunistic diseases. Clin Dermatol. 2014;32:592-598. doi:10.1016/j.clindermatol.2014.04.006
  53. Kohorst JJ, Shah KK, Hallemeier CL, et al. Squamous cell carcinoma in perineal, perianal, and gluteal hidradenitis suppurativa: experience in 12 patients. Dermatol Surg. 2019;45:519-526. doi:10.1097/DSS.0000000000001713
  54. Huang C, Lai Z, He M, et al. Successful surgical treatment for squamous cell carcinoma arising from hidradenitis suppurativa: a case report and literature review. Medicine (Baltimore). 2017;96:e5857. doi:10.1097/MD.0000000000005857
  55. Lavogiez C, Delaporte E, Darras-Vercambre S, et al. Clinicopathological study of 13 cases of squamous cell carcinoma complicating hidradenitis suppurativa. Dermatology. 2010;220:147-153. doi:10.1159/000269836
  56. Makris G-M, Poulakaki N, Papanota A-M, et al. Vulvar, perianal and perineal cancer after hidradenitis suppurativa: a systematic review and pooled analysis. Dermatol Surg. 2017;43:107-115. doi:10.1097/DSS.0000000000000944
  57. Cosmatos I, Matcho A, Weinstein R, et al. Analysis of patient claims data to determine the prevalence of hidradenitis suppurativa in the United States. J Am Acad Dermatol. 2013;68:412-419. doi:10.1016/j.jaad.2012.07.027
  58. Hollestein LM, de Vries E, Nijsten T. Trends of cutaneous squamous cell carcinoma in the Netherlands: increased incidence rates, but stable relative survival and mortality 1989-2008. Eur J Cancer. 2012;48:2046-2053. doi:10.1016/j.ejca.2012.01.003
  59. Uva L, Freitas J, Soares de Almeida L, et al. Squamous cell carcinoma arising in ulcerated necrobiosis lipoidica diabeticorum. Int Wound J. 2015;12:741-743. doi:10.1111/iwj.12206
  60. McGrath JA, Schofield OM, Mayou BJ, et al. Epidermolysis bullosa complicated by squamous cell carcinoma: report of 10 cases. J Cutan Pathol. 1992;19:116-123. doi:10.1111/j.1600-0560.1992.tb01352.x
  61. Montaudié H, Chiaverini C, Sbidian E, et al. Inherited epidermolysis bullosa and squamous cell carcinoma: a systematic review of 117 cases. Orphanet J Rare Dis. 2016;11:117. doi:10.1186/s13023-016-0489-9.
  62. Fine J-D. Inherited epidermolysis bullosa: past, present, and future. Ann N Y Acad Sci. 2010;1194:213-222. doi:10.1111/j.1749-6632.2010.05463.x
  63. Kim M, Li M, Intong-Wheeler LRA, et al. Epidemiology and outcome of squamous cell carcinoma in epidermolysis bullosa in Australia and New Zealand. Acta Derm Venereol. 2018;98:70-76. doi:10.2340/00015555-2781
  64. Bruckner-Tuderman L, Mitsuhashi Y, Schnyder UW, et al. Anchoring fibrils and type VII collagen are absent from skin in severe recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 1989;93:3-9. doi:10.1111/1523-1747.ep12277331
  65. Ng Y-Z, Pourreyron C, Salas-Alanis JC, et al. Fibroblast-derived dermal matrix drives development of aggressive cutaneous squamous cell carcinoma in patients with recessive dystrophic epidermolysis bullosa. Cancer Res. 2012;72:3522-3534. doi:10.1158/0008-5472.CAN-11-2996
  66. Arbiser JL, Fan C-Y, Su X, et al. Involvement of p53 and p16 tumor suppressor genes in recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2004;123:788-790. doi:10.1111/j.0022-202X.2004.23418.x
  67. Knaup J, Gruber C, Krammer B, et al. TGFbeta-signaling in squamous cell carcinoma occurring in recessive dystrophic epidermolysis bullosa. Anal Cell Pathol (Amst). 2011;34:339-353. doi:10.3233/ACP-2011-0039
  68. Kivisaari AK, Kallajoki M, Mirtti T, et al. Transformation-specific matrix metalloproteinases (MMP)-7 and MMP-13 are expressed by tumour cells in epidermolysis bullosa-associated squamous cell carcinomas. Br J Dermatol. 2008;158:778-785. doi:10.1111/j.1365-2133.2008.08466.x
  69. Rodeck U, Fertala A, Uitto J. Anchorless keratinocyte survival: an emerging pathogenic mechanism for squamous cell carcinoma in recessive dystrophic epidermolysis bullosa. Exp Dermatol. 2007;16:465-467. doi:10.1111/j.1600-0625.2007.00563.x
  70. Ortiz-Urda S, Garcia J, Green CL, et al. Type VII collagen is required for Ras-driven human epidermal tumorigenesis. Science. 2005;307:1773-1776. doi:10.1126/science.1106209
  71. Pourreyron C, Chen M, McGrath JA, et al. High levels of type VII collagen expression in recessive dystrophic epidermolysis bullosa cutaneous squamous cell carcinoma keratinocytes increases PI3K and MAPK signalling, cell migration and invasion. Br J Dermatol. 2014;170:1256-1265. doi:10.1111/bjd.12715
  72. Purdie KJ, Pourreyron C, Fassihi H, et al. No evidence that human papillomavirus is responsible for the aggressive nature of recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2010;130:2853-2855. doi:10.1038/jid.2010.243
  73. South AP, O’Toole EA. Understanding the pathogenesis of recessive dystrophic epidermolysis bullosa squamous cell carcinoma. Dermatol Clin. 2010;28:171-178. doi:10.1016/j.det.2009.10.023
References
  1. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007;121:2373-2380. doi:10.1002/ijc.23173
  2. Mantovani A, Allavena P, Sica A, et al. Cancer-related inflammation. Nature. 2008;454:436-444. doi:10.1038/nature07205
  3. Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development. Front Immunol. 2011;2:98. doi:10.3389/fimmu.2011.00098
  4. Tebbe B. Clinical course and prognosis of cutaneous lupus erythematosus. Clin Dermatol. 2004;22:121-124. doi:10.1016/j.clindermatol.2003.12.018
  5. Tao J, Zhang X, Guo N, et al. Squamous cell carcinoma complicating discoid lupus erythematosus in Chinese patients: review of the literature, 1964-2010. J Am Acad Dermatol. 2012;66:695-696. doi:10.1016 /j.jaad.2011.09.033
  6. Fernandes MS, Girisha BS, Viswanathan N, et al. Discoid lupus erythematosus with squamous cell carcinoma: a case report and review of the literature in Indian patients. Lupus. 2015;24:1562-1566. doi:10.1177/0961203315599245
  7. Makita E, Akasaka E, Sakuraba Y, et al. Squamous cell carcinoma on the lip arising from discoid lupus erythematosus: a case report and review of Japanese patients. Eur J Dermatol. 2016;26:395-396. doi:10.1684/ejd.2016.2780
  8. Clayman GL, Lee JJ, Holsinger FC, et al. Mortality risk from squamous cell skin cancer. J Clin Oncol. 2005;23:759-765. doi:10.1200/JCO.2005.02.155
  9. Arvanitidou I-E, Nikitakis NG, Georgaki M, et al. Multiple primary squamous cell carcinomas of the lower lip and tongue arising in discoid lupus erythematosus: a case report. Oral Surg Oral Med Oral Pathol Oral Radiol. 2018;125:e22-e30. doi:10.1016/j.oooo.2017.08.012
  10. Alsanafi S, Werth VP. Squamous cell carcinomas arising in discoid lupus erythematosus scars: unusual occurrence in an African-American and in a sun-protected area. J Clin Rheumatol. 2011;17:35-36. doi:10.1097/RHU.0b013e3182051928
  11. Goobie GC, Bernatsky S, Ramsey-Goldman R, et al. Malignancies in systemic lupus erythematosus: a 2015 update. Curr Opin Rheumatol. 2015;27:454-460. doi:10.1097/BOR.0000000000000202
  12. Simpson JK, Medina-Flores R, Deng J-S. Squamous cell carcinoma arising in discoid lupus erythematosus lesions of the ears infected with human papillomavirus. Cutis. 2010;86:195-198.
  13. Sigurgeirsson B, Lindelöf B. Lichen planus and malignancy. an epidemiologic study of 2071 patients and a review of the literature. Arch Dermatol. 1991;127:1684-1688. doi:10.1001/archderm.127.11.1684
  14. Fitzpatrick SG, Hirsch SA, Gordon SC. The malignant transformation of oral lichen planus and oral lichenoid lesions: a systematic review. J Am Dent Assoc. 2014;145:45-56. doi:10.14219/jada.2013.10
  15. Laniosz V, Torgerson RR, Ramos-Rodriguez AJ, et al. Incidence of squamous cell carcinoma in oral lichen planus: a 25-year population-based study. Int J Dermatol. 2019;58:296-301. doi:10.1111/ijd.14215
  16. Aghbari SMH, Abushouk AI, Attia A, et al. Malignant transformation of oral lichen planus and oral lichenoid lesions: a meta-analysis of 20095 patient data. Oral Oncol. 2017;68:92-102. doi:10.1016/j.oraloncology.2017.03.012
  17. Morita M, Asoda S, Tsunoda K, et al. The onset risk of carcinoma in patients continuing tacrolimus topical treatment for oral lichen planus: a case report. Odontology. 2017;105:262-266. doi:10.1007/s10266-016-0255-4
  18. Knackstedt TJ, Collins LK, Li Z, et al. Squamous cell carcinoma arising in hypertrophic lichen planus: a review and analysis of 38 cases. Dermatol Surg. 2015;41:1411-1418. doi:10.1097/DSS.0000000000000565
  19. Tong LX, Weinstock MJ, Drews R, et al. Widely metastatic squamous cell carcinoma originating from malignant transformation of hypertrophic lichen planus in a 24-year-old woman: case report and review of the literature. Pediatr Dermatol. 2015;32:e98-e101. doi:10.1111/pde.12549
  20. Ardabili M, Gambichler T, Rotterdam S, et al. Metastatic cutaneous squamous cell carcinoma arising from a previous area of chronic hypertrophic lichen planus. Dermatol Online J. 2003;9:10.
  21. Bowen AR, Burt L, Boucher K, et al. Use of proliferation rate, p53 staining and perforating elastic fibers in distinguishing keratoacanthoma from hypertrophic lichen planus: a pilot study. J Cutan Pathol. 2012;39:243-250. doi:10.1111/j.1600-0560.2011.01834.x
  22. Totonchy MB, Leventhal JS, Ko CJ, et al. Hypertrophic lichen planus and well-differentiated squamous cell carcinoma: a diagnostic conundrum. Dermatol Surg. 2018;44:1466-1470. doi:10.1097/DSS.0000000000001465
  23. Levandoski KA, Nazarian RM, Asgari MM. Hypertrophic lichen planus mimicking squamous cell carcinoma: the importance of clinicopathologic correlation. JAAD Case Rep. 2017;3:151-154. doi: 10.1016/j.jdcr.2017.01.020
  24. Okiyama N, Satoh T, Yokozeki H, et al. Squamous cell carcinoma arising from lichen planus of nail matrix and nail bed. J Am Acad Dermatol. 2005;53:908-909. doi:10.1016/j.jaad.2005.04.052
  25. Riddel C, Rashid R, Thomas V. Ungual and periungual human papillomavirus-associated squamous cell carcinoma: a review. J Am Acad Dermatol. 2011;64:1147-1153. doi:10.1016/j.jaad.2010.02.057
  26. Shimizu A, Kuriyama Y, Hasegawa M, et al. Nail squamous cell carcinoma: a hidden high-risk human papillomavirus reservoir for sexually transmitted infections. J Am Acad Dermatol. 2019;81:1358-1370. doi:10.1016/j.jaad.2019.03.070
  27. Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. J Am Acad Dermatol. 1995;32:393-416. doi:10.1016/0190-9622(95)90060-8
  28. Leibowitch M, Neill S, Pelisse M, et al. The epithelial changes associated with squamous cell carcinoma of the vulva: a review of the clinical, histological and viral findings in 78 women. Br J Obstet Gynaecol. 1990;97:1135-1139. doi:10.1111/j.1471-0528.1990.tb02502.x
  29. Bleeker MCG, Visser PJ, Overbeek LIH, et al. Lichen sclerosus: incidence and risk of vulvar squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev. 2016;25:1224-1230. doi:10.1158/1055-9965.EPI-16-0019
  30. Carlson JA, Ambros R, Malfetano J, et al. Vulvar lichen sclerosus and squamous cell carcinoma: a cohort, case control, and investigational study with historical perspective; implications for chronic inflammation and sclerosis in the development of neoplasia. Hum Pathol. 1998;29:932-948. doi:10.1016/s0046-8177(98)90198-8
  31. Micheletti L, Preti M, Radici G, et al. Vulvar lichen sclerosus and neoplastic transformation: a retrospective study of 976 cases. J Low Genit Tract Dis. 2016;20:180-183. doi:10.1097/LGT.0000000000000186
  32. Cooper SM, Madnani N, Margesson L. Reduced risk of squamous cell carcinoma with adequate treatment of vulvar lichen sclerosus. JAMA Dermatol. 2015;151:1059-1060. doi:10.1001/jamadermatol.2015.0644
  33. Rakislova N, Alemany L, Clavero O, et al; VVAP Study Group. Differentiated vulvar intraepithelial neoplasia-like and lichen sclerosus-like lesions in HPV-associated squamous cell carcinomas of the vulva. Am J Surg Pathol. 2018;42:828-835. doi:10.1097/PAS.0000000000001047
  34. Val I, Almeida G. An overview of lichen sclerosus. Clin Obstet Gynecol. 2005;48:808-817. doi:10.1097/01.grf.0000179635.64663.3d
  35. Lee A, Bradford J, Fischer G. Long-term management of adult vulvar lichen sclerosus: a prospective cohort study of 507 women. JAMA Dermatol. 2015;151:1061-1067. doi:10.1001/jamadermatol.2015.0643
  36. Renaud-Vilmer C, Cavelier-Balloy B, Porcher R, et al. Vulvar lichen sclerosus: effect of long-term topical application of a potent steroid on the course of the disease. Arch Dermatol. 2004;140:709-712. doi:10.1001/archderm.140.6.709
  37. Minhas S, Manseck A, Watya S, et al. Penile cancer—prevention and premalignant conditions. Urology. 2010;76(2 suppl 1):S24-S35. doi:10.1016/j.urology.2010.04.007
  38. Nasca MR, Innocenzi D, Micali G. Penile cancer among patients with genital lichen sclerosus. J Am Acad Dermatol. 1999;41:911-914. doi:10.1016/s0190-9622(99)70245-8
  39. Philippou P, Shabbir M, Ralph DJ, et al. Genital lichen sclerosus/balanitis xerotica obliterans in men with penile carcinoma: a critical analysis. BJU Int. 2013;111:970-976. doi:10.1111/j.1464-410X.2012.11773.x
  40. Velazquez EF, Cubilla AL. Lichen sclerosus in 68 patients with squamous cell carcinoma of the penis: frequent atypias and correlation with special carcinoma variants suggests a precancerous role. Am J Surg Pathol. 2003;27:1448-1453. doi:10.1097/00000478-200311000-00007
  41. Pekarek B, Buck S, Osher L. A comprehensive review on Marjolin’s ulcers: diagnosis and treatment. J Am Col Certif Wound Spec. 2011;3:60-64. doi:10.1016/j.jcws.2012.04.001
  42. Aydogdu E, Yildirim S, Akoz T. Is surgery an effective and adequate treatment in advanced Marjolin’s ulcer? Burns. 2005;31:421-431. doi:10.1016/j.burns.2005.02.008
  43. Xiao H, Deng K, Liu R, et al. A review of 31 cases of Marjolin’s ulcer on scalp: is it necessary to preventively remove the scar? Int Wound J. 2019;16:479-485. doi:10.1111/iwj.13058
  44. Chaturvedi G, Gupta AK, Das S, et al. Marjolin ulcer: an observational epidemiological study from a tertiary care centre in India. Ann Plast Surg. 2019;83:518-522. doi:10.1097/SAP.0000000000001995
  45. Karasoy Yesilada A, Zeynep Sevim K, Özgur Sucu D, et al. Marjolin ulcer: clinical experience with 34 patients over 15 years. J Cutan Med Surg. 2013;17:404-409. doi:10.2310/7750.2013.13016
  46. Bazalin´ski D, Przybek-Mita J, Baran´ska B, et al. Marjolin’s ulcer in chronic wounds - review of available literature. Contemp Oncol (Pozn). 2017;21:197-202. doi:10.5114/wo.2017.70109
  47. Visuthikosol V, Boonpucknavig V, Nitiyanant P. Squamous carcinoma in scars: clinicopathological correlations. Ann Plast Surg. 1986;16:42-48. doi:10.1097/00000637-198601000-00004
  48. Bostwick J 3rd, Pendergrast WJ Jr, Vasconez LO. Marjolin’s ulcer: an immunologically privileged tumor? Plast Reconstr Surg. 1976;57:66-69.
  49. Kerr-Valentic MA, Samimi K, Rohlen BH, et al. Marjolin’s ulcer: modern analysis of an ancient problem. Plast Reconstr Surg. 2009;123:184-191. doi:10.1097/PRS.0b013e3181904d86
  50. Constantinou C, Widom K, Desantis J, et al. Hidradenitis suppurativa complicated by squamous cell carcinoma. Am Surg. 2008;74:1177-1181.
  51. Fabbrocini G, Ruocco E, De Vita V, et al. Squamous cell carcinoma arising in long-standing hidradenitis suppurativa: an overlooked facet of the immunocompromised district. Clin Dermatol. 2017;35:225-227. doi:10.1016/j.clindermatol.2016.10.019
  52. Baroni A, Buommino E, Piccolo V, et al. Alterations of skin innate immunity in lymphedematous limbs: correlations with opportunistic diseases. Clin Dermatol. 2014;32:592-598. doi:10.1016/j.clindermatol.2014.04.006
  53. Kohorst JJ, Shah KK, Hallemeier CL, et al. Squamous cell carcinoma in perineal, perianal, and gluteal hidradenitis suppurativa: experience in 12 patients. Dermatol Surg. 2019;45:519-526. doi:10.1097/DSS.0000000000001713
  54. Huang C, Lai Z, He M, et al. Successful surgical treatment for squamous cell carcinoma arising from hidradenitis suppurativa: a case report and literature review. Medicine (Baltimore). 2017;96:e5857. doi:10.1097/MD.0000000000005857
  55. Lavogiez C, Delaporte E, Darras-Vercambre S, et al. Clinicopathological study of 13 cases of squamous cell carcinoma complicating hidradenitis suppurativa. Dermatology. 2010;220:147-153. doi:10.1159/000269836
  56. Makris G-M, Poulakaki N, Papanota A-M, et al. Vulvar, perianal and perineal cancer after hidradenitis suppurativa: a systematic review and pooled analysis. Dermatol Surg. 2017;43:107-115. doi:10.1097/DSS.0000000000000944
  57. Cosmatos I, Matcho A, Weinstein R, et al. Analysis of patient claims data to determine the prevalence of hidradenitis suppurativa in the United States. J Am Acad Dermatol. 2013;68:412-419. doi:10.1016/j.jaad.2012.07.027
  58. Hollestein LM, de Vries E, Nijsten T. Trends of cutaneous squamous cell carcinoma in the Netherlands: increased incidence rates, but stable relative survival and mortality 1989-2008. Eur J Cancer. 2012;48:2046-2053. doi:10.1016/j.ejca.2012.01.003
  59. Uva L, Freitas J, Soares de Almeida L, et al. Squamous cell carcinoma arising in ulcerated necrobiosis lipoidica diabeticorum. Int Wound J. 2015;12:741-743. doi:10.1111/iwj.12206
  60. McGrath JA, Schofield OM, Mayou BJ, et al. Epidermolysis bullosa complicated by squamous cell carcinoma: report of 10 cases. J Cutan Pathol. 1992;19:116-123. doi:10.1111/j.1600-0560.1992.tb01352.x
  61. Montaudié H, Chiaverini C, Sbidian E, et al. Inherited epidermolysis bullosa and squamous cell carcinoma: a systematic review of 117 cases. Orphanet J Rare Dis. 2016;11:117. doi:10.1186/s13023-016-0489-9.
  62. Fine J-D. Inherited epidermolysis bullosa: past, present, and future. Ann N Y Acad Sci. 2010;1194:213-222. doi:10.1111/j.1749-6632.2010.05463.x
  63. Kim M, Li M, Intong-Wheeler LRA, et al. Epidemiology and outcome of squamous cell carcinoma in epidermolysis bullosa in Australia and New Zealand. Acta Derm Venereol. 2018;98:70-76. doi:10.2340/00015555-2781
  64. Bruckner-Tuderman L, Mitsuhashi Y, Schnyder UW, et al. Anchoring fibrils and type VII collagen are absent from skin in severe recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 1989;93:3-9. doi:10.1111/1523-1747.ep12277331
  65. Ng Y-Z, Pourreyron C, Salas-Alanis JC, et al. Fibroblast-derived dermal matrix drives development of aggressive cutaneous squamous cell carcinoma in patients with recessive dystrophic epidermolysis bullosa. Cancer Res. 2012;72:3522-3534. doi:10.1158/0008-5472.CAN-11-2996
  66. Arbiser JL, Fan C-Y, Su X, et al. Involvement of p53 and p16 tumor suppressor genes in recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2004;123:788-790. doi:10.1111/j.0022-202X.2004.23418.x
  67. Knaup J, Gruber C, Krammer B, et al. TGFbeta-signaling in squamous cell carcinoma occurring in recessive dystrophic epidermolysis bullosa. Anal Cell Pathol (Amst). 2011;34:339-353. doi:10.3233/ACP-2011-0039
  68. Kivisaari AK, Kallajoki M, Mirtti T, et al. Transformation-specific matrix metalloproteinases (MMP)-7 and MMP-13 are expressed by tumour cells in epidermolysis bullosa-associated squamous cell carcinomas. Br J Dermatol. 2008;158:778-785. doi:10.1111/j.1365-2133.2008.08466.x
  69. Rodeck U, Fertala A, Uitto J. Anchorless keratinocyte survival: an emerging pathogenic mechanism for squamous cell carcinoma in recessive dystrophic epidermolysis bullosa. Exp Dermatol. 2007;16:465-467. doi:10.1111/j.1600-0625.2007.00563.x
  70. Ortiz-Urda S, Garcia J, Green CL, et al. Type VII collagen is required for Ras-driven human epidermal tumorigenesis. Science. 2005;307:1773-1776. doi:10.1126/science.1106209
  71. Pourreyron C, Chen M, McGrath JA, et al. High levels of type VII collagen expression in recessive dystrophic epidermolysis bullosa cutaneous squamous cell carcinoma keratinocytes increases PI3K and MAPK signalling, cell migration and invasion. Br J Dermatol. 2014;170:1256-1265. doi:10.1111/bjd.12715
  72. Purdie KJ, Pourreyron C, Fassihi H, et al. No evidence that human papillomavirus is responsible for the aggressive nature of recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2010;130:2853-2855. doi:10.1038/jid.2010.243
  73. South AP, O’Toole EA. Understanding the pathogenesis of recessive dystrophic epidermolysis bullosa squamous cell carcinoma. Dermatol Clin. 2010;28:171-178. doi:10.1016/j.det.2009.10.023
Issue
Cutis - 113(1)
Issue
Cutis - 113(1)
Page Number
29-34
Page Number
29-34
Publications
Publications
Topics
Article Type
Display Headline
Squamous Cell Carcinoma Arising in Chronic Inflammatory Dermatoses
Display Headline
Squamous Cell Carcinoma Arising in Chronic Inflammatory Dermatoses
Sections
Inside the Article

PRACTICE POINTS

  • Squamous cell carcinoma can develop within chronic inflammatory dermatoses.
  • Orolabial discoid lupus erythematosus (DLE), oral lichen planus, and lichen sclerosus can lead to relatively rapid tumorigenesis. Squamous cell carcinoma arising in cutaneous DLE, hidradenitis suppurativa (HS), necrobiosis lipoidica, chronic wounds, and hypertrophic lichen planus tends to appear after decades of inflammation.
  • Be especially mindful of new orolabial DLE cases and chronic cases of HS and Marjolin ulcer because malignancies in these settings are particularly aggressive.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Multiple New-Onset Pyogenic Granulomas During Treatment With Paclitaxel and Ramucirumab

Article Type
Changed
Thu, 01/04/2024 - 13:30
Display Headline
Multiple New-Onset Pyogenic Granulomas During Treatment With Paclitaxel and Ramucirumab

To the Editor:

Pyogenic granuloma (PG) is a benign vascular tumor that clinically is characterized as a small eruptive friable papule.1 Lesions typically are solitary and most commonly occur in children but also are associated with pregnancy; trauma to the skin or mucosa; and use of certain medications such as isotretinoin, capecitabine, vemurafenib, or indinavir.1 Numerous antineoplastic medications have been associated with the development of solitary PGs, including the taxane mitotic inhibitor paclitaxel (PTX) and the vascular endothelial growth factor receptor 2 (VEGFR2) monoclonal antibody ramucirumab.2 We report a case of multiple PGs in a patient undergoing treatment with PTX and ramucirumab.

New-onset pyogenic granuloma on the left cheek following combination therapy with paclitaxel and ramucirumab.
FIGURE 1. New-onset pyogenic granuloma on the left cheek following combination therapy with paclitaxel and ramucirumab.

A 59-year-old woman presented to the dermatology clinic with red, itchy, bleeding skin lesions on the breast, superior chest, left cheek, and forearm of 1 month’s duration. She denied any preceding trauma to the areas. Her medical history was notable for gastroesophageal junction adenocarcinoma diagnosed more than 2 years prior to presentation. Her original treatment regimen included nivolumab, which was discontinued for unknown reasons 5 months prior to presentation, and she was started on combination therapy with PTX and ramucirumab at that time. She noted the formation of small red papules 2 months after the initiation of PTX-ramucirumab combination therapy, which grew larger over the course of the next month. Physical examination revealed 5 friable hemorrhagic papules and nodules ranging in size from 3 to 10 mm on the chest, cheek, and forearm consistent with PGs (Figure 1). Several scattered cherry angiomas were noted on the scalp and torso, but the patient reported these were not new. Biopsies of the PGs demonstrated lobular aggregates of small-caliber vessels set in an edematous inflamed stroma and partially enclosed by small collarettes of adnexal epithelium, confirming the clinical diagnosis of multiple PGs (Figure 2).

Histopathology from the left cheek, medial breast, and medial superior chest demonstrated lobular aggregates of small-caliber vessels set in an edematous inflamed stroma and partially enclosed by small collarettes of adnexal epithelium
FIGURE 2. A–C, Histopathology from the left cheek, medial breast, and medial superior chest demonstrated lobular aggregates of small-caliber vessels set in an edematous inflamed stroma and partially enclosed by small collarettes of adnexal epithelium, consistent with pyogenic granuloma (H&E; original magnifications ×40, ×100, and ×40, respectively).

The first case of PTX-associated PG was reported in 2012.3 Based on a PubMed search of articles indexed for MEDLINE using the terms pyogenic granuloma, lobular capillary hemangioma, paclitaxel, taxane, and ramucirumab, there have been 9 cases of solitary PG development in the setting of PTX alone or in combination with ramucirumab since 2019 (Table).3-8 Pyogenic granulomas reported in patients who were treated exclusively with PTX were subungual, while the cases resulting from combined therapy were present on the scalp, face, oral mucosa, and surfaces of the hands sparing the nails. Ibe et al6 reported PG in a patient who received ramucirumab therapy without PTX but in combination with another taxane, docetaxel, which itself has been reported to cause subungual PG when used alone.9 Our case of the simultaneous development of multiple PGs in the setting of combined PTX and ramucirumab therapy added to the cutaneous distributions for which therapy-induced PGs have been observed (Table).

Pyogenic Granulomas in Patients Undergoing Treatment With Paclitaxel and Ramucirumab

The development of PG, a vascular tumor, during treatment with the VEGFR2 inhibitor ramucirumab—whose mechanism of action is to inhibit angioneogenesis—is inherently paradoxical. In 2015, a rapidly expanding angioma with a mutation in the kinase domain receptor gene, KDR, that encodes VEGFR2 was identified in a patient undergoing ramucirumab therapy. The authors suggested that KDR mutation resulted in paradoxical activation of VEGFR2 in the setting of ramucirumab therapy.10 Since then, ramucirumab and PTX were suggested to have a synergistic effect in vascular proliferation,5 though an exact mechanism has not been proposed. Other authors have identified increased expression of VEGFR2 in biopsy specimens of PG during combined ramucirumab and taxane therapy.6 Although genetic studies have not been used to evaluate for the presence of KDR mutations specifically in our patient population, it is possible that patients who develop PG and other vascular tumors during combined taxane and ramucirumab therapy have a mutation that makes them more susceptible to VEGFR2 upregulation. UV exposure may have a role in the formation of PG in patients on combined ramucirumab and taxane therapy7; however, our patient’s lesions were distributed on both sun-exposed and unexposed areas. Although potential clinical implications have not yet been thoroughly investigated, following long-term outcomes for these patients may provide important information on the efficacy of the antineoplastic regimen in the subset of patients who develop cutaneous vascular tumors during antiangiogenic treatment.

Combination therapy with PTX and ramucirumab has been associated with the paradoxical development of cutaneous vascular tumors. We report a case of multiple new-onset PGs in a patient undergoing this treatment regimen.

References
  1. Elston D, Neuhaus I, James WD, et al. Andrews’ Diseases of the Skin: Clinical Dermatology. 13th ed. Elsevier; 2020.
  2. Pierson JC. Pyogenic granuloma (lobular capillary hemangioma) clinical presentation. Medscape. Updated February 21, 2020. Accessed December 26, 2023. https://emedicine.medscape.com/article/1084701-clinical#showall
  3. Paul LJ, Cohen PR. Paclitaxel-associated subungual pyogenic granuloma: report in a patient with breast cancer receiving paclitaxel and review of drug-induced pyogenic granulomas adjacent to and beneath the nail. J Drugs Dermatol. 2012;11:262-268.
  4. Alessandrini A, Starace M, Cerè G, et al. Management and outcome of taxane-induced nail side effects: experience of 79 patients from a single centre. Skin Appendage Disord. 2019;5:276-282.
  5. Watanabe R, Nakano E, Kawazoe A, et al. Four cases of paradoxical cephalocervical pyogenic granuloma during treatment with paclitaxel and ramucirumab. J Dermatol. 2019;46:E178-E180.
  6. Ibe T, Hamamoto Y, Takabatake M, et al. Development of pyogenic granuloma with strong vascular endothelial growth factor receptor-2 expression during ramucirumab treatment. BMJ Case Rep. 2019;12:E231464.
  7. Choi YH, Byun HJ, Lee JH, et al. Multiple cherry angiomas and pyogenic granuloma in a patient treated with ramucirumab and paclitaxel. Indian J Dermatol Venereol Leprol. 2020;86:199-202.
  8. Aragaki T, Tomomatsu N, Michi Y, et al. Ramucirumab-related oral pyogenic granuloma: a report of two cases [published online March 8, 2021]. Intern Med. 2021;60:2601-2605. doi:10.2169/internalmedicine.6650-20
  9. Devillers C, Vanhooteghem O, Henrijean A, et al. Subungual pyogenic granuloma secondary to docetaxel therapy. Clin Exp Dermatol. 2009;34:251-252.
  10. Lim YH, Odell ID, Ko CJ, et al. Somatic p.T771R KDR (VEGFR2) mutation arising in a sporadic angioma during ramucirumab therapy. JAMA Dermatol. 2015;151:1240-1243.
Article PDF
Author and Disclosure Information

Dr. Savell is from the Department of Dermatology, University of California Davis. Dr. Heaphy is from the School of Medicine, University of Nevada, Reno, and the Skin Cancer and Dermatology Institute, Reno.

The authors report no conflict of interest.

Correspondence: Anita S. Savell, MD, 3301 C St, Ste 1400, Sacramento, CA 95816 (asavell@ucdavis.edu).

Issue
Cutis - 113(1)
Publications
Topics
Page Number
E5-E7
Sections
Author and Disclosure Information

Dr. Savell is from the Department of Dermatology, University of California Davis. Dr. Heaphy is from the School of Medicine, University of Nevada, Reno, and the Skin Cancer and Dermatology Institute, Reno.

The authors report no conflict of interest.

Correspondence: Anita S. Savell, MD, 3301 C St, Ste 1400, Sacramento, CA 95816 (asavell@ucdavis.edu).

Author and Disclosure Information

Dr. Savell is from the Department of Dermatology, University of California Davis. Dr. Heaphy is from the School of Medicine, University of Nevada, Reno, and the Skin Cancer and Dermatology Institute, Reno.

The authors report no conflict of interest.

Correspondence: Anita S. Savell, MD, 3301 C St, Ste 1400, Sacramento, CA 95816 (asavell@ucdavis.edu).

Article PDF
Article PDF

To the Editor:

Pyogenic granuloma (PG) is a benign vascular tumor that clinically is characterized as a small eruptive friable papule.1 Lesions typically are solitary and most commonly occur in children but also are associated with pregnancy; trauma to the skin or mucosa; and use of certain medications such as isotretinoin, capecitabine, vemurafenib, or indinavir.1 Numerous antineoplastic medications have been associated with the development of solitary PGs, including the taxane mitotic inhibitor paclitaxel (PTX) and the vascular endothelial growth factor receptor 2 (VEGFR2) monoclonal antibody ramucirumab.2 We report a case of multiple PGs in a patient undergoing treatment with PTX and ramucirumab.

New-onset pyogenic granuloma on the left cheek following combination therapy with paclitaxel and ramucirumab.
FIGURE 1. New-onset pyogenic granuloma on the left cheek following combination therapy with paclitaxel and ramucirumab.

A 59-year-old woman presented to the dermatology clinic with red, itchy, bleeding skin lesions on the breast, superior chest, left cheek, and forearm of 1 month’s duration. She denied any preceding trauma to the areas. Her medical history was notable for gastroesophageal junction adenocarcinoma diagnosed more than 2 years prior to presentation. Her original treatment regimen included nivolumab, which was discontinued for unknown reasons 5 months prior to presentation, and she was started on combination therapy with PTX and ramucirumab at that time. She noted the formation of small red papules 2 months after the initiation of PTX-ramucirumab combination therapy, which grew larger over the course of the next month. Physical examination revealed 5 friable hemorrhagic papules and nodules ranging in size from 3 to 10 mm on the chest, cheek, and forearm consistent with PGs (Figure 1). Several scattered cherry angiomas were noted on the scalp and torso, but the patient reported these were not new. Biopsies of the PGs demonstrated lobular aggregates of small-caliber vessels set in an edematous inflamed stroma and partially enclosed by small collarettes of adnexal epithelium, confirming the clinical diagnosis of multiple PGs (Figure 2).

Histopathology from the left cheek, medial breast, and medial superior chest demonstrated lobular aggregates of small-caliber vessels set in an edematous inflamed stroma and partially enclosed by small collarettes of adnexal epithelium
FIGURE 2. A–C, Histopathology from the left cheek, medial breast, and medial superior chest demonstrated lobular aggregates of small-caliber vessels set in an edematous inflamed stroma and partially enclosed by small collarettes of adnexal epithelium, consistent with pyogenic granuloma (H&E; original magnifications ×40, ×100, and ×40, respectively).

The first case of PTX-associated PG was reported in 2012.3 Based on a PubMed search of articles indexed for MEDLINE using the terms pyogenic granuloma, lobular capillary hemangioma, paclitaxel, taxane, and ramucirumab, there have been 9 cases of solitary PG development in the setting of PTX alone or in combination with ramucirumab since 2019 (Table).3-8 Pyogenic granulomas reported in patients who were treated exclusively with PTX were subungual, while the cases resulting from combined therapy were present on the scalp, face, oral mucosa, and surfaces of the hands sparing the nails. Ibe et al6 reported PG in a patient who received ramucirumab therapy without PTX but in combination with another taxane, docetaxel, which itself has been reported to cause subungual PG when used alone.9 Our case of the simultaneous development of multiple PGs in the setting of combined PTX and ramucirumab therapy added to the cutaneous distributions for which therapy-induced PGs have been observed (Table).

Pyogenic Granulomas in Patients Undergoing Treatment With Paclitaxel and Ramucirumab

The development of PG, a vascular tumor, during treatment with the VEGFR2 inhibitor ramucirumab—whose mechanism of action is to inhibit angioneogenesis—is inherently paradoxical. In 2015, a rapidly expanding angioma with a mutation in the kinase domain receptor gene, KDR, that encodes VEGFR2 was identified in a patient undergoing ramucirumab therapy. The authors suggested that KDR mutation resulted in paradoxical activation of VEGFR2 in the setting of ramucirumab therapy.10 Since then, ramucirumab and PTX were suggested to have a synergistic effect in vascular proliferation,5 though an exact mechanism has not been proposed. Other authors have identified increased expression of VEGFR2 in biopsy specimens of PG during combined ramucirumab and taxane therapy.6 Although genetic studies have not been used to evaluate for the presence of KDR mutations specifically in our patient population, it is possible that patients who develop PG and other vascular tumors during combined taxane and ramucirumab therapy have a mutation that makes them more susceptible to VEGFR2 upregulation. UV exposure may have a role in the formation of PG in patients on combined ramucirumab and taxane therapy7; however, our patient’s lesions were distributed on both sun-exposed and unexposed areas. Although potential clinical implications have not yet been thoroughly investigated, following long-term outcomes for these patients may provide important information on the efficacy of the antineoplastic regimen in the subset of patients who develop cutaneous vascular tumors during antiangiogenic treatment.

Combination therapy with PTX and ramucirumab has been associated with the paradoxical development of cutaneous vascular tumors. We report a case of multiple new-onset PGs in a patient undergoing this treatment regimen.

To the Editor:

Pyogenic granuloma (PG) is a benign vascular tumor that clinically is characterized as a small eruptive friable papule.1 Lesions typically are solitary and most commonly occur in children but also are associated with pregnancy; trauma to the skin or mucosa; and use of certain medications such as isotretinoin, capecitabine, vemurafenib, or indinavir.1 Numerous antineoplastic medications have been associated with the development of solitary PGs, including the taxane mitotic inhibitor paclitaxel (PTX) and the vascular endothelial growth factor receptor 2 (VEGFR2) monoclonal antibody ramucirumab.2 We report a case of multiple PGs in a patient undergoing treatment with PTX and ramucirumab.

New-onset pyogenic granuloma on the left cheek following combination therapy with paclitaxel and ramucirumab.
FIGURE 1. New-onset pyogenic granuloma on the left cheek following combination therapy with paclitaxel and ramucirumab.

A 59-year-old woman presented to the dermatology clinic with red, itchy, bleeding skin lesions on the breast, superior chest, left cheek, and forearm of 1 month’s duration. She denied any preceding trauma to the areas. Her medical history was notable for gastroesophageal junction adenocarcinoma diagnosed more than 2 years prior to presentation. Her original treatment regimen included nivolumab, which was discontinued for unknown reasons 5 months prior to presentation, and she was started on combination therapy with PTX and ramucirumab at that time. She noted the formation of small red papules 2 months after the initiation of PTX-ramucirumab combination therapy, which grew larger over the course of the next month. Physical examination revealed 5 friable hemorrhagic papules and nodules ranging in size from 3 to 10 mm on the chest, cheek, and forearm consistent with PGs (Figure 1). Several scattered cherry angiomas were noted on the scalp and torso, but the patient reported these were not new. Biopsies of the PGs demonstrated lobular aggregates of small-caliber vessels set in an edematous inflamed stroma and partially enclosed by small collarettes of adnexal epithelium, confirming the clinical diagnosis of multiple PGs (Figure 2).

Histopathology from the left cheek, medial breast, and medial superior chest demonstrated lobular aggregates of small-caliber vessels set in an edematous inflamed stroma and partially enclosed by small collarettes of adnexal epithelium
FIGURE 2. A–C, Histopathology from the left cheek, medial breast, and medial superior chest demonstrated lobular aggregates of small-caliber vessels set in an edematous inflamed stroma and partially enclosed by small collarettes of adnexal epithelium, consistent with pyogenic granuloma (H&E; original magnifications ×40, ×100, and ×40, respectively).

The first case of PTX-associated PG was reported in 2012.3 Based on a PubMed search of articles indexed for MEDLINE using the terms pyogenic granuloma, lobular capillary hemangioma, paclitaxel, taxane, and ramucirumab, there have been 9 cases of solitary PG development in the setting of PTX alone or in combination with ramucirumab since 2019 (Table).3-8 Pyogenic granulomas reported in patients who were treated exclusively with PTX were subungual, while the cases resulting from combined therapy were present on the scalp, face, oral mucosa, and surfaces of the hands sparing the nails. Ibe et al6 reported PG in a patient who received ramucirumab therapy without PTX but in combination with another taxane, docetaxel, which itself has been reported to cause subungual PG when used alone.9 Our case of the simultaneous development of multiple PGs in the setting of combined PTX and ramucirumab therapy added to the cutaneous distributions for which therapy-induced PGs have been observed (Table).

Pyogenic Granulomas in Patients Undergoing Treatment With Paclitaxel and Ramucirumab

The development of PG, a vascular tumor, during treatment with the VEGFR2 inhibitor ramucirumab—whose mechanism of action is to inhibit angioneogenesis—is inherently paradoxical. In 2015, a rapidly expanding angioma with a mutation in the kinase domain receptor gene, KDR, that encodes VEGFR2 was identified in a patient undergoing ramucirumab therapy. The authors suggested that KDR mutation resulted in paradoxical activation of VEGFR2 in the setting of ramucirumab therapy.10 Since then, ramucirumab and PTX were suggested to have a synergistic effect in vascular proliferation,5 though an exact mechanism has not been proposed. Other authors have identified increased expression of VEGFR2 in biopsy specimens of PG during combined ramucirumab and taxane therapy.6 Although genetic studies have not been used to evaluate for the presence of KDR mutations specifically in our patient population, it is possible that patients who develop PG and other vascular tumors during combined taxane and ramucirumab therapy have a mutation that makes them more susceptible to VEGFR2 upregulation. UV exposure may have a role in the formation of PG in patients on combined ramucirumab and taxane therapy7; however, our patient’s lesions were distributed on both sun-exposed and unexposed areas. Although potential clinical implications have not yet been thoroughly investigated, following long-term outcomes for these patients may provide important information on the efficacy of the antineoplastic regimen in the subset of patients who develop cutaneous vascular tumors during antiangiogenic treatment.

Combination therapy with PTX and ramucirumab has been associated with the paradoxical development of cutaneous vascular tumors. We report a case of multiple new-onset PGs in a patient undergoing this treatment regimen.

References
  1. Elston D, Neuhaus I, James WD, et al. Andrews’ Diseases of the Skin: Clinical Dermatology. 13th ed. Elsevier; 2020.
  2. Pierson JC. Pyogenic granuloma (lobular capillary hemangioma) clinical presentation. Medscape. Updated February 21, 2020. Accessed December 26, 2023. https://emedicine.medscape.com/article/1084701-clinical#showall
  3. Paul LJ, Cohen PR. Paclitaxel-associated subungual pyogenic granuloma: report in a patient with breast cancer receiving paclitaxel and review of drug-induced pyogenic granulomas adjacent to and beneath the nail. J Drugs Dermatol. 2012;11:262-268.
  4. Alessandrini A, Starace M, Cerè G, et al. Management and outcome of taxane-induced nail side effects: experience of 79 patients from a single centre. Skin Appendage Disord. 2019;5:276-282.
  5. Watanabe R, Nakano E, Kawazoe A, et al. Four cases of paradoxical cephalocervical pyogenic granuloma during treatment with paclitaxel and ramucirumab. J Dermatol. 2019;46:E178-E180.
  6. Ibe T, Hamamoto Y, Takabatake M, et al. Development of pyogenic granuloma with strong vascular endothelial growth factor receptor-2 expression during ramucirumab treatment. BMJ Case Rep. 2019;12:E231464.
  7. Choi YH, Byun HJ, Lee JH, et al. Multiple cherry angiomas and pyogenic granuloma in a patient treated with ramucirumab and paclitaxel. Indian J Dermatol Venereol Leprol. 2020;86:199-202.
  8. Aragaki T, Tomomatsu N, Michi Y, et al. Ramucirumab-related oral pyogenic granuloma: a report of two cases [published online March 8, 2021]. Intern Med. 2021;60:2601-2605. doi:10.2169/internalmedicine.6650-20
  9. Devillers C, Vanhooteghem O, Henrijean A, et al. Subungual pyogenic granuloma secondary to docetaxel therapy. Clin Exp Dermatol. 2009;34:251-252.
  10. Lim YH, Odell ID, Ko CJ, et al. Somatic p.T771R KDR (VEGFR2) mutation arising in a sporadic angioma during ramucirumab therapy. JAMA Dermatol. 2015;151:1240-1243.
References
  1. Elston D, Neuhaus I, James WD, et al. Andrews’ Diseases of the Skin: Clinical Dermatology. 13th ed. Elsevier; 2020.
  2. Pierson JC. Pyogenic granuloma (lobular capillary hemangioma) clinical presentation. Medscape. Updated February 21, 2020. Accessed December 26, 2023. https://emedicine.medscape.com/article/1084701-clinical#showall
  3. Paul LJ, Cohen PR. Paclitaxel-associated subungual pyogenic granuloma: report in a patient with breast cancer receiving paclitaxel and review of drug-induced pyogenic granulomas adjacent to and beneath the nail. J Drugs Dermatol. 2012;11:262-268.
  4. Alessandrini A, Starace M, Cerè G, et al. Management and outcome of taxane-induced nail side effects: experience of 79 patients from a single centre. Skin Appendage Disord. 2019;5:276-282.
  5. Watanabe R, Nakano E, Kawazoe A, et al. Four cases of paradoxical cephalocervical pyogenic granuloma during treatment with paclitaxel and ramucirumab. J Dermatol. 2019;46:E178-E180.
  6. Ibe T, Hamamoto Y, Takabatake M, et al. Development of pyogenic granuloma with strong vascular endothelial growth factor receptor-2 expression during ramucirumab treatment. BMJ Case Rep. 2019;12:E231464.
  7. Choi YH, Byun HJ, Lee JH, et al. Multiple cherry angiomas and pyogenic granuloma in a patient treated with ramucirumab and paclitaxel. Indian J Dermatol Venereol Leprol. 2020;86:199-202.
  8. Aragaki T, Tomomatsu N, Michi Y, et al. Ramucirumab-related oral pyogenic granuloma: a report of two cases [published online March 8, 2021]. Intern Med. 2021;60:2601-2605. doi:10.2169/internalmedicine.6650-20
  9. Devillers C, Vanhooteghem O, Henrijean A, et al. Subungual pyogenic granuloma secondary to docetaxel therapy. Clin Exp Dermatol. 2009;34:251-252.
  10. Lim YH, Odell ID, Ko CJ, et al. Somatic p.T771R KDR (VEGFR2) mutation arising in a sporadic angioma during ramucirumab therapy. JAMA Dermatol. 2015;151:1240-1243.
Issue
Cutis - 113(1)
Issue
Cutis - 113(1)
Page Number
E5-E7
Page Number
E5-E7
Publications
Publications
Topics
Article Type
Display Headline
Multiple New-Onset Pyogenic Granulomas During Treatment With Paclitaxel and Ramucirumab
Display Headline
Multiple New-Onset Pyogenic Granulomas During Treatment With Paclitaxel and Ramucirumab
Sections
Inside the Article

Practice Points

  • Pyogenic granulomas (PGs) are benign vascular tumors that clinically are characterized as small, eruptive, friable papules.
  • Ramucirumab is a monoclonal antibody against vascular endothelial growth factor receptor 2.
  • Some patients experience paradoxical formation of vascular tumors such as PGs when treated with combination therapy with ramucirumab and a taxane such as paclitaxel.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Cemiplimab-Associated Eruption of Generalized Eruptive Keratoacanthoma of Grzybowski

Article Type
Changed
Thu, 01/04/2024 - 09:54
Display Headline
Cemiplimab-Associated Eruption of Generalized Eruptive Keratoacanthoma of Grzybowski

To the Editor:

Treatment of cancer, including cutaneous malignancy, has been transformed by the use of immunotherapeutic agents such as immune checkpoint inhibitors (ICIs) that target cytotoxic T lymphocyte-associated antigen 4, programmed cell-death protein 1 (PD-1), or programmed cell-death ligand 1 (PD-L1). However, these drugs are associated with a distinct set of immune-related adverse events (IRAEs). We present a case of generalized eruptive keratoacanthoma of Grzybowski associated with the ICI cemiplimab.

A 94-year-old White woman presented to the dermatology clinic with acute onset of extensive, locally advanced cutaneous squamous cell carcinoma (cSCC) of the upper right posterolateral calf as well as multiple noninvasive cSCCs of the arms and legs. Her medical history was remarkable for widespread actinic keratoses and numerous cSCCs. The patient had no personal or family history of melanoma. Various cSCCs had required treatment with electrodesiccation and curettage, topical or intralesional 5-fluorouracil, and Mohs micrographic surgery. Approximately 1 year prior to presentation, oral acitretin was initiated to help control the cSCC. Given the extent of locally advanced disease, which was considered unresectable, she was referred to oncology but continued to follow up with dermatology. Positron emission tomography was remarkable for hypermetabolic cutaneous thickening in the upper right posterolateral calf with no evidence of visceral disease.

Clinical presentation of well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type, on the arms and legs, respectively, with widespread red, tender, scaly papules and nodules.
FIGURE 1. A and B, Clinical presentation of well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type, on the arms and legs, respectively, with widespread red, tender, scaly papules and nodules.

The patient was started on cemiplimab, an anti-PD-1 monoclonal antibody ICI indicated for the treatment of both metastatic and advanced cSCC. After 4 cycles of intravenous cemiplimab, the patient developed widespread nodules covering the arms and legs (Figure 1) as well as associated tenderness and pruritus. Biopsies of nodules revealed superficially invasive, well-differentiated cSCC consistent with keratoacanthoma. Although a lymphocytic infiltrate was present, no other specific reaction pattern, such as a lichenoid infiltrate, was present (Figure 2).

Well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type.
FIGURE 2. Well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type. Histopathology of a biopsy specimen from the right proximal lateral calf lesion revealed nests of well-differentiated tumor cells with low-grade nuclei and abundant, glassy, eosinophilic cytoplasm, as well as abundant accumulation of keratin (H&E, original magnification ×40).

Positron emission tomography was repeated, demonstrating resolution of the right calf lesion; however, new diffuse cutaneous lesions and inguinal lymph node involvement were present, again without evidence of visceral disease. Given the clinical and histologic findings, a diagnosis of generalized eruptive keratoacanthoma of Grzybowski was made. Cemiplimab was discontinued after the fifth cycle. The patient declined further systemic treatment, instead choosing a regimen of topical steroids and an emollient. 

Immunotherapeutics have transformed cancer therapy, which includes ICIs that target cytotoxic T lymphocyte-associated antigen 4, PD-1, or PD-L1. Increased activity of these checkpoints allows tumor cells to downregulate T-cell activation, thereby evading immune destruction. When PD-1 on T cells binds PD-L1 on tumor cells, T lymphocytes are inhibited from cytotoxic-mediated killing. Therefore, anti-PD-1 ICIs such as cemiplimab permit T-lymphocyte activation and destruction of malignant cells. However, this unique mechanism of immunotherapy is associated with an array of IRAEs, which often manifest in a delayed and prolonged fashion.1 Immune-related adverse events most commonly affect the gastrointestinal tract as well as the endocrine and dermatologic systems.2 Notably, patients with certain tumors who experience these adverse effects might be more likely to have superior overall survival; therefore, IRAEs are sometimes used as an indicator of favorable treatment response.2,3

Dermatologic IRAEs associated with the use of a PD-1 inhibitor include lichenoid reactions, pruritus, morbilliform eruptions, vitiligo, and bullous pemphigoid.4,5 Eruptions of keratoacanthoma rarely have been reported following treatment with the PD-1 inhibitors nivolumab and pembrolizumab.3,6,7 In our patient, we believe the profound and generalized eruptive keratoacanthoma—a well-differentiated cSCC variant—was related to treatment of locally advanced cSCC with cemiplimab. The mechanism underlying the formation of anti-PD-1 eruptive keratoacanthoma is not well understood. In susceptible patients, it is plausible that the inflammatory environment permitted by ICIs paradoxically induces regression of tumors such as locally invasive cSCC and simultaneously promotes formation of keratoacanthoma.

The role of inflammation in the pathogenesis and progression of cSCC is complex and possibly involves contrasting roles of leukocyte subpopulations.8 The increased incidence of cSCC in the immunocompromised population,8 PD-L1 overexpression in cSCC,9,10 and successful treatment of cSCC with PD-1 inhibition10 all suggest that inhibition of specific inflammatory pathways is pivotal in tumor pathogenesis. However, increased inflammation, particularly inflammation driven by T lymphocytes and Langerhans cells, also is believed to play a key role in the formation of cSCCs, including the degeneration of actinic keratosis into cSCC. Moreover, because keratoacanthomas are believed to be a cSCC variant and also are associated with PD-L1 overexpression,9 it is perplexing that PD-1 blockade may result in eruptive keratoacanthoma in some patients while also treating locally advanced cSCC, as seen in our patient. Successful treatment of keratoacanthoma with anti-inflammatory intralesional or topical corticosteroids adds to this complicated picture.3

We hypothesize that the pathogenesis of invasive cSCC and keratoacanthoma shares certain immune-mediated mechanisms but also differs in distinct manners. To understand the relationship between systemic treatment of cSCC and eruptive keratoacanthoma, further research is required.

In addition, the RAS/BRAF/MEK oncogenic pathway may be involved in the development of cSCCs associated with anti-PD-1. It is hypothesized that BRAF and MEK inhibition increases T-cell infiltration and increases PD-L1 expression on tumor cells,11 thus increasing the susceptibility of those cells to PD-1 blockade. Further supporting a relationship between the RAS/BRAF/MEK and PD-1 pathways, BRAF inhibitors are associated with development of SCCs and verrucal keratosis by upregulation of the RAS pathway.12,13 Perhaps a common mechanism underlying these pathways results in their shared association for an increased risk for cSCC upon blockade. More research is needed to fully elucidate the underlying biochemical mechanism of immunotherapy and formation of SCCs, such as keratoacanthoma. 

Treatment of solitary keratoacanthoma often involves surgical excision; however, the sheer number of lesions in eruptive keratoacanthoma presents a larger dilemma. Because oral systemic retinoids have been shown to be most effective for treating eruptive keratoacanthoma, they are considered first-line therapy as monotherapy or in combination with surgical excision.3 Other treatment options include intralesional or topical corticosteroids, cyclosporine, 5-fluorouracil, imiquimod, and cryotherapy.3,6

The development of ICIs has revolutionized the treatment of cutaneous malignancy, yet we have a great deal more to comprehend on the systemic effects of these medications. Although IRAEs may signal a better response to therapy, some of these effects regrettably can be dose limiting. In our patient, cemiplimab was successful in treating locally advanced cSCC, but treatment also resulted in devastating widespread eruptive keratoacanthoma. The mechanism of this kind of eruption has yet to be understood; we hypothesize that it likely involves T lymphocyte–driven inflammation and the interplay of molecular and immune-mediated pathways.

References
  1. Ramos-Casals M, Brahmer JR, Callahan MK, et al. Immune-related adverse events of checkpoint inhibitors. Nat Rev Dis Primers. 2020;6:38. doi:10.1038/s41572-020-0160-6
  2. Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer. 2019;7:306. doi:10.1186/s40425-019-0805-8
  3. Freites-Martinez A, Kwong BY, Rieger KE, et al. Eruptive keratoacanthomas associated with pembrolizumab therapy. JAMA Dermatol. 2017;153:694-697. doi:10.1001/jamadermatol.2017.0989
  4. Shen J, Chang J, Mendenhall M, et al. Diverse cutaneous adverse eruptions caused by anti-programmed cell death-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) immunotherapies: clinicalfeatures and management. Ther Adv Med Oncol. 2018;10:1758834017751634. doi:10.1177/1758834017751634
  5. Bandino JP, Perry DM, Clarke CE, et al. Two cases of anti-programmed cell death 1-associated bullous pemphigoid-like disease and eruptive keratoacanthomas featuring combined histopathology. J Eur Acad Dermatol Venereol. 2017;31:E378-E380. doi:10.1111/jdv.14179
  6. Marsh RL, Kolodney JA, Iyengar S, et al. Formation of eruptive cutaneous squamous cell carcinomas after programmed cell death protein-1 blockade. JAAD Case Rep. 2020;6:390-393. doi:10.1016/j.jdcr.2020.02.024
  7. Antonov NK, Nair KG, Halasz CL. Transient eruptive keratoacanthomas associated with nivolumab. JAAD Case Rep. 2019;5:342-345. doi:10.1016/j.jdcr.2019.01.025
  8. Bottomley MJ, Thomson J, Harwood C, et al. The role of the immune system in cutaneous squamous cell carcinoma. Int J Mol Sci. 2019;20:2009. doi:10.3390/ijms20082009
  9. Gambichler T, Gnielka M, Rüddel I, et al. Expression of PD-L1 in keratoacanthoma and different stages of progression in cutaneous squamous cell carcinoma. Cancer Immunol Immunother. 2017;66:1199-1204. doi:10.1007/s00262-017-2015-x
  10. Patel R, Chang ALS. Immune checkpoint inhibitors for treating advanced cutaneous squamous cell carcinoma. Am J Clin Dermatol. 2019;20:477-482. doi:10.1007/s40257-019-00426-w
  11. Rozeman EA, Blank CU. Combining checkpoint inhibition and targeted therapy in melanoma. Nat Med. 2019;25:879-882. doi:10.1038/s41591-019-0482-7
  12. Dubauskas Z, Kunishige J, Prieto VG, Jonasch E, Hwu P, Tannir NM. Cutaneous squamous cell carcinoma and inflammation of actinic keratoses associated with sorafenib. Clin Genitourin Cancer. 2009;7:20-23. doi:10.3816/CGC.2009.n.003
  13. Chen P, Chen F, Zhou B. Systematic review and meta-analysis of prevalence of dermatological toxicities associated with vemurafenib treatment in patients with melanoma. Clin Exp Dermatol. 2019;44:243-251. doi:10.1111/ced.13751
Article PDF
Author and Disclosure Information

Drs. Kang, Khanna, M.H. Patel, and N.B. Patel are from Creighton University School of Medicine, Phoenix Regional Campus, Arizona.Drs. Glembocki and N.B. Patel are from Southwest Skin Specialists, Phoenix. Drs. Jeffy and Thosani are from Spectrum Dermatology, Phoenix.

The authors report no conflict of interest.

Correspondence: Bianca Y. Kang, MD, Creighton University School of Medicine, Phoenix Regional Campus, 350 W Thomas Rd, Phoenix, AZ 85013 (biancakang94@gmail.com).

Issue
Cutis - 113(1)
Publications
Topics
Page Number
E8-E10
Sections
Author and Disclosure Information

Drs. Kang, Khanna, M.H. Patel, and N.B. Patel are from Creighton University School of Medicine, Phoenix Regional Campus, Arizona.Drs. Glembocki and N.B. Patel are from Southwest Skin Specialists, Phoenix. Drs. Jeffy and Thosani are from Spectrum Dermatology, Phoenix.

The authors report no conflict of interest.

Correspondence: Bianca Y. Kang, MD, Creighton University School of Medicine, Phoenix Regional Campus, 350 W Thomas Rd, Phoenix, AZ 85013 (biancakang94@gmail.com).

Author and Disclosure Information

Drs. Kang, Khanna, M.H. Patel, and N.B. Patel are from Creighton University School of Medicine, Phoenix Regional Campus, Arizona.Drs. Glembocki and N.B. Patel are from Southwest Skin Specialists, Phoenix. Drs. Jeffy and Thosani are from Spectrum Dermatology, Phoenix.

The authors report no conflict of interest.

Correspondence: Bianca Y. Kang, MD, Creighton University School of Medicine, Phoenix Regional Campus, 350 W Thomas Rd, Phoenix, AZ 85013 (biancakang94@gmail.com).

Article PDF
Article PDF

To the Editor:

Treatment of cancer, including cutaneous malignancy, has been transformed by the use of immunotherapeutic agents such as immune checkpoint inhibitors (ICIs) that target cytotoxic T lymphocyte-associated antigen 4, programmed cell-death protein 1 (PD-1), or programmed cell-death ligand 1 (PD-L1). However, these drugs are associated with a distinct set of immune-related adverse events (IRAEs). We present a case of generalized eruptive keratoacanthoma of Grzybowski associated with the ICI cemiplimab.

A 94-year-old White woman presented to the dermatology clinic with acute onset of extensive, locally advanced cutaneous squamous cell carcinoma (cSCC) of the upper right posterolateral calf as well as multiple noninvasive cSCCs of the arms and legs. Her medical history was remarkable for widespread actinic keratoses and numerous cSCCs. The patient had no personal or family history of melanoma. Various cSCCs had required treatment with electrodesiccation and curettage, topical or intralesional 5-fluorouracil, and Mohs micrographic surgery. Approximately 1 year prior to presentation, oral acitretin was initiated to help control the cSCC. Given the extent of locally advanced disease, which was considered unresectable, she was referred to oncology but continued to follow up with dermatology. Positron emission tomography was remarkable for hypermetabolic cutaneous thickening in the upper right posterolateral calf with no evidence of visceral disease.

Clinical presentation of well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type, on the arms and legs, respectively, with widespread red, tender, scaly papules and nodules.
FIGURE 1. A and B, Clinical presentation of well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type, on the arms and legs, respectively, with widespread red, tender, scaly papules and nodules.

The patient was started on cemiplimab, an anti-PD-1 monoclonal antibody ICI indicated for the treatment of both metastatic and advanced cSCC. After 4 cycles of intravenous cemiplimab, the patient developed widespread nodules covering the arms and legs (Figure 1) as well as associated tenderness and pruritus. Biopsies of nodules revealed superficially invasive, well-differentiated cSCC consistent with keratoacanthoma. Although a lymphocytic infiltrate was present, no other specific reaction pattern, such as a lichenoid infiltrate, was present (Figure 2).

Well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type.
FIGURE 2. Well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type. Histopathology of a biopsy specimen from the right proximal lateral calf lesion revealed nests of well-differentiated tumor cells with low-grade nuclei and abundant, glassy, eosinophilic cytoplasm, as well as abundant accumulation of keratin (H&E, original magnification ×40).

Positron emission tomography was repeated, demonstrating resolution of the right calf lesion; however, new diffuse cutaneous lesions and inguinal lymph node involvement were present, again without evidence of visceral disease. Given the clinical and histologic findings, a diagnosis of generalized eruptive keratoacanthoma of Grzybowski was made. Cemiplimab was discontinued after the fifth cycle. The patient declined further systemic treatment, instead choosing a regimen of topical steroids and an emollient. 

Immunotherapeutics have transformed cancer therapy, which includes ICIs that target cytotoxic T lymphocyte-associated antigen 4, PD-1, or PD-L1. Increased activity of these checkpoints allows tumor cells to downregulate T-cell activation, thereby evading immune destruction. When PD-1 on T cells binds PD-L1 on tumor cells, T lymphocytes are inhibited from cytotoxic-mediated killing. Therefore, anti-PD-1 ICIs such as cemiplimab permit T-lymphocyte activation and destruction of malignant cells. However, this unique mechanism of immunotherapy is associated with an array of IRAEs, which often manifest in a delayed and prolonged fashion.1 Immune-related adverse events most commonly affect the gastrointestinal tract as well as the endocrine and dermatologic systems.2 Notably, patients with certain tumors who experience these adverse effects might be more likely to have superior overall survival; therefore, IRAEs are sometimes used as an indicator of favorable treatment response.2,3

Dermatologic IRAEs associated with the use of a PD-1 inhibitor include lichenoid reactions, pruritus, morbilliform eruptions, vitiligo, and bullous pemphigoid.4,5 Eruptions of keratoacanthoma rarely have been reported following treatment with the PD-1 inhibitors nivolumab and pembrolizumab.3,6,7 In our patient, we believe the profound and generalized eruptive keratoacanthoma—a well-differentiated cSCC variant—was related to treatment of locally advanced cSCC with cemiplimab. The mechanism underlying the formation of anti-PD-1 eruptive keratoacanthoma is not well understood. In susceptible patients, it is plausible that the inflammatory environment permitted by ICIs paradoxically induces regression of tumors such as locally invasive cSCC and simultaneously promotes formation of keratoacanthoma.

The role of inflammation in the pathogenesis and progression of cSCC is complex and possibly involves contrasting roles of leukocyte subpopulations.8 The increased incidence of cSCC in the immunocompromised population,8 PD-L1 overexpression in cSCC,9,10 and successful treatment of cSCC with PD-1 inhibition10 all suggest that inhibition of specific inflammatory pathways is pivotal in tumor pathogenesis. However, increased inflammation, particularly inflammation driven by T lymphocytes and Langerhans cells, also is believed to play a key role in the formation of cSCCs, including the degeneration of actinic keratosis into cSCC. Moreover, because keratoacanthomas are believed to be a cSCC variant and also are associated with PD-L1 overexpression,9 it is perplexing that PD-1 blockade may result in eruptive keratoacanthoma in some patients while also treating locally advanced cSCC, as seen in our patient. Successful treatment of keratoacanthoma with anti-inflammatory intralesional or topical corticosteroids adds to this complicated picture.3

We hypothesize that the pathogenesis of invasive cSCC and keratoacanthoma shares certain immune-mediated mechanisms but also differs in distinct manners. To understand the relationship between systemic treatment of cSCC and eruptive keratoacanthoma, further research is required.

In addition, the RAS/BRAF/MEK oncogenic pathway may be involved in the development of cSCCs associated with anti-PD-1. It is hypothesized that BRAF and MEK inhibition increases T-cell infiltration and increases PD-L1 expression on tumor cells,11 thus increasing the susceptibility of those cells to PD-1 blockade. Further supporting a relationship between the RAS/BRAF/MEK and PD-1 pathways, BRAF inhibitors are associated with development of SCCs and verrucal keratosis by upregulation of the RAS pathway.12,13 Perhaps a common mechanism underlying these pathways results in their shared association for an increased risk for cSCC upon blockade. More research is needed to fully elucidate the underlying biochemical mechanism of immunotherapy and formation of SCCs, such as keratoacanthoma. 

Treatment of solitary keratoacanthoma often involves surgical excision; however, the sheer number of lesions in eruptive keratoacanthoma presents a larger dilemma. Because oral systemic retinoids have been shown to be most effective for treating eruptive keratoacanthoma, they are considered first-line therapy as monotherapy or in combination with surgical excision.3 Other treatment options include intralesional or topical corticosteroids, cyclosporine, 5-fluorouracil, imiquimod, and cryotherapy.3,6

The development of ICIs has revolutionized the treatment of cutaneous malignancy, yet we have a great deal more to comprehend on the systemic effects of these medications. Although IRAEs may signal a better response to therapy, some of these effects regrettably can be dose limiting. In our patient, cemiplimab was successful in treating locally advanced cSCC, but treatment also resulted in devastating widespread eruptive keratoacanthoma. The mechanism of this kind of eruption has yet to be understood; we hypothesize that it likely involves T lymphocyte–driven inflammation and the interplay of molecular and immune-mediated pathways.

To the Editor:

Treatment of cancer, including cutaneous malignancy, has been transformed by the use of immunotherapeutic agents such as immune checkpoint inhibitors (ICIs) that target cytotoxic T lymphocyte-associated antigen 4, programmed cell-death protein 1 (PD-1), or programmed cell-death ligand 1 (PD-L1). However, these drugs are associated with a distinct set of immune-related adverse events (IRAEs). We present a case of generalized eruptive keratoacanthoma of Grzybowski associated with the ICI cemiplimab.

A 94-year-old White woman presented to the dermatology clinic with acute onset of extensive, locally advanced cutaneous squamous cell carcinoma (cSCC) of the upper right posterolateral calf as well as multiple noninvasive cSCCs of the arms and legs. Her medical history was remarkable for widespread actinic keratoses and numerous cSCCs. The patient had no personal or family history of melanoma. Various cSCCs had required treatment with electrodesiccation and curettage, topical or intralesional 5-fluorouracil, and Mohs micrographic surgery. Approximately 1 year prior to presentation, oral acitretin was initiated to help control the cSCC. Given the extent of locally advanced disease, which was considered unresectable, she was referred to oncology but continued to follow up with dermatology. Positron emission tomography was remarkable for hypermetabolic cutaneous thickening in the upper right posterolateral calf with no evidence of visceral disease.

Clinical presentation of well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type, on the arms and legs, respectively, with widespread red, tender, scaly papules and nodules.
FIGURE 1. A and B, Clinical presentation of well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type, on the arms and legs, respectively, with widespread red, tender, scaly papules and nodules.

The patient was started on cemiplimab, an anti-PD-1 monoclonal antibody ICI indicated for the treatment of both metastatic and advanced cSCC. After 4 cycles of intravenous cemiplimab, the patient developed widespread nodules covering the arms and legs (Figure 1) as well as associated tenderness and pruritus. Biopsies of nodules revealed superficially invasive, well-differentiated cSCC consistent with keratoacanthoma. Although a lymphocytic infiltrate was present, no other specific reaction pattern, such as a lichenoid infiltrate, was present (Figure 2).

Well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type.
FIGURE 2. Well-differentiated cutaneous squamous cell carcinoma, keratoacanthoma type. Histopathology of a biopsy specimen from the right proximal lateral calf lesion revealed nests of well-differentiated tumor cells with low-grade nuclei and abundant, glassy, eosinophilic cytoplasm, as well as abundant accumulation of keratin (H&E, original magnification ×40).

Positron emission tomography was repeated, demonstrating resolution of the right calf lesion; however, new diffuse cutaneous lesions and inguinal lymph node involvement were present, again without evidence of visceral disease. Given the clinical and histologic findings, a diagnosis of generalized eruptive keratoacanthoma of Grzybowski was made. Cemiplimab was discontinued after the fifth cycle. The patient declined further systemic treatment, instead choosing a regimen of topical steroids and an emollient. 

Immunotherapeutics have transformed cancer therapy, which includes ICIs that target cytotoxic T lymphocyte-associated antigen 4, PD-1, or PD-L1. Increased activity of these checkpoints allows tumor cells to downregulate T-cell activation, thereby evading immune destruction. When PD-1 on T cells binds PD-L1 on tumor cells, T lymphocytes are inhibited from cytotoxic-mediated killing. Therefore, anti-PD-1 ICIs such as cemiplimab permit T-lymphocyte activation and destruction of malignant cells. However, this unique mechanism of immunotherapy is associated with an array of IRAEs, which often manifest in a delayed and prolonged fashion.1 Immune-related adverse events most commonly affect the gastrointestinal tract as well as the endocrine and dermatologic systems.2 Notably, patients with certain tumors who experience these adverse effects might be more likely to have superior overall survival; therefore, IRAEs are sometimes used as an indicator of favorable treatment response.2,3

Dermatologic IRAEs associated with the use of a PD-1 inhibitor include lichenoid reactions, pruritus, morbilliform eruptions, vitiligo, and bullous pemphigoid.4,5 Eruptions of keratoacanthoma rarely have been reported following treatment with the PD-1 inhibitors nivolumab and pembrolizumab.3,6,7 In our patient, we believe the profound and generalized eruptive keratoacanthoma—a well-differentiated cSCC variant—was related to treatment of locally advanced cSCC with cemiplimab. The mechanism underlying the formation of anti-PD-1 eruptive keratoacanthoma is not well understood. In susceptible patients, it is plausible that the inflammatory environment permitted by ICIs paradoxically induces regression of tumors such as locally invasive cSCC and simultaneously promotes formation of keratoacanthoma.

The role of inflammation in the pathogenesis and progression of cSCC is complex and possibly involves contrasting roles of leukocyte subpopulations.8 The increased incidence of cSCC in the immunocompromised population,8 PD-L1 overexpression in cSCC,9,10 and successful treatment of cSCC with PD-1 inhibition10 all suggest that inhibition of specific inflammatory pathways is pivotal in tumor pathogenesis. However, increased inflammation, particularly inflammation driven by T lymphocytes and Langerhans cells, also is believed to play a key role in the formation of cSCCs, including the degeneration of actinic keratosis into cSCC. Moreover, because keratoacanthomas are believed to be a cSCC variant and also are associated with PD-L1 overexpression,9 it is perplexing that PD-1 blockade may result in eruptive keratoacanthoma in some patients while also treating locally advanced cSCC, as seen in our patient. Successful treatment of keratoacanthoma with anti-inflammatory intralesional or topical corticosteroids adds to this complicated picture.3

We hypothesize that the pathogenesis of invasive cSCC and keratoacanthoma shares certain immune-mediated mechanisms but also differs in distinct manners. To understand the relationship between systemic treatment of cSCC and eruptive keratoacanthoma, further research is required.

In addition, the RAS/BRAF/MEK oncogenic pathway may be involved in the development of cSCCs associated with anti-PD-1. It is hypothesized that BRAF and MEK inhibition increases T-cell infiltration and increases PD-L1 expression on tumor cells,11 thus increasing the susceptibility of those cells to PD-1 blockade. Further supporting a relationship between the RAS/BRAF/MEK and PD-1 pathways, BRAF inhibitors are associated with development of SCCs and verrucal keratosis by upregulation of the RAS pathway.12,13 Perhaps a common mechanism underlying these pathways results in their shared association for an increased risk for cSCC upon blockade. More research is needed to fully elucidate the underlying biochemical mechanism of immunotherapy and formation of SCCs, such as keratoacanthoma. 

Treatment of solitary keratoacanthoma often involves surgical excision; however, the sheer number of lesions in eruptive keratoacanthoma presents a larger dilemma. Because oral systemic retinoids have been shown to be most effective for treating eruptive keratoacanthoma, they are considered first-line therapy as monotherapy or in combination with surgical excision.3 Other treatment options include intralesional or topical corticosteroids, cyclosporine, 5-fluorouracil, imiquimod, and cryotherapy.3,6

The development of ICIs has revolutionized the treatment of cutaneous malignancy, yet we have a great deal more to comprehend on the systemic effects of these medications. Although IRAEs may signal a better response to therapy, some of these effects regrettably can be dose limiting. In our patient, cemiplimab was successful in treating locally advanced cSCC, but treatment also resulted in devastating widespread eruptive keratoacanthoma. The mechanism of this kind of eruption has yet to be understood; we hypothesize that it likely involves T lymphocyte–driven inflammation and the interplay of molecular and immune-mediated pathways.

References
  1. Ramos-Casals M, Brahmer JR, Callahan MK, et al. Immune-related adverse events of checkpoint inhibitors. Nat Rev Dis Primers. 2020;6:38. doi:10.1038/s41572-020-0160-6
  2. Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer. 2019;7:306. doi:10.1186/s40425-019-0805-8
  3. Freites-Martinez A, Kwong BY, Rieger KE, et al. Eruptive keratoacanthomas associated with pembrolizumab therapy. JAMA Dermatol. 2017;153:694-697. doi:10.1001/jamadermatol.2017.0989
  4. Shen J, Chang J, Mendenhall M, et al. Diverse cutaneous adverse eruptions caused by anti-programmed cell death-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) immunotherapies: clinicalfeatures and management. Ther Adv Med Oncol. 2018;10:1758834017751634. doi:10.1177/1758834017751634
  5. Bandino JP, Perry DM, Clarke CE, et al. Two cases of anti-programmed cell death 1-associated bullous pemphigoid-like disease and eruptive keratoacanthomas featuring combined histopathology. J Eur Acad Dermatol Venereol. 2017;31:E378-E380. doi:10.1111/jdv.14179
  6. Marsh RL, Kolodney JA, Iyengar S, et al. Formation of eruptive cutaneous squamous cell carcinomas after programmed cell death protein-1 blockade. JAAD Case Rep. 2020;6:390-393. doi:10.1016/j.jdcr.2020.02.024
  7. Antonov NK, Nair KG, Halasz CL. Transient eruptive keratoacanthomas associated with nivolumab. JAAD Case Rep. 2019;5:342-345. doi:10.1016/j.jdcr.2019.01.025
  8. Bottomley MJ, Thomson J, Harwood C, et al. The role of the immune system in cutaneous squamous cell carcinoma. Int J Mol Sci. 2019;20:2009. doi:10.3390/ijms20082009
  9. Gambichler T, Gnielka M, Rüddel I, et al. Expression of PD-L1 in keratoacanthoma and different stages of progression in cutaneous squamous cell carcinoma. Cancer Immunol Immunother. 2017;66:1199-1204. doi:10.1007/s00262-017-2015-x
  10. Patel R, Chang ALS. Immune checkpoint inhibitors for treating advanced cutaneous squamous cell carcinoma. Am J Clin Dermatol. 2019;20:477-482. doi:10.1007/s40257-019-00426-w
  11. Rozeman EA, Blank CU. Combining checkpoint inhibition and targeted therapy in melanoma. Nat Med. 2019;25:879-882. doi:10.1038/s41591-019-0482-7
  12. Dubauskas Z, Kunishige J, Prieto VG, Jonasch E, Hwu P, Tannir NM. Cutaneous squamous cell carcinoma and inflammation of actinic keratoses associated with sorafenib. Clin Genitourin Cancer. 2009;7:20-23. doi:10.3816/CGC.2009.n.003
  13. Chen P, Chen F, Zhou B. Systematic review and meta-analysis of prevalence of dermatological toxicities associated with vemurafenib treatment in patients with melanoma. Clin Exp Dermatol. 2019;44:243-251. doi:10.1111/ced.13751
References
  1. Ramos-Casals M, Brahmer JR, Callahan MK, et al. Immune-related adverse events of checkpoint inhibitors. Nat Rev Dis Primers. 2020;6:38. doi:10.1038/s41572-020-0160-6
  2. Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer. 2019;7:306. doi:10.1186/s40425-019-0805-8
  3. Freites-Martinez A, Kwong BY, Rieger KE, et al. Eruptive keratoacanthomas associated with pembrolizumab therapy. JAMA Dermatol. 2017;153:694-697. doi:10.1001/jamadermatol.2017.0989
  4. Shen J, Chang J, Mendenhall M, et al. Diverse cutaneous adverse eruptions caused by anti-programmed cell death-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) immunotherapies: clinicalfeatures and management. Ther Adv Med Oncol. 2018;10:1758834017751634. doi:10.1177/1758834017751634
  5. Bandino JP, Perry DM, Clarke CE, et al. Two cases of anti-programmed cell death 1-associated bullous pemphigoid-like disease and eruptive keratoacanthomas featuring combined histopathology. J Eur Acad Dermatol Venereol. 2017;31:E378-E380. doi:10.1111/jdv.14179
  6. Marsh RL, Kolodney JA, Iyengar S, et al. Formation of eruptive cutaneous squamous cell carcinomas after programmed cell death protein-1 blockade. JAAD Case Rep. 2020;6:390-393. doi:10.1016/j.jdcr.2020.02.024
  7. Antonov NK, Nair KG, Halasz CL. Transient eruptive keratoacanthomas associated with nivolumab. JAAD Case Rep. 2019;5:342-345. doi:10.1016/j.jdcr.2019.01.025
  8. Bottomley MJ, Thomson J, Harwood C, et al. The role of the immune system in cutaneous squamous cell carcinoma. Int J Mol Sci. 2019;20:2009. doi:10.3390/ijms20082009
  9. Gambichler T, Gnielka M, Rüddel I, et al. Expression of PD-L1 in keratoacanthoma and different stages of progression in cutaneous squamous cell carcinoma. Cancer Immunol Immunother. 2017;66:1199-1204. doi:10.1007/s00262-017-2015-x
  10. Patel R, Chang ALS. Immune checkpoint inhibitors for treating advanced cutaneous squamous cell carcinoma. Am J Clin Dermatol. 2019;20:477-482. doi:10.1007/s40257-019-00426-w
  11. Rozeman EA, Blank CU. Combining checkpoint inhibition and targeted therapy in melanoma. Nat Med. 2019;25:879-882. doi:10.1038/s41591-019-0482-7
  12. Dubauskas Z, Kunishige J, Prieto VG, Jonasch E, Hwu P, Tannir NM. Cutaneous squamous cell carcinoma and inflammation of actinic keratoses associated with sorafenib. Clin Genitourin Cancer. 2009;7:20-23. doi:10.3816/CGC.2009.n.003
  13. Chen P, Chen F, Zhou B. Systematic review and meta-analysis of prevalence of dermatological toxicities associated with vemurafenib treatment in patients with melanoma. Clin Exp Dermatol. 2019;44:243-251. doi:10.1111/ced.13751
Issue
Cutis - 113(1)
Issue
Cutis - 113(1)
Page Number
E8-E10
Page Number
E8-E10
Publications
Publications
Topics
Article Type
Display Headline
Cemiplimab-Associated Eruption of Generalized Eruptive Keratoacanthoma of Grzybowski
Display Headline
Cemiplimab-Associated Eruption of Generalized Eruptive Keratoacanthoma of Grzybowski
Sections
Inside the Article

Practice Points

  • Immunotherapy, including immune checkpoint inhibitors such as programmed cell-death protein 1 (PD-1) inhibitors, is associated with an array of immune-related adverse events that often manifest in a delayed and prolonged manner. They most commonly affect the gastrointestinal tract as well as the endocrine and dermatologic systems.
  • Dermatologic adverse effects associated with PD-1 inhibitors include lichenoid reactions, pruritus, morbilliform eruptions, vitiligo, and bullous pemphigoid.
  • Eruptions of keratoacanthoma rarely have been reported following treatment with PD-1 inhibitors such as cemiplimab, nivolumab, and pembrolizumab.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Thalidomide Analogue Drug Eruption Along the Lines of Blaschko

Article Type
Changed
Thu, 12/21/2023 - 10:31
Display Headline
Thalidomide Analogue Drug Eruption Along the Lines of Blaschko

To the Editor:

Lenalidomide is a thalidomide analogue used to treat various hematologic malignancies, including non-Hodgkin lymphoma, myelodysplastic syndrome, and multiple myeloma (MM).1 Lenalidomide is referred to as a degrader therapeutic because it induces targeted protein degradation of disease-relevant proteins (eg, Ikaros family zinc finger protein 1 [IKZF1], Ikaros family zinc finger protein 3 [IKZF3], and casein kinase I isoform-α [CK1α]) as its primary mechanism of action.1,2 Although cutaneous adverse events are relatively common among thalidomide analogues, the morphologic and histopathologic descriptions of these drug eruptions have not been fully elucidated.3,4 We report a novel pityriasiform drug eruption followed by a clinical eruption suggestive of blaschkitis in a patient with MM who was being treated with lenalidomide.

A 76-year-old man presented to the dermatology clinic with a progressive, mildly pruritic eruption on the chest and axillae of 1 year’s duration. He had a medical history of chronic hepatitis B, malignant carcinoid tumor of the colon, prostate cancer, and MM. The eruption emerged 1 to 2 weeks after the patient started oral lenalidomide 10 mg/d and oral dexamethasone40 mg/wk following autologous stem cell transplantation for MM. The patient had not received any other therapy for MM.

Physical examination revealed multiple erythematous, hyperpigmented, scaly papules and plaques on the lateral chest and within the axillae (Figure 1). A skin biopsy from the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate with scattered eosinophils, neutrophils, and extravasated erythrocytes. The overlying epidermis showed spongiosis with parakeratosis in addition to lymphocytic exocytosis (Figure 2). No fungal organisms were highlighted on periodic acid–Schiff staining. After this evaluation, we recommended that the patient discontinue lenalidomide and start taking a topical over-the-counter corticosteroid for 2 weeks. Over time, he noted marked improvement in the eruption and associated pruritus.

Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.
FIGURE 1. Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.

After a drug holiday of 2 months, the patient resumed a maintenance dosage of oral lenalidomide 10 mg/d. Four or 5 days after restarting lenalidomide, a pruritic eruption appeared that involved the axillae and the left lower abdomen, circling around to the left lower back. The axillary eruption resolved with a topical over-the-counter corticosteroid; the abdominal eruption persisted.

A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes.
FIGURE 2. A and B, A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes. The overlying epidermis was spongiotic with parakeratosis and lymphocytic exocytosis (H&E, original magnifications ×100 and ×200).

At the 3-month follow-up visit, physical examination revealed erythematous macules and papules that coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline (Figure 3).

At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossi
FIGURE 3. A and B, At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline.

We recommended that the patient continue treatment through this eruption; he was instructed to apply a corticosteroid cream and resume lenalidomide at the maintenance dosage. A month later, he reported that the eruption and associated pruritus resolved with the corticosteroid cream and resumption of the maintenance dose of lenalidomide. The patient noted no further spread of the eruption.

Cutaneous adverse events are common following lenalidomide. In prior trials, the overall incidence of any-grade rash following lenalidomide exposure was 22% to 33%.5 A meta-analysis of 10 trials determined the overall incidence of all-grade and high-grade cutaneous adverse events after exposure to lenalidomide was 27.2% and 3.6%, respectively.6 Our case represents a pityriasiform eruption due to lenalidomide followed by a secondary eruption suggestive of blaschkitis.

 

 

The rash due to lenalidomide has been described as morbilliform, urticarial, dermatitic, acneform, and undefined.7 Lenalidomide-induced rash typically develops during the first month of therapy, similar to our patient’s presentation. It has even been observed in the first week of therapy.8 Severe reactions such as Stevens-Johnson syndrome and toxic epidermal necrolysis have been reported.5,6 Risk factors associated with rash secondary to lenalidomide include advanced age (≥70 years), presence of Bence-Jones protein-type MM in urine, and no prior chemotherapy.8 Our patient had 2 of these risk factors: advanced age and no prior chemotherapy for MM. The exact pathogenesis by which lenalidomide leads to a pityriasiform eruption, as in our patient, or to a rash in general is unclear. Studies have hypothesized that a lenalidomide-induced rash could be attributable to a delayed hypersensitivity type IV reaction or to a reaction related to the molecular mechanism of action of the drug.9

At the molecular level, the antimyeloma effects of lenalidomide include promoting degradation of transcription factors IKZF1 and IKZF3, which subsequently increases production of IL-2.1,2,9 Recombinant IL-2 has been associated with an increased incidence of rash in other cancers.9 Overexpression of programmed death 1(PD-1) and its ligand (PD-L1) has been demonstrated in MM; lenalidomide has been shown to downregulate both PD-1 and PD-L1. Patients receiving PD-1 and PD-L1 inhibitors commonly have developed rash.9 However, the association between lenalidomide and its downregulation of PD-1 and PD-L1 leading to rash has not been fully elucidated. Given the multiple malignancies in our patient—MM, prostate cancer, malignant carcinoid tumor—an underlying paraneoplastic phenomenon may be possible. Additionally, because our patient initially received dexamethasone along with lenalidomide, the manifestation of the initial pityriasiform rash may have been less severe due to the steroid use. Although our patient underwent a 2-month drug holiday following the initial pityriasiform eruption, most lenalidomide-induced rashes do not necessitate discontinuation of the drug.5,7

Our patient’s secondary drug eruption was clinically suggestive of lenalidomide-induced blaschkitis. A report of a German patient with plasmacytoma described a unilateral papular exanthem that developed 4 months after lenalidomide was initiated.10 The papular exanthem following the lines of Blaschko lines extended from that patient’s posterior left foot to the calf and on to the thigh and flank,10 which was more extensive than our patient’s eruption. Blaschkitis in this patient resolved with a corticosteroid cream and UV light therapy10; lenalidomide was not discontinued, similar to our patient.

The pathogenesis of our patient’s secondary eruption that preferentially involved the lines of Blaschko is unclear. After the initial pityriasiform eruption, the secondary eruption was blaschkitis. Distinguishing dermatomes from the lines of Blaschko, which are thought to represent pathways of epidermal cell migration and proliferation during embryologic development, is important. Genodermatoses such as incontinentia pigmenti and hypomelanosis of Ito involve the lines of Blaschko11; other disorders in the differential diagnosis of linear configurations include linear lichen planus, linear cutaneous lupus erythematosus, linear morphea, and lichen striatus.11 Notably, drug-induced blaschkitis is rare.

Cutaneous adverse reactions from thalidomide analogues are relatively common. Our case of lenalidomide-associated blaschkitis that developed following an initial pityriasiform drug eruption in a patient with MM highlights that dermatologists need to collaborate with the oncologist regarding the severity of drug eruptions to determine if the patient should continue treatment through the cutaneous eruptions or discontinue a vital medication.

References
  1. Jan M, Sperling AS, Ebert BL. Cancer therapies based on targeted protein degradation—lessons learned with lenalidomide. Nat Rev Clin Oncol. 2021;18:401-417. doi:10.1038/s41571-021-00479-z
  2. Shah UA, Mailankody S. Emerging immunotherapies in multiple myeloma. BMJ. 2020;370:3176. doi:10.1136/BMJ.M3176
  3. Richardson PG, Blood E, Mitsiades CS, et al. A randomized phase 2 study of lenalidomide therapy for patients with relapsed or relapsed and refractory multiple myeloma. Blood. 2006;108:3458-3464. doi:10.1182/BLOOD-2006-04-015909
  4. Benboubker L, Dimopoulos MA, Dispenzieri A, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med. 2014;371:906-917. doi:10.1056/NEJMOA1402551
  5. Tinsley SM, Kurtin SE, Ridgeway JA. Practical management of lenalidomide-related rash. Clin Lymphoma Myeloma Leuk. 2015;15(suppl):S64-S69. doi:10.1016/J.CLML.2015.02.008
  6. Nardone B, Wu S, Garden BC, et al. Risk of rash associated with lenalidomide in cancer patients: a systematic review of the literature and meta-analysis. Clin Lymphoma Myeloma Leuk. 2013;13:424-429. doi:10.1016/J.CLML.2013.03.006
  7. Sviggum HP, Davis MDP, Rajkumar SV, et al. Dermatologic adverse effects of lenalidomide therapy for amyloidosis and multiple myeloma. Arch Dermatol. 2006;142:1298-1302. doi:10.1001/ARCHDERM.142.10.1298
  8. Sugi T, Nishigami Y, Saigo H, et al. Analysis of risk factors for lenalidomide-associated skin rash in patients with multiple myeloma. Leuk Lymphoma. 2021;62:1405-1410. doi:10.1080/10428194.2021.1876867
  9. Barley K, He W, Agarwal S, et al. Outcomes and management of lenalidomide-associated rash in patients with multiple myeloma. Leuk Lymphoma. 2016;57:2510-2515. doi:10.3109/10428194.2016.1151507
  10. Grape J, Frosch P. Papular drug eruption along the lines of Blaschko caused by lenalidomide [in German]. Hautarzt. 2011;62:618-620. doi:10.1007/S00105-010-2121-6
  11. Bolognia JL, Orlow SJ, Glick SA. Lines of Blaschko. J Am Acad Dermatol. 1994;31(2 pt 1):157-190. doi:10.1016/S0190-9622(94)70143-1
Article PDF
Author and Disclosure Information

From the Tulane University School of Medicine, New Orleans, Louisiana. Drs. Jafari and Williams are from the Department of Dermatology, and Dr. Vick is from the Department of Dermatopathology.

The authors report no conflict of interest.

Correspondence: Alexander J. Jafari, MD, MPH, Department of Dermatology, Tulane University School of Medicine, 1430 Tulane Ave, #8036, New Orleans, LA 70112 (ajafari@tulane.edu).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E27-E29
Sections
Author and Disclosure Information

From the Tulane University School of Medicine, New Orleans, Louisiana. Drs. Jafari and Williams are from the Department of Dermatology, and Dr. Vick is from the Department of Dermatopathology.

The authors report no conflict of interest.

Correspondence: Alexander J. Jafari, MD, MPH, Department of Dermatology, Tulane University School of Medicine, 1430 Tulane Ave, #8036, New Orleans, LA 70112 (ajafari@tulane.edu).

Author and Disclosure Information

From the Tulane University School of Medicine, New Orleans, Louisiana. Drs. Jafari and Williams are from the Department of Dermatology, and Dr. Vick is from the Department of Dermatopathology.

The authors report no conflict of interest.

Correspondence: Alexander J. Jafari, MD, MPH, Department of Dermatology, Tulane University School of Medicine, 1430 Tulane Ave, #8036, New Orleans, LA 70112 (ajafari@tulane.edu).

Article PDF
Article PDF

To the Editor:

Lenalidomide is a thalidomide analogue used to treat various hematologic malignancies, including non-Hodgkin lymphoma, myelodysplastic syndrome, and multiple myeloma (MM).1 Lenalidomide is referred to as a degrader therapeutic because it induces targeted protein degradation of disease-relevant proteins (eg, Ikaros family zinc finger protein 1 [IKZF1], Ikaros family zinc finger protein 3 [IKZF3], and casein kinase I isoform-α [CK1α]) as its primary mechanism of action.1,2 Although cutaneous adverse events are relatively common among thalidomide analogues, the morphologic and histopathologic descriptions of these drug eruptions have not been fully elucidated.3,4 We report a novel pityriasiform drug eruption followed by a clinical eruption suggestive of blaschkitis in a patient with MM who was being treated with lenalidomide.

A 76-year-old man presented to the dermatology clinic with a progressive, mildly pruritic eruption on the chest and axillae of 1 year’s duration. He had a medical history of chronic hepatitis B, malignant carcinoid tumor of the colon, prostate cancer, and MM. The eruption emerged 1 to 2 weeks after the patient started oral lenalidomide 10 mg/d and oral dexamethasone40 mg/wk following autologous stem cell transplantation for MM. The patient had not received any other therapy for MM.

Physical examination revealed multiple erythematous, hyperpigmented, scaly papules and plaques on the lateral chest and within the axillae (Figure 1). A skin biopsy from the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate with scattered eosinophils, neutrophils, and extravasated erythrocytes. The overlying epidermis showed spongiosis with parakeratosis in addition to lymphocytic exocytosis (Figure 2). No fungal organisms were highlighted on periodic acid–Schiff staining. After this evaluation, we recommended that the patient discontinue lenalidomide and start taking a topical over-the-counter corticosteroid for 2 weeks. Over time, he noted marked improvement in the eruption and associated pruritus.

Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.
FIGURE 1. Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.

After a drug holiday of 2 months, the patient resumed a maintenance dosage of oral lenalidomide 10 mg/d. Four or 5 days after restarting lenalidomide, a pruritic eruption appeared that involved the axillae and the left lower abdomen, circling around to the left lower back. The axillary eruption resolved with a topical over-the-counter corticosteroid; the abdominal eruption persisted.

A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes.
FIGURE 2. A and B, A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes. The overlying epidermis was spongiotic with parakeratosis and lymphocytic exocytosis (H&E, original magnifications ×100 and ×200).

At the 3-month follow-up visit, physical examination revealed erythematous macules and papules that coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline (Figure 3).

At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossi
FIGURE 3. A and B, At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline.

We recommended that the patient continue treatment through this eruption; he was instructed to apply a corticosteroid cream and resume lenalidomide at the maintenance dosage. A month later, he reported that the eruption and associated pruritus resolved with the corticosteroid cream and resumption of the maintenance dose of lenalidomide. The patient noted no further spread of the eruption.

Cutaneous adverse events are common following lenalidomide. In prior trials, the overall incidence of any-grade rash following lenalidomide exposure was 22% to 33%.5 A meta-analysis of 10 trials determined the overall incidence of all-grade and high-grade cutaneous adverse events after exposure to lenalidomide was 27.2% and 3.6%, respectively.6 Our case represents a pityriasiform eruption due to lenalidomide followed by a secondary eruption suggestive of blaschkitis.

 

 

The rash due to lenalidomide has been described as morbilliform, urticarial, dermatitic, acneform, and undefined.7 Lenalidomide-induced rash typically develops during the first month of therapy, similar to our patient’s presentation. It has even been observed in the first week of therapy.8 Severe reactions such as Stevens-Johnson syndrome and toxic epidermal necrolysis have been reported.5,6 Risk factors associated with rash secondary to lenalidomide include advanced age (≥70 years), presence of Bence-Jones protein-type MM in urine, and no prior chemotherapy.8 Our patient had 2 of these risk factors: advanced age and no prior chemotherapy for MM. The exact pathogenesis by which lenalidomide leads to a pityriasiform eruption, as in our patient, or to a rash in general is unclear. Studies have hypothesized that a lenalidomide-induced rash could be attributable to a delayed hypersensitivity type IV reaction or to a reaction related to the molecular mechanism of action of the drug.9

At the molecular level, the antimyeloma effects of lenalidomide include promoting degradation of transcription factors IKZF1 and IKZF3, which subsequently increases production of IL-2.1,2,9 Recombinant IL-2 has been associated with an increased incidence of rash in other cancers.9 Overexpression of programmed death 1(PD-1) and its ligand (PD-L1) has been demonstrated in MM; lenalidomide has been shown to downregulate both PD-1 and PD-L1. Patients receiving PD-1 and PD-L1 inhibitors commonly have developed rash.9 However, the association between lenalidomide and its downregulation of PD-1 and PD-L1 leading to rash has not been fully elucidated. Given the multiple malignancies in our patient—MM, prostate cancer, malignant carcinoid tumor—an underlying paraneoplastic phenomenon may be possible. Additionally, because our patient initially received dexamethasone along with lenalidomide, the manifestation of the initial pityriasiform rash may have been less severe due to the steroid use. Although our patient underwent a 2-month drug holiday following the initial pityriasiform eruption, most lenalidomide-induced rashes do not necessitate discontinuation of the drug.5,7

Our patient’s secondary drug eruption was clinically suggestive of lenalidomide-induced blaschkitis. A report of a German patient with plasmacytoma described a unilateral papular exanthem that developed 4 months after lenalidomide was initiated.10 The papular exanthem following the lines of Blaschko lines extended from that patient’s posterior left foot to the calf and on to the thigh and flank,10 which was more extensive than our patient’s eruption. Blaschkitis in this patient resolved with a corticosteroid cream and UV light therapy10; lenalidomide was not discontinued, similar to our patient.

The pathogenesis of our patient’s secondary eruption that preferentially involved the lines of Blaschko is unclear. After the initial pityriasiform eruption, the secondary eruption was blaschkitis. Distinguishing dermatomes from the lines of Blaschko, which are thought to represent pathways of epidermal cell migration and proliferation during embryologic development, is important. Genodermatoses such as incontinentia pigmenti and hypomelanosis of Ito involve the lines of Blaschko11; other disorders in the differential diagnosis of linear configurations include linear lichen planus, linear cutaneous lupus erythematosus, linear morphea, and lichen striatus.11 Notably, drug-induced blaschkitis is rare.

Cutaneous adverse reactions from thalidomide analogues are relatively common. Our case of lenalidomide-associated blaschkitis that developed following an initial pityriasiform drug eruption in a patient with MM highlights that dermatologists need to collaborate with the oncologist regarding the severity of drug eruptions to determine if the patient should continue treatment through the cutaneous eruptions or discontinue a vital medication.

To the Editor:

Lenalidomide is a thalidomide analogue used to treat various hematologic malignancies, including non-Hodgkin lymphoma, myelodysplastic syndrome, and multiple myeloma (MM).1 Lenalidomide is referred to as a degrader therapeutic because it induces targeted protein degradation of disease-relevant proteins (eg, Ikaros family zinc finger protein 1 [IKZF1], Ikaros family zinc finger protein 3 [IKZF3], and casein kinase I isoform-α [CK1α]) as its primary mechanism of action.1,2 Although cutaneous adverse events are relatively common among thalidomide analogues, the morphologic and histopathologic descriptions of these drug eruptions have not been fully elucidated.3,4 We report a novel pityriasiform drug eruption followed by a clinical eruption suggestive of blaschkitis in a patient with MM who was being treated with lenalidomide.

A 76-year-old man presented to the dermatology clinic with a progressive, mildly pruritic eruption on the chest and axillae of 1 year’s duration. He had a medical history of chronic hepatitis B, malignant carcinoid tumor of the colon, prostate cancer, and MM. The eruption emerged 1 to 2 weeks after the patient started oral lenalidomide 10 mg/d and oral dexamethasone40 mg/wk following autologous stem cell transplantation for MM. The patient had not received any other therapy for MM.

Physical examination revealed multiple erythematous, hyperpigmented, scaly papules and plaques on the lateral chest and within the axillae (Figure 1). A skin biopsy from the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate with scattered eosinophils, neutrophils, and extravasated erythrocytes. The overlying epidermis showed spongiosis with parakeratosis in addition to lymphocytic exocytosis (Figure 2). No fungal organisms were highlighted on periodic acid–Schiff staining. After this evaluation, we recommended that the patient discontinue lenalidomide and start taking a topical over-the-counter corticosteroid for 2 weeks. Over time, he noted marked improvement in the eruption and associated pruritus.

Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.
FIGURE 1. Multiple erythematous, hyperpigmented, pityriasiform papules and plaques within the axillae.

After a drug holiday of 2 months, the patient resumed a maintenance dosage of oral lenalidomide 10 mg/d. Four or 5 days after restarting lenalidomide, a pruritic eruption appeared that involved the axillae and the left lower abdomen, circling around to the left lower back. The axillary eruption resolved with a topical over-the-counter corticosteroid; the abdominal eruption persisted.

A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes.
FIGURE 2. A and B, A biopsy specimen of the left axilla demonstrated a mild lichenoid and perivascular lymphocytic infiltrate containing scattered eosinophils, neutrophils, and a few extravasated erythrocytes. The overlying epidermis was spongiotic with parakeratosis and lymphocytic exocytosis (H&E, original magnifications ×100 and ×200).

At the 3-month follow-up visit, physical examination revealed erythematous macules and papules that coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline (Figure 3).

At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossi
FIGURE 3. A and B, At a 3-month follow-up visit, erythematous macules and papules coalesced over a salmon-colored base along the lines of Blaschko extending from the left lower abdominal quadrant, crossing the left flank, and continuing to the left lower back without crossing the midline.

We recommended that the patient continue treatment through this eruption; he was instructed to apply a corticosteroid cream and resume lenalidomide at the maintenance dosage. A month later, he reported that the eruption and associated pruritus resolved with the corticosteroid cream and resumption of the maintenance dose of lenalidomide. The patient noted no further spread of the eruption.

Cutaneous adverse events are common following lenalidomide. In prior trials, the overall incidence of any-grade rash following lenalidomide exposure was 22% to 33%.5 A meta-analysis of 10 trials determined the overall incidence of all-grade and high-grade cutaneous adverse events after exposure to lenalidomide was 27.2% and 3.6%, respectively.6 Our case represents a pityriasiform eruption due to lenalidomide followed by a secondary eruption suggestive of blaschkitis.

 

 

The rash due to lenalidomide has been described as morbilliform, urticarial, dermatitic, acneform, and undefined.7 Lenalidomide-induced rash typically develops during the first month of therapy, similar to our patient’s presentation. It has even been observed in the first week of therapy.8 Severe reactions such as Stevens-Johnson syndrome and toxic epidermal necrolysis have been reported.5,6 Risk factors associated with rash secondary to lenalidomide include advanced age (≥70 years), presence of Bence-Jones protein-type MM in urine, and no prior chemotherapy.8 Our patient had 2 of these risk factors: advanced age and no prior chemotherapy for MM. The exact pathogenesis by which lenalidomide leads to a pityriasiform eruption, as in our patient, or to a rash in general is unclear. Studies have hypothesized that a lenalidomide-induced rash could be attributable to a delayed hypersensitivity type IV reaction or to a reaction related to the molecular mechanism of action of the drug.9

At the molecular level, the antimyeloma effects of lenalidomide include promoting degradation of transcription factors IKZF1 and IKZF3, which subsequently increases production of IL-2.1,2,9 Recombinant IL-2 has been associated with an increased incidence of rash in other cancers.9 Overexpression of programmed death 1(PD-1) and its ligand (PD-L1) has been demonstrated in MM; lenalidomide has been shown to downregulate both PD-1 and PD-L1. Patients receiving PD-1 and PD-L1 inhibitors commonly have developed rash.9 However, the association between lenalidomide and its downregulation of PD-1 and PD-L1 leading to rash has not been fully elucidated. Given the multiple malignancies in our patient—MM, prostate cancer, malignant carcinoid tumor—an underlying paraneoplastic phenomenon may be possible. Additionally, because our patient initially received dexamethasone along with lenalidomide, the manifestation of the initial pityriasiform rash may have been less severe due to the steroid use. Although our patient underwent a 2-month drug holiday following the initial pityriasiform eruption, most lenalidomide-induced rashes do not necessitate discontinuation of the drug.5,7

Our patient’s secondary drug eruption was clinically suggestive of lenalidomide-induced blaschkitis. A report of a German patient with plasmacytoma described a unilateral papular exanthem that developed 4 months after lenalidomide was initiated.10 The papular exanthem following the lines of Blaschko lines extended from that patient’s posterior left foot to the calf and on to the thigh and flank,10 which was more extensive than our patient’s eruption. Blaschkitis in this patient resolved with a corticosteroid cream and UV light therapy10; lenalidomide was not discontinued, similar to our patient.

The pathogenesis of our patient’s secondary eruption that preferentially involved the lines of Blaschko is unclear. After the initial pityriasiform eruption, the secondary eruption was blaschkitis. Distinguishing dermatomes from the lines of Blaschko, which are thought to represent pathways of epidermal cell migration and proliferation during embryologic development, is important. Genodermatoses such as incontinentia pigmenti and hypomelanosis of Ito involve the lines of Blaschko11; other disorders in the differential diagnosis of linear configurations include linear lichen planus, linear cutaneous lupus erythematosus, linear morphea, and lichen striatus.11 Notably, drug-induced blaschkitis is rare.

Cutaneous adverse reactions from thalidomide analogues are relatively common. Our case of lenalidomide-associated blaschkitis that developed following an initial pityriasiform drug eruption in a patient with MM highlights that dermatologists need to collaborate with the oncologist regarding the severity of drug eruptions to determine if the patient should continue treatment through the cutaneous eruptions or discontinue a vital medication.

References
  1. Jan M, Sperling AS, Ebert BL. Cancer therapies based on targeted protein degradation—lessons learned with lenalidomide. Nat Rev Clin Oncol. 2021;18:401-417. doi:10.1038/s41571-021-00479-z
  2. Shah UA, Mailankody S. Emerging immunotherapies in multiple myeloma. BMJ. 2020;370:3176. doi:10.1136/BMJ.M3176
  3. Richardson PG, Blood E, Mitsiades CS, et al. A randomized phase 2 study of lenalidomide therapy for patients with relapsed or relapsed and refractory multiple myeloma. Blood. 2006;108:3458-3464. doi:10.1182/BLOOD-2006-04-015909
  4. Benboubker L, Dimopoulos MA, Dispenzieri A, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med. 2014;371:906-917. doi:10.1056/NEJMOA1402551
  5. Tinsley SM, Kurtin SE, Ridgeway JA. Practical management of lenalidomide-related rash. Clin Lymphoma Myeloma Leuk. 2015;15(suppl):S64-S69. doi:10.1016/J.CLML.2015.02.008
  6. Nardone B, Wu S, Garden BC, et al. Risk of rash associated with lenalidomide in cancer patients: a systematic review of the literature and meta-analysis. Clin Lymphoma Myeloma Leuk. 2013;13:424-429. doi:10.1016/J.CLML.2013.03.006
  7. Sviggum HP, Davis MDP, Rajkumar SV, et al. Dermatologic adverse effects of lenalidomide therapy for amyloidosis and multiple myeloma. Arch Dermatol. 2006;142:1298-1302. doi:10.1001/ARCHDERM.142.10.1298
  8. Sugi T, Nishigami Y, Saigo H, et al. Analysis of risk factors for lenalidomide-associated skin rash in patients with multiple myeloma. Leuk Lymphoma. 2021;62:1405-1410. doi:10.1080/10428194.2021.1876867
  9. Barley K, He W, Agarwal S, et al. Outcomes and management of lenalidomide-associated rash in patients with multiple myeloma. Leuk Lymphoma. 2016;57:2510-2515. doi:10.3109/10428194.2016.1151507
  10. Grape J, Frosch P. Papular drug eruption along the lines of Blaschko caused by lenalidomide [in German]. Hautarzt. 2011;62:618-620. doi:10.1007/S00105-010-2121-6
  11. Bolognia JL, Orlow SJ, Glick SA. Lines of Blaschko. J Am Acad Dermatol. 1994;31(2 pt 1):157-190. doi:10.1016/S0190-9622(94)70143-1
References
  1. Jan M, Sperling AS, Ebert BL. Cancer therapies based on targeted protein degradation—lessons learned with lenalidomide. Nat Rev Clin Oncol. 2021;18:401-417. doi:10.1038/s41571-021-00479-z
  2. Shah UA, Mailankody S. Emerging immunotherapies in multiple myeloma. BMJ. 2020;370:3176. doi:10.1136/BMJ.M3176
  3. Richardson PG, Blood E, Mitsiades CS, et al. A randomized phase 2 study of lenalidomide therapy for patients with relapsed or relapsed and refractory multiple myeloma. Blood. 2006;108:3458-3464. doi:10.1182/BLOOD-2006-04-015909
  4. Benboubker L, Dimopoulos MA, Dispenzieri A, et al. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med. 2014;371:906-917. doi:10.1056/NEJMOA1402551
  5. Tinsley SM, Kurtin SE, Ridgeway JA. Practical management of lenalidomide-related rash. Clin Lymphoma Myeloma Leuk. 2015;15(suppl):S64-S69. doi:10.1016/J.CLML.2015.02.008
  6. Nardone B, Wu S, Garden BC, et al. Risk of rash associated with lenalidomide in cancer patients: a systematic review of the literature and meta-analysis. Clin Lymphoma Myeloma Leuk. 2013;13:424-429. doi:10.1016/J.CLML.2013.03.006
  7. Sviggum HP, Davis MDP, Rajkumar SV, et al. Dermatologic adverse effects of lenalidomide therapy for amyloidosis and multiple myeloma. Arch Dermatol. 2006;142:1298-1302. doi:10.1001/ARCHDERM.142.10.1298
  8. Sugi T, Nishigami Y, Saigo H, et al. Analysis of risk factors for lenalidomide-associated skin rash in patients with multiple myeloma. Leuk Lymphoma. 2021;62:1405-1410. doi:10.1080/10428194.2021.1876867
  9. Barley K, He W, Agarwal S, et al. Outcomes and management of lenalidomide-associated rash in patients with multiple myeloma. Leuk Lymphoma. 2016;57:2510-2515. doi:10.3109/10428194.2016.1151507
  10. Grape J, Frosch P. Papular drug eruption along the lines of Blaschko caused by lenalidomide [in German]. Hautarzt. 2011;62:618-620. doi:10.1007/S00105-010-2121-6
  11. Bolognia JL, Orlow SJ, Glick SA. Lines of Blaschko. J Am Acad Dermatol. 1994;31(2 pt 1):157-190. doi:10.1016/S0190-9622(94)70143-1
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E27-E29
Page Number
E27-E29
Publications
Publications
Topics
Article Type
Display Headline
Thalidomide Analogue Drug Eruption Along the Lines of Blaschko
Display Headline
Thalidomide Analogue Drug Eruption Along the Lines of Blaschko
Sections
Inside the Article

Practice Points

  • Dermatologists should be aware of the variety of cutaneous adverse events that can arise from the use of immunotherapeutic agents for hematologic malignancies.
  • Some cutaneous reactions to immunotherapeutic medications, such as pityriasiform eruption and blaschkitis, generally are benign and may not necessitate halting an important therapy.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

The Role of Toluidine Blue in Mohs Micrographic Surgery: A Systematic Review

Article Type
Changed
Thu, 12/21/2023 - 08:12
Display Headline
The Role of Toluidine Blue in Mohs Micrographic Surgery: A Systematic Review

Toluidine blue (TB), a dye with metachromatic staining properties, was developed in 1856 by William Henry Perkin.1 Metachromasia is a perceptible change in the color of staining of living tissue due to the electrochemical properties of the tissue. Tissues that contain high concentrations of ionized sulfate and phosphate groups (high concentrations of free electronegative groups) form polymeric aggregates of the basic dye solution that alter the absorbed wavelengths of light.2 The function of this characteristic is to use a single dye to highlight different structures in tissue based on their relative chemical differences.3

Toluidine blue primarily was used within the dye industry until the 1960s, when it was first used in vital staining of the oral mucosa.2 Because of the tissue absorption potential, this technique was used to detect the location of oral malignancies.4 Since then, TB has progressively been used for staining fresh frozen sections in Mohs micrographic surgery (MMS). In a 2003 survey study (N=310), 16.8% of surgeons performing MMS reported using TB in their laboratory.5 We sought to systematically review the published literature describing the uses of TB in the setting of fresh frozen sections and MMS.

Methods

We conducted a systematic search of the PubMed and Cochrane databases for articles published before December 1, 2019, to identify any relevant studies in English. Electronic searches were performed using the terms toluidine blue and Mohs or Mohs micrographic surgery. We manually checked the bibliographies of the identified articles to further identify eligible studies.

Eligibility Criteria—The inclusion criteria were articles that (1) considered TB in the context of MMS, (2) were published in peer-reviewed journals, (3) were published in English, and (4) were available as full text. Systematic reviews were excluded.

Data Extraction and Outcomes—All relevant information regarding the study characteristics, including design, level of evidence, methodologic quality of evidence, pathology examined, and outcome measures, were collected by 2 independent reviewers (T.L. and A.D.) using a predetermined data sheet. The same 2 reviewers were used for all steps of the review process, data were independently obtained, and any discrepancy was introduced for a third opinion (D.H.) and agreed upon by the majority.

Quality Assessment—The level of evidence was evaluated based on the criteria of the Oxford Centre for Evidence-Based Medicine. Two reviewers (T.L. and A.D.) graded each article included in the review.

PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.
FIGURE 1. PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.

Results

A total of 25 articles were reviewed. After the titles and abstracts were screened for relevance, 12 articles remained (Figure 1). Of these, 1 compared basal cell carcinoma (BCC) and squamous cell carcinoma (SCC), 4 were related to BCC, 3 were related to SCC, 1 was related to microcystic adnexal carcinoma (MAC), 1 was related to primary cutaneous adenoid cystic carcinoma (PCACC), and 2 were related to technical aspects of the staining process (Table 1).

Summary of Articles Published on Toluidine Blue in Mohs Micrographic Surgery

 

 

A majority of the articles included in this review were qualitative and observational in nature, describing the staining characteristics of TB. Study characteristics are summarized in Table 1.

Comment

Basal Cell Carcinoma—Toluidine blue staining characteristics help to identify BCC nests by differentiating them from hair follicles in frozen sections. The metachromatic characteristic of TB stains the inner root sheath deep blue and highlights the surrounding stromal mucin of BCC a magenta color.18,19 In hematoxylin and eosin (H&E) stains, these 2 distinct structures can be differentiated by cleft formation around tumor nests, mitotic figures, and the lack of a fibrous sheath present in BCC tumors.20 The advantages and limitations of TB staining of BCC are presented in Table 2.

Advantages and Limitations of Toluidine Blue Staining in BCC and SCC

Humphreys et al6 suggested a noticeable difference between H&E and TB in the staining of cellular and stromal components. The nuclear detail of tumor cells was subjectively sharper and clearer with TB staining. The staining of stromal components may provide the most assistance in locating BCC islands. Mucopolysaccharide staining may be absent in H&E but stain a deep magenta with TB. Although the presence of mucopolysaccharides does not specifically indicate a tumor, it may prompt further attention and provide an indicator for sparse and infiltrative tumor cells.6 The metachromatic stromal change may indicate a narrow tumor-free margin where additional deeper sections often reveal tumor that may warrant additional resection margin in more aggressive malignancies. In particular, sclerosing/morpheaform BCCs have been shown to induce glycosaminoglycan synthesis and are highlighted more readily with TB than with H&E when compared to surrounding tissue.21 This differentiation in staining has remained a popular reason to routinely incorporate TB into the staining of infiltrative and morpheaform variants of BCC. Additionally, stromal mast cells are believed to be more abundant in the stroma of BCC and are more readily visualized in tissue specimens stained with TB, appearing as bright purple metachromatic granules. These granules are larger than normal and are increased in number.6

The margin behavior of BCC stained with TB was further characterized by Goldberg et al,8 who coined the term setting sun sign, which may be present in sequential sections of a disappearing nodule of a BCC tumor. Stroma, inflammatory infiltrate, and mast cells produce a magenta glow surrounding BCC tumors that is reminiscent of a setting sun (Figure 2). Invasive BCC is considered variable in this presentation, primarily because of zones of cell-free fluid and edema or the second area of inflammatory cells. This unique sign may benefit the inspecting Mohs surgeon by providing a clue to an underlying process that may have residual BCC tumors. The setting sun sign also may assist in identifying exact surgical margins.8

Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.
FIGURE 2. A and B, Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.

The nasal surface has a predilection for BCC.22 The skin of the nose has numerous look-alike structures to consider for complete tumor removal and avoidance of unnecessary removal. One challenge is distinguishing follicular basaloid proliferations (FBP) from BCC, a scenario that is more common on the nose.22 When TB staining was used, the sensitivity for detecting FBP reached 100% in 34 cases reviewed by Donaldson and Weber.10 None of the cases examined showed TB metachromasia surrounding FBP, thus indicating that TB can dependably identify this benign entity. Conversely, 5% (N=279) of BCCs confirmed on H&E did not exhibit surrounding TB metachromasia. This finding is concerning regarding the specificity of TB staining for BCC, but the authors of this study suggested the possibility that these exceptions were benign “simulants” (ie, trichoepithelioma) of BCC.10

The use of TB also has been shown to be statistically beneficial in Mohs training. In a single-center, single-fellow experiment, the sensitivity and specificity of using TB for BCC were extrapolated.9 Using TB as an adjunct in deep sections showed superior sensitivity to H&E alone in identifying BCC, increasing sensitivity from 96.3% to 99.7%. In a cohort of 352 BCC excisions and frozen sections, only 1 BCC was not completely excised. If H&E only had been performed, the fellow would have missed 13 residual BCC tumors.9

Bennett and Taher7 described a case in which hyaluronic acid (HA) from a filler injection was confused with the HA surrounding BCC tumor nests. They found that when TB is used as an adjunct, the HA filler is easier to differentiate from the HA surrounding the BCC tumor nests. In frozen sections stained with TB, the HA filler appeared as an amorphous, metachromatic, reddish-purple, whereas the HA surrounding the BCC tumor nests appeared as a well-defined red. These findings were less obvious in the same sections stained with H&E alone.7

 

 

Squamous Cell Carcinoma—In early investigations, the utility of TB in identifying SCC in frozen sections was thought to be limited. The description by Humphreys and colleagues6 of staining characteristics in SCC suggested that the nuclear detail that H&E provides is more easily recognized. The deep aqua nuclear staining produced with TB was considered more difficult to observe than the cytoplasmic eosinophilia of pyknotic and keratinizing cells in H&E.6

Toluidine blue may be beneficial in providing unique staining characteristics to further detail tumors that are difficult to interpret, such as spindle cell SCC and perineural invasion of aggressive SCC. In H&E, squamous cells of spindle cell SCC (scSCC) blend into the background of inflammatory cells and can be perceptibly difficult to locate. A small cohort of 3 Mohs surgeons who routinely use H&E were surveyed on their ability to detect a proven scSCC in H&E or TB by photograph.12 All 3 were able to detect the scSCC in the TB photographs, but only 2 of 3 were able to detect it in H&E photographs. All 3 surgeons agreed that TB was preferable to H&E for this tumor type. These findings suggested that TB may be superior and preferred over H&E for visualizing tumor cells of scSCC.12 The TB staining characteristics of perineural invasion of aggressive SCC have been referred to as the perineural corona sign because of the bright magenta stain that forms around affected nerves.13 Drosou et al13 suggested that TB may enhance the diagnostic accuracy for perineural SCC.

Rare Tumors—The adjunctive use of TB with H&E has been examined in rare tumors. Published reports have highlighted its use in MMS for treating MAC and PCACC. Toluidine blue exhibits staining advantages for these tumors. It may render isolated nests and perineural invasion of MAC more easily visible on frozen section.15

Although PCACC is rare, the recurrence rate is high.23 Toluidine blue has been used with MMS to ensure complete removal and higher cure rates. The metachromatic nature of TB is advantageous in staining the HA present in these tumors. Those who have reported the use of TB for PCACC prefer it to H&E for frozen sections.14

Technical Aspects—The staining time for TB-treated slides is reduced compared to H&E staining; staining can be efficiently done in frozen sections in less than 2.5 minutes using the method shown in Table 3.17 In comparison, typical H&E staining takes 9 minutes, and older TB techniques take 7 minutes.6

Rapid Toluidine Blue Staining Protocol

Conclusion

Toluidine blue may play an important and helpful role in the successful diagnosis and treatment of particular cutaneous tumors by providing additional diagnostic information. Although surgeons performing MMS will continue using the staining protocols with which they are most comfortable, adjunctive use of TB over time may provide an additional benefit at low risk for disrupting practice efficiency or workflow. Many Mohs surgeons are accustomed to using this stain, even preferring to interpret only TB-stained slides for cutaneous malignancy. Most published studies on this topic have been observational in nature, and additional controlled trials may be warranted to determine the effects on outcomes in real-world practice.

References
  1. Culling CF, Allison TR. Cellular Pathology Technique. 4th ed. Butterworths; 1985.
  2. Bergeron JA, Singer M. Metachromasy: an experimental and theoretical reevaluation. J Biophys Biochem Cytol. 1958;4:433-457. doi:10.1083/jcb.4.4.433
  3. Epstein JB, Scully C, Spinelli J. Toluidine blue and Lugol’s iodine application in the assessment of oral malignant disease and lesions at risk of malignancy. J Oral Pathol Med. 1992;21:160-163. doi:10.1111/j.1600-0714.1992.tb00094.x
  4. Warnakulasuriya KA, Johnson NW. Sensitivity and specificity of OraScan (R) toluidine blue mouthrinse in the detection of oral cancer and precancer. J Oral Pathol Med. 1996;25:97-103. doi:10.1111/j.1600-0714.1996.tb00201.x
  5. Silapunt S, Peterson SR, Alcalay J, et al. Mohs tissue mapping and processing: a survey study. Dermatol Surg. 2003;29:1109-1112; discussion 1112.
  6. Humphreys TR, Nemeth A, McCrevey S, et al. A pilot study comparing toluidine blue and hematoxylin and eosin staining of basal cell and squamous cell carcinoma during Mohs surgery. Dermatol Surg. 1996;22:693-697. doi:10.1111/j.1524-4725.1996.tb00619.x
  7. Bennett R, Taher M. Restylane persistent for 23 months found during Mohs micrographic surgery: a source of confusion with hyaluronic acid surrounding basal cell carcinoma. Dermatol Surg. 2005;31:1366-1369. doi:10.1111/j.1524-4725.2005.31223
  8. Goldberg LH, Wang SQ, Kimyai-Asadi A. The setting sun sign: visualizing the margins of a basal cell carcinoma on serial frozen sections stained with toluidine blue. Dermatol Surg. 2007;33:761-763. doi:10.1111/j.1524-4725.2007.33158.x
  9. Tehrani H, May K, Morris A, et al. Does the dual use of toluidine blue and hematoxylin and eosin staining improve basal cell carcinoma detection by Mohs surgery trainees? Dermatol Surg. 2013;39:995-1000. doi:10.1111/dsu.12180
  10. Donaldson MR, Weber LA. Toluidine blue supports differentiation of folliculocentric basaloid proliferation from basal cell carcinoma on frozen sections in a small single-practice cohort. Dermatol Surg. 2017;43:1303-1306. doi:10.1097/DSS.0000000000001107
  11. Styperek AR, Goldberg LH, Goldschmidt LE, et al. Toluidine blue and hematoxylin and eosin stains are comparable in evaluating squamous cell carcinoma during Mohs. Dermatol Surg. 2016;42:1279-1284. doi:10.1097/DSS.0000000000000872
  12. Trieu D, Drosou A, Goldberg LH, et al. Detecting spindle cell squamous cell carcinomas with toluidine blue on frozen sections. Dermatol Surg. 2014;40:1259-1260. doi:10.1097/DSS.0000000000000147
  13. Drosou A, Trieu D, Goldberg LH, et al. The perineural corona sign: enhancing detection of perineural squamous cell carcinoma during Mohs micrographic surgery with toluidine blue stain. J Am Acad Dermatol. 2014;71:826-827. doi:10.1016/j.jaad.2014.04.076
  14. Chesser RS, Bertler DE, Fitzpatrick JE, et al. Primary cutaneous adenoid cystic carcinoma treated with Mohs micrographic surgery toluidine blue technique. J Dermatol Surg Oncol. 1992;18:175-176. doi:10.1111/j.1524-4725.1992.tb02794.x
  15. Wang SQ, Goldberg LH, Nemeth A. The merits of adding toluidine blue-stained slides in Mohs surgery in the treatment of a microcystic adnexal carcinoma. J Am Acad Dermatol. 2007;56:1067-1069. doi:10.1016/j.jaad.2007.01.008
  16. Chen CL, Wilson S, Afzalneia R, et al. Topical aluminum chloride and Monsel’s solution block toluidine blue staining in Mohs frozen sections: mechanism and solution. Dermatol Surg. 2019;45:1019-1025. doi:10.1097/DSS.0000000000001761
  17. Todd MM, Lee JW, Marks VJ. Rapid toluidine blue stain for Mohs’ micrographic surgery. Dermatol Surg. 2005;31:244-245. doi:10.1111/j.1524-4725.2005.31053
  18. Picoto AM, Picoto A. Technical procedures for Mohs fresh tissue surgery. J Derm Surg Oncol. 1986;12:134-138. doi:10.1111/j.1524-4725.1986.tb01442.x
  19. Sperling LC, Winton GB. The transverse anatomy of androgenic alopecia. J Derm Surg Oncol. 1990;16:1127-1133. doi:10.1111/j.1524 -4725.1990.tb00024.x
  20. Smith-Zagone MJ, Schwartz MR. Frozen section of skin specimens. Arch Pathol Lab Med. 2005;129:1536-1543. doi:10.5858/2005-129-1536-FSOSS
  21. Moy RL, Potter TS, Uitto J. Increased glycosaminoglycans production in sclerosing basal cell carcinoma–derived fibroblasts and stimulation of normal skin fibroblast glycosaminoglycans production by a cytokine-derived from sclerosing basal cell carcinoma. Dermatol Surg. 2000;26:1029-1036. doi:10.1046/j.1524-4725.2000.0260111029.x
  22. Leshin B, White WL. Folliculocentric basaloid proliferation. The bulge (der Wulst) revisited. Arch Dermatol. 1990;126:900-906. doi:10.1001/archderm.126.7.900
  23. Seab JA, Graham JH. Primary cutaneous adenoid cystic carcinoma.J Am Acad Dermatol. 1987;17:113-118. doi:10.1016/s0190 -9622(87)70182-0
Article PDF
Author and Disclosure Information

Dr. Long is from the Health Corporation of America and Virginia College of Osteopathic Medicine, Blacksburg. Dr. Dunn is in private practice, Tampa, Florida. Drs. Hill and Akin are from the Department of Dermatology, Texas Tech University Health Sciences Center, Lubbock. Dr. Akin also is from Midland Dermatology and Skin Cancer Center, Texas. Dr. Goldberg is from DermSurgery Associates, Houston, Texas.

The authors report no conflicts of interest.

Correspondence: Tyler Long, DO, HCA LewisGale Hospital Montgomery Medical Education, 700 S Main St, Blacksburg, VA 24060 (longjtyler@yahoo.com).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E6-E11
Sections
Author and Disclosure Information

Dr. Long is from the Health Corporation of America and Virginia College of Osteopathic Medicine, Blacksburg. Dr. Dunn is in private practice, Tampa, Florida. Drs. Hill and Akin are from the Department of Dermatology, Texas Tech University Health Sciences Center, Lubbock. Dr. Akin also is from Midland Dermatology and Skin Cancer Center, Texas. Dr. Goldberg is from DermSurgery Associates, Houston, Texas.

The authors report no conflicts of interest.

Correspondence: Tyler Long, DO, HCA LewisGale Hospital Montgomery Medical Education, 700 S Main St, Blacksburg, VA 24060 (longjtyler@yahoo.com).

Author and Disclosure Information

Dr. Long is from the Health Corporation of America and Virginia College of Osteopathic Medicine, Blacksburg. Dr. Dunn is in private practice, Tampa, Florida. Drs. Hill and Akin are from the Department of Dermatology, Texas Tech University Health Sciences Center, Lubbock. Dr. Akin also is from Midland Dermatology and Skin Cancer Center, Texas. Dr. Goldberg is from DermSurgery Associates, Houston, Texas.

The authors report no conflicts of interest.

Correspondence: Tyler Long, DO, HCA LewisGale Hospital Montgomery Medical Education, 700 S Main St, Blacksburg, VA 24060 (longjtyler@yahoo.com).

Article PDF
Article PDF

Toluidine blue (TB), a dye with metachromatic staining properties, was developed in 1856 by William Henry Perkin.1 Metachromasia is a perceptible change in the color of staining of living tissue due to the electrochemical properties of the tissue. Tissues that contain high concentrations of ionized sulfate and phosphate groups (high concentrations of free electronegative groups) form polymeric aggregates of the basic dye solution that alter the absorbed wavelengths of light.2 The function of this characteristic is to use a single dye to highlight different structures in tissue based on their relative chemical differences.3

Toluidine blue primarily was used within the dye industry until the 1960s, when it was first used in vital staining of the oral mucosa.2 Because of the tissue absorption potential, this technique was used to detect the location of oral malignancies.4 Since then, TB has progressively been used for staining fresh frozen sections in Mohs micrographic surgery (MMS). In a 2003 survey study (N=310), 16.8% of surgeons performing MMS reported using TB in their laboratory.5 We sought to systematically review the published literature describing the uses of TB in the setting of fresh frozen sections and MMS.

Methods

We conducted a systematic search of the PubMed and Cochrane databases for articles published before December 1, 2019, to identify any relevant studies in English. Electronic searches were performed using the terms toluidine blue and Mohs or Mohs micrographic surgery. We manually checked the bibliographies of the identified articles to further identify eligible studies.

Eligibility Criteria—The inclusion criteria were articles that (1) considered TB in the context of MMS, (2) were published in peer-reviewed journals, (3) were published in English, and (4) were available as full text. Systematic reviews were excluded.

Data Extraction and Outcomes—All relevant information regarding the study characteristics, including design, level of evidence, methodologic quality of evidence, pathology examined, and outcome measures, were collected by 2 independent reviewers (T.L. and A.D.) using a predetermined data sheet. The same 2 reviewers were used for all steps of the review process, data were independently obtained, and any discrepancy was introduced for a third opinion (D.H.) and agreed upon by the majority.

Quality Assessment—The level of evidence was evaluated based on the criteria of the Oxford Centre for Evidence-Based Medicine. Two reviewers (T.L. and A.D.) graded each article included in the review.

PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.
FIGURE 1. PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.

Results

A total of 25 articles were reviewed. After the titles and abstracts were screened for relevance, 12 articles remained (Figure 1). Of these, 1 compared basal cell carcinoma (BCC) and squamous cell carcinoma (SCC), 4 were related to BCC, 3 were related to SCC, 1 was related to microcystic adnexal carcinoma (MAC), 1 was related to primary cutaneous adenoid cystic carcinoma (PCACC), and 2 were related to technical aspects of the staining process (Table 1).

Summary of Articles Published on Toluidine Blue in Mohs Micrographic Surgery

 

 

A majority of the articles included in this review were qualitative and observational in nature, describing the staining characteristics of TB. Study characteristics are summarized in Table 1.

Comment

Basal Cell Carcinoma—Toluidine blue staining characteristics help to identify BCC nests by differentiating them from hair follicles in frozen sections. The metachromatic characteristic of TB stains the inner root sheath deep blue and highlights the surrounding stromal mucin of BCC a magenta color.18,19 In hematoxylin and eosin (H&E) stains, these 2 distinct structures can be differentiated by cleft formation around tumor nests, mitotic figures, and the lack of a fibrous sheath present in BCC tumors.20 The advantages and limitations of TB staining of BCC are presented in Table 2.

Advantages and Limitations of Toluidine Blue Staining in BCC and SCC

Humphreys et al6 suggested a noticeable difference between H&E and TB in the staining of cellular and stromal components. The nuclear detail of tumor cells was subjectively sharper and clearer with TB staining. The staining of stromal components may provide the most assistance in locating BCC islands. Mucopolysaccharide staining may be absent in H&E but stain a deep magenta with TB. Although the presence of mucopolysaccharides does not specifically indicate a tumor, it may prompt further attention and provide an indicator for sparse and infiltrative tumor cells.6 The metachromatic stromal change may indicate a narrow tumor-free margin where additional deeper sections often reveal tumor that may warrant additional resection margin in more aggressive malignancies. In particular, sclerosing/morpheaform BCCs have been shown to induce glycosaminoglycan synthesis and are highlighted more readily with TB than with H&E when compared to surrounding tissue.21 This differentiation in staining has remained a popular reason to routinely incorporate TB into the staining of infiltrative and morpheaform variants of BCC. Additionally, stromal mast cells are believed to be more abundant in the stroma of BCC and are more readily visualized in tissue specimens stained with TB, appearing as bright purple metachromatic granules. These granules are larger than normal and are increased in number.6

The margin behavior of BCC stained with TB was further characterized by Goldberg et al,8 who coined the term setting sun sign, which may be present in sequential sections of a disappearing nodule of a BCC tumor. Stroma, inflammatory infiltrate, and mast cells produce a magenta glow surrounding BCC tumors that is reminiscent of a setting sun (Figure 2). Invasive BCC is considered variable in this presentation, primarily because of zones of cell-free fluid and edema or the second area of inflammatory cells. This unique sign may benefit the inspecting Mohs surgeon by providing a clue to an underlying process that may have residual BCC tumors. The setting sun sign also may assist in identifying exact surgical margins.8

Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.
FIGURE 2. A and B, Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.

The nasal surface has a predilection for BCC.22 The skin of the nose has numerous look-alike structures to consider for complete tumor removal and avoidance of unnecessary removal. One challenge is distinguishing follicular basaloid proliferations (FBP) from BCC, a scenario that is more common on the nose.22 When TB staining was used, the sensitivity for detecting FBP reached 100% in 34 cases reviewed by Donaldson and Weber.10 None of the cases examined showed TB metachromasia surrounding FBP, thus indicating that TB can dependably identify this benign entity. Conversely, 5% (N=279) of BCCs confirmed on H&E did not exhibit surrounding TB metachromasia. This finding is concerning regarding the specificity of TB staining for BCC, but the authors of this study suggested the possibility that these exceptions were benign “simulants” (ie, trichoepithelioma) of BCC.10

The use of TB also has been shown to be statistically beneficial in Mohs training. In a single-center, single-fellow experiment, the sensitivity and specificity of using TB for BCC were extrapolated.9 Using TB as an adjunct in deep sections showed superior sensitivity to H&E alone in identifying BCC, increasing sensitivity from 96.3% to 99.7%. In a cohort of 352 BCC excisions and frozen sections, only 1 BCC was not completely excised. If H&E only had been performed, the fellow would have missed 13 residual BCC tumors.9

Bennett and Taher7 described a case in which hyaluronic acid (HA) from a filler injection was confused with the HA surrounding BCC tumor nests. They found that when TB is used as an adjunct, the HA filler is easier to differentiate from the HA surrounding the BCC tumor nests. In frozen sections stained with TB, the HA filler appeared as an amorphous, metachromatic, reddish-purple, whereas the HA surrounding the BCC tumor nests appeared as a well-defined red. These findings were less obvious in the same sections stained with H&E alone.7

 

 

Squamous Cell Carcinoma—In early investigations, the utility of TB in identifying SCC in frozen sections was thought to be limited. The description by Humphreys and colleagues6 of staining characteristics in SCC suggested that the nuclear detail that H&E provides is more easily recognized. The deep aqua nuclear staining produced with TB was considered more difficult to observe than the cytoplasmic eosinophilia of pyknotic and keratinizing cells in H&E.6

Toluidine blue may be beneficial in providing unique staining characteristics to further detail tumors that are difficult to interpret, such as spindle cell SCC and perineural invasion of aggressive SCC. In H&E, squamous cells of spindle cell SCC (scSCC) blend into the background of inflammatory cells and can be perceptibly difficult to locate. A small cohort of 3 Mohs surgeons who routinely use H&E were surveyed on their ability to detect a proven scSCC in H&E or TB by photograph.12 All 3 were able to detect the scSCC in the TB photographs, but only 2 of 3 were able to detect it in H&E photographs. All 3 surgeons agreed that TB was preferable to H&E for this tumor type. These findings suggested that TB may be superior and preferred over H&E for visualizing tumor cells of scSCC.12 The TB staining characteristics of perineural invasion of aggressive SCC have been referred to as the perineural corona sign because of the bright magenta stain that forms around affected nerves.13 Drosou et al13 suggested that TB may enhance the diagnostic accuracy for perineural SCC.

Rare Tumors—The adjunctive use of TB with H&E has been examined in rare tumors. Published reports have highlighted its use in MMS for treating MAC and PCACC. Toluidine blue exhibits staining advantages for these tumors. It may render isolated nests and perineural invasion of MAC more easily visible on frozen section.15

Although PCACC is rare, the recurrence rate is high.23 Toluidine blue has been used with MMS to ensure complete removal and higher cure rates. The metachromatic nature of TB is advantageous in staining the HA present in these tumors. Those who have reported the use of TB for PCACC prefer it to H&E for frozen sections.14

Technical Aspects—The staining time for TB-treated slides is reduced compared to H&E staining; staining can be efficiently done in frozen sections in less than 2.5 minutes using the method shown in Table 3.17 In comparison, typical H&E staining takes 9 minutes, and older TB techniques take 7 minutes.6

Rapid Toluidine Blue Staining Protocol

Conclusion

Toluidine blue may play an important and helpful role in the successful diagnosis and treatment of particular cutaneous tumors by providing additional diagnostic information. Although surgeons performing MMS will continue using the staining protocols with which they are most comfortable, adjunctive use of TB over time may provide an additional benefit at low risk for disrupting practice efficiency or workflow. Many Mohs surgeons are accustomed to using this stain, even preferring to interpret only TB-stained slides for cutaneous malignancy. Most published studies on this topic have been observational in nature, and additional controlled trials may be warranted to determine the effects on outcomes in real-world practice.

Toluidine blue (TB), a dye with metachromatic staining properties, was developed in 1856 by William Henry Perkin.1 Metachromasia is a perceptible change in the color of staining of living tissue due to the electrochemical properties of the tissue. Tissues that contain high concentrations of ionized sulfate and phosphate groups (high concentrations of free electronegative groups) form polymeric aggregates of the basic dye solution that alter the absorbed wavelengths of light.2 The function of this characteristic is to use a single dye to highlight different structures in tissue based on their relative chemical differences.3

Toluidine blue primarily was used within the dye industry until the 1960s, when it was first used in vital staining of the oral mucosa.2 Because of the tissue absorption potential, this technique was used to detect the location of oral malignancies.4 Since then, TB has progressively been used for staining fresh frozen sections in Mohs micrographic surgery (MMS). In a 2003 survey study (N=310), 16.8% of surgeons performing MMS reported using TB in their laboratory.5 We sought to systematically review the published literature describing the uses of TB in the setting of fresh frozen sections and MMS.

Methods

We conducted a systematic search of the PubMed and Cochrane databases for articles published before December 1, 2019, to identify any relevant studies in English. Electronic searches were performed using the terms toluidine blue and Mohs or Mohs micrographic surgery. We manually checked the bibliographies of the identified articles to further identify eligible studies.

Eligibility Criteria—The inclusion criteria were articles that (1) considered TB in the context of MMS, (2) were published in peer-reviewed journals, (3) were published in English, and (4) were available as full text. Systematic reviews were excluded.

Data Extraction and Outcomes—All relevant information regarding the study characteristics, including design, level of evidence, methodologic quality of evidence, pathology examined, and outcome measures, were collected by 2 independent reviewers (T.L. and A.D.) using a predetermined data sheet. The same 2 reviewers were used for all steps of the review process, data were independently obtained, and any discrepancy was introduced for a third opinion (D.H.) and agreed upon by the majority.

Quality Assessment—The level of evidence was evaluated based on the criteria of the Oxford Centre for Evidence-Based Medicine. Two reviewers (T.L. and A.D.) graded each article included in the review.

PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.
FIGURE 1. PRISMA (Preferred Reporting Items for Systematic Reviews and Meta Analyses) flow diagram.

Results

A total of 25 articles were reviewed. After the titles and abstracts were screened for relevance, 12 articles remained (Figure 1). Of these, 1 compared basal cell carcinoma (BCC) and squamous cell carcinoma (SCC), 4 were related to BCC, 3 were related to SCC, 1 was related to microcystic adnexal carcinoma (MAC), 1 was related to primary cutaneous adenoid cystic carcinoma (PCACC), and 2 were related to technical aspects of the staining process (Table 1).

Summary of Articles Published on Toluidine Blue in Mohs Micrographic Surgery

 

 

A majority of the articles included in this review were qualitative and observational in nature, describing the staining characteristics of TB. Study characteristics are summarized in Table 1.

Comment

Basal Cell Carcinoma—Toluidine blue staining characteristics help to identify BCC nests by differentiating them from hair follicles in frozen sections. The metachromatic characteristic of TB stains the inner root sheath deep blue and highlights the surrounding stromal mucin of BCC a magenta color.18,19 In hematoxylin and eosin (H&E) stains, these 2 distinct structures can be differentiated by cleft formation around tumor nests, mitotic figures, and the lack of a fibrous sheath present in BCC tumors.20 The advantages and limitations of TB staining of BCC are presented in Table 2.

Advantages and Limitations of Toluidine Blue Staining in BCC and SCC

Humphreys et al6 suggested a noticeable difference between H&E and TB in the staining of cellular and stromal components. The nuclear detail of tumor cells was subjectively sharper and clearer with TB staining. The staining of stromal components may provide the most assistance in locating BCC islands. Mucopolysaccharide staining may be absent in H&E but stain a deep magenta with TB. Although the presence of mucopolysaccharides does not specifically indicate a tumor, it may prompt further attention and provide an indicator for sparse and infiltrative tumor cells.6 The metachromatic stromal change may indicate a narrow tumor-free margin where additional deeper sections often reveal tumor that may warrant additional resection margin in more aggressive malignancies. In particular, sclerosing/morpheaform BCCs have been shown to induce glycosaminoglycan synthesis and are highlighted more readily with TB than with H&E when compared to surrounding tissue.21 This differentiation in staining has remained a popular reason to routinely incorporate TB into the staining of infiltrative and morpheaform variants of BCC. Additionally, stromal mast cells are believed to be more abundant in the stroma of BCC and are more readily visualized in tissue specimens stained with TB, appearing as bright purple metachromatic granules. These granules are larger than normal and are increased in number.6

The margin behavior of BCC stained with TB was further characterized by Goldberg et al,8 who coined the term setting sun sign, which may be present in sequential sections of a disappearing nodule of a BCC tumor. Stroma, inflammatory infiltrate, and mast cells produce a magenta glow surrounding BCC tumors that is reminiscent of a setting sun (Figure 2). Invasive BCC is considered variable in this presentation, primarily because of zones of cell-free fluid and edema or the second area of inflammatory cells. This unique sign may benefit the inspecting Mohs surgeon by providing a clue to an underlying process that may have residual BCC tumors. The setting sun sign also may assist in identifying exact surgical margins.8

Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.
FIGURE 2. A and B, Frozen sections of a basal cell carcinoma (original magnification ×100 for both). The basophilic tumor cells were surrounded by an immediate magenta zone of stroma and an inflammatory response of mast cells, lymphocytes, and fibroblasts.

The nasal surface has a predilection for BCC.22 The skin of the nose has numerous look-alike structures to consider for complete tumor removal and avoidance of unnecessary removal. One challenge is distinguishing follicular basaloid proliferations (FBP) from BCC, a scenario that is more common on the nose.22 When TB staining was used, the sensitivity for detecting FBP reached 100% in 34 cases reviewed by Donaldson and Weber.10 None of the cases examined showed TB metachromasia surrounding FBP, thus indicating that TB can dependably identify this benign entity. Conversely, 5% (N=279) of BCCs confirmed on H&E did not exhibit surrounding TB metachromasia. This finding is concerning regarding the specificity of TB staining for BCC, but the authors of this study suggested the possibility that these exceptions were benign “simulants” (ie, trichoepithelioma) of BCC.10

The use of TB also has been shown to be statistically beneficial in Mohs training. In a single-center, single-fellow experiment, the sensitivity and specificity of using TB for BCC were extrapolated.9 Using TB as an adjunct in deep sections showed superior sensitivity to H&E alone in identifying BCC, increasing sensitivity from 96.3% to 99.7%. In a cohort of 352 BCC excisions and frozen sections, only 1 BCC was not completely excised. If H&E only had been performed, the fellow would have missed 13 residual BCC tumors.9

Bennett and Taher7 described a case in which hyaluronic acid (HA) from a filler injection was confused with the HA surrounding BCC tumor nests. They found that when TB is used as an adjunct, the HA filler is easier to differentiate from the HA surrounding the BCC tumor nests. In frozen sections stained with TB, the HA filler appeared as an amorphous, metachromatic, reddish-purple, whereas the HA surrounding the BCC tumor nests appeared as a well-defined red. These findings were less obvious in the same sections stained with H&E alone.7

 

 

Squamous Cell Carcinoma—In early investigations, the utility of TB in identifying SCC in frozen sections was thought to be limited. The description by Humphreys and colleagues6 of staining characteristics in SCC suggested that the nuclear detail that H&E provides is more easily recognized. The deep aqua nuclear staining produced with TB was considered more difficult to observe than the cytoplasmic eosinophilia of pyknotic and keratinizing cells in H&E.6

Toluidine blue may be beneficial in providing unique staining characteristics to further detail tumors that are difficult to interpret, such as spindle cell SCC and perineural invasion of aggressive SCC. In H&E, squamous cells of spindle cell SCC (scSCC) blend into the background of inflammatory cells and can be perceptibly difficult to locate. A small cohort of 3 Mohs surgeons who routinely use H&E were surveyed on their ability to detect a proven scSCC in H&E or TB by photograph.12 All 3 were able to detect the scSCC in the TB photographs, but only 2 of 3 were able to detect it in H&E photographs. All 3 surgeons agreed that TB was preferable to H&E for this tumor type. These findings suggested that TB may be superior and preferred over H&E for visualizing tumor cells of scSCC.12 The TB staining characteristics of perineural invasion of aggressive SCC have been referred to as the perineural corona sign because of the bright magenta stain that forms around affected nerves.13 Drosou et al13 suggested that TB may enhance the diagnostic accuracy for perineural SCC.

Rare Tumors—The adjunctive use of TB with H&E has been examined in rare tumors. Published reports have highlighted its use in MMS for treating MAC and PCACC. Toluidine blue exhibits staining advantages for these tumors. It may render isolated nests and perineural invasion of MAC more easily visible on frozen section.15

Although PCACC is rare, the recurrence rate is high.23 Toluidine blue has been used with MMS to ensure complete removal and higher cure rates. The metachromatic nature of TB is advantageous in staining the HA present in these tumors. Those who have reported the use of TB for PCACC prefer it to H&E for frozen sections.14

Technical Aspects—The staining time for TB-treated slides is reduced compared to H&E staining; staining can be efficiently done in frozen sections in less than 2.5 minutes using the method shown in Table 3.17 In comparison, typical H&E staining takes 9 minutes, and older TB techniques take 7 minutes.6

Rapid Toluidine Blue Staining Protocol

Conclusion

Toluidine blue may play an important and helpful role in the successful diagnosis and treatment of particular cutaneous tumors by providing additional diagnostic information. Although surgeons performing MMS will continue using the staining protocols with which they are most comfortable, adjunctive use of TB over time may provide an additional benefit at low risk for disrupting practice efficiency or workflow. Many Mohs surgeons are accustomed to using this stain, even preferring to interpret only TB-stained slides for cutaneous malignancy. Most published studies on this topic have been observational in nature, and additional controlled trials may be warranted to determine the effects on outcomes in real-world practice.

References
  1. Culling CF, Allison TR. Cellular Pathology Technique. 4th ed. Butterworths; 1985.
  2. Bergeron JA, Singer M. Metachromasy: an experimental and theoretical reevaluation. J Biophys Biochem Cytol. 1958;4:433-457. doi:10.1083/jcb.4.4.433
  3. Epstein JB, Scully C, Spinelli J. Toluidine blue and Lugol’s iodine application in the assessment of oral malignant disease and lesions at risk of malignancy. J Oral Pathol Med. 1992;21:160-163. doi:10.1111/j.1600-0714.1992.tb00094.x
  4. Warnakulasuriya KA, Johnson NW. Sensitivity and specificity of OraScan (R) toluidine blue mouthrinse in the detection of oral cancer and precancer. J Oral Pathol Med. 1996;25:97-103. doi:10.1111/j.1600-0714.1996.tb00201.x
  5. Silapunt S, Peterson SR, Alcalay J, et al. Mohs tissue mapping and processing: a survey study. Dermatol Surg. 2003;29:1109-1112; discussion 1112.
  6. Humphreys TR, Nemeth A, McCrevey S, et al. A pilot study comparing toluidine blue and hematoxylin and eosin staining of basal cell and squamous cell carcinoma during Mohs surgery. Dermatol Surg. 1996;22:693-697. doi:10.1111/j.1524-4725.1996.tb00619.x
  7. Bennett R, Taher M. Restylane persistent for 23 months found during Mohs micrographic surgery: a source of confusion with hyaluronic acid surrounding basal cell carcinoma. Dermatol Surg. 2005;31:1366-1369. doi:10.1111/j.1524-4725.2005.31223
  8. Goldberg LH, Wang SQ, Kimyai-Asadi A. The setting sun sign: visualizing the margins of a basal cell carcinoma on serial frozen sections stained with toluidine blue. Dermatol Surg. 2007;33:761-763. doi:10.1111/j.1524-4725.2007.33158.x
  9. Tehrani H, May K, Morris A, et al. Does the dual use of toluidine blue and hematoxylin and eosin staining improve basal cell carcinoma detection by Mohs surgery trainees? Dermatol Surg. 2013;39:995-1000. doi:10.1111/dsu.12180
  10. Donaldson MR, Weber LA. Toluidine blue supports differentiation of folliculocentric basaloid proliferation from basal cell carcinoma on frozen sections in a small single-practice cohort. Dermatol Surg. 2017;43:1303-1306. doi:10.1097/DSS.0000000000001107
  11. Styperek AR, Goldberg LH, Goldschmidt LE, et al. Toluidine blue and hematoxylin and eosin stains are comparable in evaluating squamous cell carcinoma during Mohs. Dermatol Surg. 2016;42:1279-1284. doi:10.1097/DSS.0000000000000872
  12. Trieu D, Drosou A, Goldberg LH, et al. Detecting spindle cell squamous cell carcinomas with toluidine blue on frozen sections. Dermatol Surg. 2014;40:1259-1260. doi:10.1097/DSS.0000000000000147
  13. Drosou A, Trieu D, Goldberg LH, et al. The perineural corona sign: enhancing detection of perineural squamous cell carcinoma during Mohs micrographic surgery with toluidine blue stain. J Am Acad Dermatol. 2014;71:826-827. doi:10.1016/j.jaad.2014.04.076
  14. Chesser RS, Bertler DE, Fitzpatrick JE, et al. Primary cutaneous adenoid cystic carcinoma treated with Mohs micrographic surgery toluidine blue technique. J Dermatol Surg Oncol. 1992;18:175-176. doi:10.1111/j.1524-4725.1992.tb02794.x
  15. Wang SQ, Goldberg LH, Nemeth A. The merits of adding toluidine blue-stained slides in Mohs surgery in the treatment of a microcystic adnexal carcinoma. J Am Acad Dermatol. 2007;56:1067-1069. doi:10.1016/j.jaad.2007.01.008
  16. Chen CL, Wilson S, Afzalneia R, et al. Topical aluminum chloride and Monsel’s solution block toluidine blue staining in Mohs frozen sections: mechanism and solution. Dermatol Surg. 2019;45:1019-1025. doi:10.1097/DSS.0000000000001761
  17. Todd MM, Lee JW, Marks VJ. Rapid toluidine blue stain for Mohs’ micrographic surgery. Dermatol Surg. 2005;31:244-245. doi:10.1111/j.1524-4725.2005.31053
  18. Picoto AM, Picoto A. Technical procedures for Mohs fresh tissue surgery. J Derm Surg Oncol. 1986;12:134-138. doi:10.1111/j.1524-4725.1986.tb01442.x
  19. Sperling LC, Winton GB. The transverse anatomy of androgenic alopecia. J Derm Surg Oncol. 1990;16:1127-1133. doi:10.1111/j.1524 -4725.1990.tb00024.x
  20. Smith-Zagone MJ, Schwartz MR. Frozen section of skin specimens. Arch Pathol Lab Med. 2005;129:1536-1543. doi:10.5858/2005-129-1536-FSOSS
  21. Moy RL, Potter TS, Uitto J. Increased glycosaminoglycans production in sclerosing basal cell carcinoma–derived fibroblasts and stimulation of normal skin fibroblast glycosaminoglycans production by a cytokine-derived from sclerosing basal cell carcinoma. Dermatol Surg. 2000;26:1029-1036. doi:10.1046/j.1524-4725.2000.0260111029.x
  22. Leshin B, White WL. Folliculocentric basaloid proliferation. The bulge (der Wulst) revisited. Arch Dermatol. 1990;126:900-906. doi:10.1001/archderm.126.7.900
  23. Seab JA, Graham JH. Primary cutaneous adenoid cystic carcinoma.J Am Acad Dermatol. 1987;17:113-118. doi:10.1016/s0190 -9622(87)70182-0
References
  1. Culling CF, Allison TR. Cellular Pathology Technique. 4th ed. Butterworths; 1985.
  2. Bergeron JA, Singer M. Metachromasy: an experimental and theoretical reevaluation. J Biophys Biochem Cytol. 1958;4:433-457. doi:10.1083/jcb.4.4.433
  3. Epstein JB, Scully C, Spinelli J. Toluidine blue and Lugol’s iodine application in the assessment of oral malignant disease and lesions at risk of malignancy. J Oral Pathol Med. 1992;21:160-163. doi:10.1111/j.1600-0714.1992.tb00094.x
  4. Warnakulasuriya KA, Johnson NW. Sensitivity and specificity of OraScan (R) toluidine blue mouthrinse in the detection of oral cancer and precancer. J Oral Pathol Med. 1996;25:97-103. doi:10.1111/j.1600-0714.1996.tb00201.x
  5. Silapunt S, Peterson SR, Alcalay J, et al. Mohs tissue mapping and processing: a survey study. Dermatol Surg. 2003;29:1109-1112; discussion 1112.
  6. Humphreys TR, Nemeth A, McCrevey S, et al. A pilot study comparing toluidine blue and hematoxylin and eosin staining of basal cell and squamous cell carcinoma during Mohs surgery. Dermatol Surg. 1996;22:693-697. doi:10.1111/j.1524-4725.1996.tb00619.x
  7. Bennett R, Taher M. Restylane persistent for 23 months found during Mohs micrographic surgery: a source of confusion with hyaluronic acid surrounding basal cell carcinoma. Dermatol Surg. 2005;31:1366-1369. doi:10.1111/j.1524-4725.2005.31223
  8. Goldberg LH, Wang SQ, Kimyai-Asadi A. The setting sun sign: visualizing the margins of a basal cell carcinoma on serial frozen sections stained with toluidine blue. Dermatol Surg. 2007;33:761-763. doi:10.1111/j.1524-4725.2007.33158.x
  9. Tehrani H, May K, Morris A, et al. Does the dual use of toluidine blue and hematoxylin and eosin staining improve basal cell carcinoma detection by Mohs surgery trainees? Dermatol Surg. 2013;39:995-1000. doi:10.1111/dsu.12180
  10. Donaldson MR, Weber LA. Toluidine blue supports differentiation of folliculocentric basaloid proliferation from basal cell carcinoma on frozen sections in a small single-practice cohort. Dermatol Surg. 2017;43:1303-1306. doi:10.1097/DSS.0000000000001107
  11. Styperek AR, Goldberg LH, Goldschmidt LE, et al. Toluidine blue and hematoxylin and eosin stains are comparable in evaluating squamous cell carcinoma during Mohs. Dermatol Surg. 2016;42:1279-1284. doi:10.1097/DSS.0000000000000872
  12. Trieu D, Drosou A, Goldberg LH, et al. Detecting spindle cell squamous cell carcinomas with toluidine blue on frozen sections. Dermatol Surg. 2014;40:1259-1260. doi:10.1097/DSS.0000000000000147
  13. Drosou A, Trieu D, Goldberg LH, et al. The perineural corona sign: enhancing detection of perineural squamous cell carcinoma during Mohs micrographic surgery with toluidine blue stain. J Am Acad Dermatol. 2014;71:826-827. doi:10.1016/j.jaad.2014.04.076
  14. Chesser RS, Bertler DE, Fitzpatrick JE, et al. Primary cutaneous adenoid cystic carcinoma treated with Mohs micrographic surgery toluidine blue technique. J Dermatol Surg Oncol. 1992;18:175-176. doi:10.1111/j.1524-4725.1992.tb02794.x
  15. Wang SQ, Goldberg LH, Nemeth A. The merits of adding toluidine blue-stained slides in Mohs surgery in the treatment of a microcystic adnexal carcinoma. J Am Acad Dermatol. 2007;56:1067-1069. doi:10.1016/j.jaad.2007.01.008
  16. Chen CL, Wilson S, Afzalneia R, et al. Topical aluminum chloride and Monsel’s solution block toluidine blue staining in Mohs frozen sections: mechanism and solution. Dermatol Surg. 2019;45:1019-1025. doi:10.1097/DSS.0000000000001761
  17. Todd MM, Lee JW, Marks VJ. Rapid toluidine blue stain for Mohs’ micrographic surgery. Dermatol Surg. 2005;31:244-245. doi:10.1111/j.1524-4725.2005.31053
  18. Picoto AM, Picoto A. Technical procedures for Mohs fresh tissue surgery. J Derm Surg Oncol. 1986;12:134-138. doi:10.1111/j.1524-4725.1986.tb01442.x
  19. Sperling LC, Winton GB. The transverse anatomy of androgenic alopecia. J Derm Surg Oncol. 1990;16:1127-1133. doi:10.1111/j.1524 -4725.1990.tb00024.x
  20. Smith-Zagone MJ, Schwartz MR. Frozen section of skin specimens. Arch Pathol Lab Med. 2005;129:1536-1543. doi:10.5858/2005-129-1536-FSOSS
  21. Moy RL, Potter TS, Uitto J. Increased glycosaminoglycans production in sclerosing basal cell carcinoma–derived fibroblasts and stimulation of normal skin fibroblast glycosaminoglycans production by a cytokine-derived from sclerosing basal cell carcinoma. Dermatol Surg. 2000;26:1029-1036. doi:10.1046/j.1524-4725.2000.0260111029.x
  22. Leshin B, White WL. Folliculocentric basaloid proliferation. The bulge (der Wulst) revisited. Arch Dermatol. 1990;126:900-906. doi:10.1001/archderm.126.7.900
  23. Seab JA, Graham JH. Primary cutaneous adenoid cystic carcinoma.J Am Acad Dermatol. 1987;17:113-118. doi:10.1016/s0190 -9622(87)70182-0
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E6-E11
Page Number
E6-E11
Publications
Publications
Topics
Article Type
Display Headline
The Role of Toluidine Blue in Mohs Micrographic Surgery: A Systematic Review
Display Headline
The Role of Toluidine Blue in Mohs Micrographic Surgery: A Systematic Review
Sections
Inside the Article

Practice Points

  • Toluidine blue (TB) staining can be integrated into Mohs micrographic surgery (MMS) for enhanced diagnosis of cutaneous tumors. Its metachromatic properties can aid in differentiating tumor cells from surrounding tissues, especially in basal cell carcinomas and squamous cell carcinomas.
  • It is important to develop expertise in interpreting TB-stained sections, as it may offer clearer visualization of nuclear details and stromal components, potentially leading to more accurate diagnosis and effective tumor margin identification.
  • Toluidine blue staining can be incorporated into routine MMS practice considering its quick staining process and low disruption to workflow. This can potentially improve diagnostic efficiency without significantly lengthening surgery time.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Reactive Angioendotheliomatosis Following Ad26.COV2.S Vaccination

Article Type
Changed
Thu, 12/21/2023 - 07:50
Display Headline
Reactive Angioendotheliomatosis Following Ad26.COV2.S Vaccination

To the Editor:

Reactive angioendotheliomatosis (RAE) is a rare self-limited cutaneous vascular proliferation of endothelial cells within blood vessels that manifests clinically as infiltrated red-blue patches and plaques with purpura that can progress to occlude vascular lumina. The etiology of RAE is mostly idiopathic; however, the disorder typically occurs in association with a range of systemic diseases, including infection, cryoglobulinemia, leukemia, antiphospholipid syndrome, peripheral vascular disease, and arteriovenous fistula. Histopathologic examination of these lesions shows marked proliferation of endothelial cells, including occlusion of the lumen of blood vessels over wide areas.

After ruling out malignancy, treatment of RAE focuses on targeting the underlying cause or disease, if any is present; 75% of reported cases occur in association with systemic disease.1 Onset can occur at any age without predilection for sex. Reactive angioendotheliomatosis commonly manifests on the extremities but may occur on the head and neck in rare instances.2

The rarity of the condition and its poorly defined clinical characteristics make it difficult to develop a treatment plan. There are no standardized treatment guidelines for the reactive form of angiomatosis. We report a case of RAE that developed 2 weeks after vaccination with the Ad26.COV2.S vaccine (Johnson & Johnson Innovative Medicine [formerly Janssen Pharmaceutical Companies of Johnson & Johnson]) that improved following 2 weeks of treatment with a topical corticosteroid and an oral antihistamine.

A 58-year-old man presented to an outpatient dermatology clinic with pruritus and occasional paresthesia associated with a rash over the left arm of 1 month’s duration. The patient suspected that the rash may have formed secondary to the bite of oak mites on the arms and chest while he was carrying milled wood. Further inquiry into the patient’s history revealed that he received the Ad26.COV2.S vaccine 2 weeks prior to the appearance of the rash. He denied mechanical trauma. His medical history included hypercholesterolemia and a mild COVID-19 infection 8 months prior to the appearance of the rash that did not require hospitalization. He denied fever or chills during the 2 weeks following vaccination. The pruritus was minimally relieved for short periods with over-the-counter calamine lotion. The patient’s medication regimen included daily pravastatin and loratadine at the time of the initial visit. He used acetaminophen as needed for knee pain.

A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.
FIGURE 1. A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.

Physical examination revealed palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula (Figure 1A), left anterolateral shoulder, left lateral volar forearm, and thenar eminence of the left hand (Figure 1B). Notably, the entire right arm, conjunctivae, tongue, lips, and bilateral fingernails were clear. Three 4-mm punch biopsies were performed at the initial presentation: 1 perilesional biopsy for direct immunofluorescence testing and 2 lesional biopsies for routine histologic evaluation. An extensive serologic workup failed to reveal abnormalities. An activated partial thromboplastin time, dilute Russell viper venom time, serum protein electrophoresis, and levels of rheumatoid factor and angiotensin-converting enzyme were within reference range. Anticardiolipin antibodies IgA, IgM, and IgG were negative. A cryoglobulin test was negative.

Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).
FIGURE 2. Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).

Histopathology revealed a proliferation of irregularly shaped vascular spaces with plump endothelium in the papillary dermis (Figure 2). Scattered leukocyte common antigen-positive lymphocytes were noted within lesions. The epidermis appeared normal, without evidence of spongiosis or alteration of the stratum corneum. Immunohistochemical studies of the perilesional skin biopsy revealed positivity for CD31 and D2-40 (Figure 3). Specimens were negative for CD20 and human herpesvirus 8. Direct immunofluorescence of the perilesional biopsy was negative.

Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).
FIGURE 3. Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).

A diagnosis of RAE was made based on clinical and histologic findings. Treatment with triamcinolone ointment 0.1% twice daily and oral cetirizine 10 mg twice daily was initiated. Re-evaluation 2 weeks later revealed notable improvement in the affected areas, including decreased edema, improvement of the purpura, and absence of pruritus. The patient noted no further spread or blister formation while the active areas were being treated with the topical steroid. The treatment regimen was modified to triamcinolone ointment 0.1% once daily, and cetirizine was discontinued. At 3-month follow-up, active areas had completely resolved (Figure 4) and triamcinolone was discontinued. To date, the patient has not had recurrence of symptoms and remains healthy.

At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.
FIGURE 4. A and B, At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.

 

 

Gottron and Nikolowski3 reported the first case of RAE in an adult patient who presented with purpuric patches secondary to skin infarction. Current definitions use the umbrella term cutaneous reactive angiomatosis to cover 3 major subtypes: reactive angioendotheliomatosis, diffuse dermal angioendotheliomatosis, and acroangiodermatitis (pseudo-Kaposi sarcoma [KS]). The manifestation of these subgroups is clinically similar, and they must be differentiated through histologic evaluation.4

Reactive angioendotheliomatosis has an unknown pathogenesis and is poorly defined clinically. The exact pathophysiology is unknown but likely is linked to vaso-occlusion and hypoxia.1 A PubMed search of articles indexed for MEDLINE, as well as a review of Science Direct, Google Scholar, and Cochrane Library, using the terms reactive angioendotheliomatosis, COVID, vaccine, Ad26.COV2.S, and RAE in any combination revealed no prior cases of RAE in association with Ad26.COV2.S vaccination.

By the late 1980s, systemic angioendotheliomatosis was segregated into 2 distinct entities: malignant and reactive.4 The differential diagnosis of malignant systemic angioendotheliomatosis includes KS and angiosarcoma; nonmalignant causes are the variants of cutaneous reactive angiomatosis. It is important to rule out KS because of its malignant and deceptive nature. It is unknown if KS originates in blood vessels or lymphatic endothelial cells; however, evidence is strongly in favor of blood vessel origin using CD31 and CD34 endothelial markers.5 CD34 positivity is more reliable than CD31 in diagnosing KS, but the absence of both markers does not offer enough evidence to rule out KS on its own.6

In our patient, histopathology revealed cells positive for CD31 and D2-40; the latter is a lymphatic endothelial cell marker that stains the endothelium of lymphatic channels but not blood vessels.7 Positive D2-40 can be indicative of KS and non-KS lesions, each with a distinct staining pattern. D2-40 staining on non-KS lesions is confined to lymphatic vessels, as it was in our patient; in contrast, spindle-shaped cells also will be stained in KS lesions.8

Another cell marker, CD20, is a B cell–specific protein that can be measured to help diagnose malignant diseases such as B-cell lymphoma and leukemia. Human herpesvirus 8 (also known as KS-associated herpesvirus) is the infectious cause of KS and traditionally has been detected using methods such as the polymerase chain reaction.9,10

Most cases of RAE are idiopathic and occur in association with systemic disease, which was not the case in our patient. We speculated that his reaction was most likely triggered by vascular transfection of endothelial cells secondary to Ad26.COV2.S vaccination. Alternatively, vaccination may have caused vascular occlusion, though the lack of cyanosis, nail changes, and route of inoculant make this less likely.

All approved COVID-19 vaccines are designed solely for intramuscular injection. In comparison to other types of tissue, muscles have superior vascularity, allowing for enhanced mobilization of compounds, which results in faster systemic circulation.11 Alternative methods of injection, including intravascular, subcutaneous, and intradermal, may lead to decreased efficacy or adverse events, or both.

 

 

Prior cases of RAE have been treated with laser therapy, topical or systemic corticosteroids, excisional removal, or topical β-blockers, such as timolol.12β-Blocking agents act on β-adrenergic receptors on endothelial cells to inhibit angiogenesis by reducing release of blood vessel growth-signaling molecules and triggering apoptosis. In this patient, topical steroids and oral antihistamines were sufficient treatment.

Vaccine-related adverse events have been reported but remain rare. The benefits of Ad26.COV2.S vaccination for protection against COVID-19 outweigh the extremely low risk for adverse events.13 For that reason, the Centers for Disease Control and Prevention recommends a booster for individuals who are eligible to maximize protection. Intramuscular injection of Ad26.COV2.S resulted in a lower incidence of moderate to severe COVID-19 cases in all age groups vs the placebo group. Hypersensitivity adverse events were reported in 0.4% of Ad26.COV2.S-vaccinated patients vs 0.4% of patients who received a placebo; the more common reactions were nonanaphylactic.13

There have been 12 reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, which sparked nationwide controversy over the safety of the Ad26.COV2.S vaccine.14 After further investigation into those reports, the US Food and Drug Administration and the Centers for Disease Control and Prevention concluded that the benefits of the Ad26.COV2.S vaccine outweigh the low risk for associated thrombosis.15

Although adverse reactions are rare, it is important that health care providers take proper safety measures before and while administering any COVID-19 vaccine. Patients should be screened for contraindications to the COVID-19 vaccine to mitigate adverse effects seen in the small percentage of patients who may need to take alternative precautions.

The broad tissue tropism and high transmissibility of SARS-CoV-2 are the main contributors to its infection having reached pandemic scale. The spike (S) protein on SARS-CoV-2 binds to ACE2, the most thoroughly studied SARS-CoV-2 receptor, which is found in a range of tissues, including arterial endothelial cells, leading to its transfection. Several studies have proposed that expression of the S protein causes endothelial dysfunction through cytokine release, activation of complement, and ultimately microvascular occlusion.16

Recent developments in the use of viral-like particles, such as vesicular stomatitis virus, may mitigate future cases of RAE that are associated with endothelial cell transfection. Vesicular stomatitis virus is a popular model virus for research applications due to its glycoprotein and matrix protein contributing to its broad tropism. Recent efforts to alter these proteins have successfully limited the broad tropism of vesicular stomatitis virus.17

The SARS-CoV-2 virus must be handled in a Biosafety Level 3 laboratory. Conversely, pseudoviruses can be handled in lower containment facilities due to their safe and efficacious nature, offering an avenue to expedite vaccine development against many viral outbreaks, including SARS-CoV-2.18

 

 

An increasing number of cutaneous manifestations have been associated with COVID-19 infection and vaccination. Eruptive pseudoangiomatosis, a rare self-limiting exanthem, has been reported in association with ­COVID-19 vaccination.19 Eruptive pseudoangiomatosis manifests as erythematous blanchable papules that resemble angiomas, typically in a widespread distribution. Eruptive pseudoangiomatosis has striking similarities to RAE histologically; both manifest as dilated dermal blood vessels with plump endothelial cells.

Our case is unique because of the vasculitic palpable nature of the lesions, which were localized to the left arm. Eruptive pseudoangiomatosis formation after COVID-19 infection or SARS-CoV-2 vaccination may suggest alteration of ACE2 by binding of S protein.20 Such alteration of the ACE2 pathway would lead to inflammation of angiotensin II, causing proliferation of endothelial cells in the formation of angiomalike lesions. This hypothesis suggests a paraviral eruption secondary to an immunologic reaction, not a classical virtual eruption from direct contact of the virus on blood vessels. Although EPA and RAE are harmless and self-limiting, these reports will spread awareness of the increasing number of skin manifestations related to COVID-19 and SARS-CoV-2 virus vaccination.

Acknowledgment—Thoughtful insights and comments on this manuscript were provided by Christine J. Ko, MD (New Haven, Connecticut); Christine L. Egan, MD (Glen Mills, Pennsylvania); Howard A. Bueller, MD (Delray Beach, Florida); and Juan Pablo Robles, PhD (Juriquilla, Mexico).

References
  1. McMenamin ME, Fletcher CDM. Reactive angioendotheliomatosis: a study of 15 cases demonstrating a wide clinicopathologic spectrum. Am J Surg Pathol. 2002;26:686-697. doi:10.1097/00000478-200206000-00001
  2. Khan S, Pujani M, Jetley S, et al. Angiomatosis: a rare vascular proliferation of head and neck region. J Cutan Aesthet Surg. 2015;8:108-110. doi:10.4103/0974-2077.158448
  3. Gottron HA, Nikolowski W. Extrarenal Lohlein focal nephritis of the skin in endocarditis. Arch Klin Exp Dermatol. 1958;207:156-176.
  4. Cooper PH. Angioendotheliomatosis: two separate diseases. J Cutan Pathol. 1988;15:259. doi:10.1111/j.1600-0560.1988.tb00556.x
  5. Cancian L, Hansen A, Boshoff C. Cellular origin of Kaposi’s sarcoma and Kaposi’s sarcoma-associated herpesvirus-induced cell reprogramming. Trends Cell Biol. Sep 2013;23:421-32. doi:10.1016/j.tcb.2013.04.001
  6. Russell Jones R, Orchard G, Zelger B, et al. Immunostaining for CD31 and CD34 in Kaposi sarcoma. J Clin Pathol. 1995;48:1011-1016. doi:10.1136/jcp.48.11.1011
  7. Kahn HJ, Bailey D, Marks A. Monoclonal antibody D2-40, a new marker of lymphatic endothelium, reacts with Kaposi’s sarcoma and a subset of angiosarcomas. Mod Pathol. 2002;15:434-440. doi:10.1038/modpathol.3880543
  8. Genedy RM, Hamza AM, Abdel Latef AA, et al. Sensitivity and specificity of D2-40 in differentiating Kaposi sarcoma from its mimickers. J Egyptian Womens Dermatolog Soc. 2021;18:67-74. doi:10.4103/jewd.jewd_61_20
  9. Mesri EA, Cesarman E, Boshoff C. Kaposi’s sarcoma and its associated herpesvirus. Nat Rev Cancer. 2010;10:707-719. doi:10.1038/nrc2888
  10. Patel RM, Goldblum JR, Hsi ED. Immunohistochemical detection of human herpes virus-8 latent nuclear antigen-1 is useful in the diagnosis of Kaposi sarcoma. Mod Pathol. 2004;17:456-460. doi:10.1038/modpathol.3800061
  11. Zuckerman JN. The importance of injecting vaccines into muscle. Different patients need different needle sizes. BMJ. 2000;321:1237-1238. doi:10.1136/bmj.321.7271.1237
  12. Bhatia R, Hazarika N, Chandrasekaran D, et al. Treatment of posttraumatic reactive angioendotheliomatosis with topical timolol maleate. JAMA Dermatol. 2021;157:1002-1004. doi:10.1001/jamadermatol.2021.1770
  13. Sadoff J, Gray G, Vandebosch A, et al; ENSEMBLE Study Group. Safety and efficacy of single-dose Ad26.COV2.S vaccine against Covid-19. N Engl J Med. 2021;384:2187-2201. doi:10.1056/NEJMoa2101544
  14. See I, Su JR, Lale A, et al. US case reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, March 2 to April 21, 2021. JAMA. 2021;325:2448-2456. doi:10.1001/jama.2021.7517
  15. Berry CT, Eliliwi M, Gallagher S, et al. Cutaneous small vessel vasculitis following single-dose Janssen Ad26.COV2.S vaccination. JAAD Case Rep. 2021;15:11-14. doi:10.1016/j.jdcr.2021.07.002
  16. Flaumenhaft R, Enjyoji K, Schmaier AA. Vasculopathy in COVID-19. Blood. 2022;140:222-235. doi:10.1182/blood.2021012250
  17. Hastie E, Cataldi M, Marriott I, et al. Understanding and altering cell tropism of vesicular stomatitis virus. Virus Res. 2013;176:16-32. doi:10.1016/j.virusres.2013.06.003
  18. Xiong H-L, Wu Y-T, Cao J-L, et al. Robust neutralization assay based on SARS-CoV-2 S-protein-bearing vesicular stomatitis virus (VSV) pseudovirus and ACE2-overexpressing BHK21 cells. Emerg Microbes Infect. 2020;9:2105-2113. doi:10.1080/22221751.2020.1815589
  19. Mohta A, Jain SK, Mehta RD, et al. Development of eruptive pseudoangiomatosis following COVID-19 immunization – apropos of 5 cases. J Eur Acad Dermatol Venereol. 2021;35:e722-e725. doi:10.1111/jdv.17499
  20. Angeli F, Spanevello A, Reboldi G, et al. SARS-CoV-2 vaccines: lights and shadows. Eur J Intern Med. 2021;88:1-8. doi:10.1016/j.ejim.2021.04.019
Article PDF
Author and Disclosure Information

Collin Faulkner is from the State University of New York at Buffalo. Dr. Jabbour is from the State University of New York Upstate Medical University, Syracuse. Dr. Kanik is from CBLPath, Rye Brook, New York. Dr. Schoeneck is from FamilyCare Medical Group, Camillus, New York. Dr. Tangoren is from I. A. Tangoren, MD, PLLC, Dermatology & Dermatologic Surgery, Syracuse.

The authors report no conflict of interest.

Correspondence: Austin J. Jabbour, MD, 850 Republican St, Seattle WA 98109 (austin.jabbour@gmail.com).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E20-E23
Sections
Author and Disclosure Information

Collin Faulkner is from the State University of New York at Buffalo. Dr. Jabbour is from the State University of New York Upstate Medical University, Syracuse. Dr. Kanik is from CBLPath, Rye Brook, New York. Dr. Schoeneck is from FamilyCare Medical Group, Camillus, New York. Dr. Tangoren is from I. A. Tangoren, MD, PLLC, Dermatology & Dermatologic Surgery, Syracuse.

The authors report no conflict of interest.

Correspondence: Austin J. Jabbour, MD, 850 Republican St, Seattle WA 98109 (austin.jabbour@gmail.com).

Author and Disclosure Information

Collin Faulkner is from the State University of New York at Buffalo. Dr. Jabbour is from the State University of New York Upstate Medical University, Syracuse. Dr. Kanik is from CBLPath, Rye Brook, New York. Dr. Schoeneck is from FamilyCare Medical Group, Camillus, New York. Dr. Tangoren is from I. A. Tangoren, MD, PLLC, Dermatology & Dermatologic Surgery, Syracuse.

The authors report no conflict of interest.

Correspondence: Austin J. Jabbour, MD, 850 Republican St, Seattle WA 98109 (austin.jabbour@gmail.com).

Article PDF
Article PDF

To the Editor:

Reactive angioendotheliomatosis (RAE) is a rare self-limited cutaneous vascular proliferation of endothelial cells within blood vessels that manifests clinically as infiltrated red-blue patches and plaques with purpura that can progress to occlude vascular lumina. The etiology of RAE is mostly idiopathic; however, the disorder typically occurs in association with a range of systemic diseases, including infection, cryoglobulinemia, leukemia, antiphospholipid syndrome, peripheral vascular disease, and arteriovenous fistula. Histopathologic examination of these lesions shows marked proliferation of endothelial cells, including occlusion of the lumen of blood vessels over wide areas.

After ruling out malignancy, treatment of RAE focuses on targeting the underlying cause or disease, if any is present; 75% of reported cases occur in association with systemic disease.1 Onset can occur at any age without predilection for sex. Reactive angioendotheliomatosis commonly manifests on the extremities but may occur on the head and neck in rare instances.2

The rarity of the condition and its poorly defined clinical characteristics make it difficult to develop a treatment plan. There are no standardized treatment guidelines for the reactive form of angiomatosis. We report a case of RAE that developed 2 weeks after vaccination with the Ad26.COV2.S vaccine (Johnson & Johnson Innovative Medicine [formerly Janssen Pharmaceutical Companies of Johnson & Johnson]) that improved following 2 weeks of treatment with a topical corticosteroid and an oral antihistamine.

A 58-year-old man presented to an outpatient dermatology clinic with pruritus and occasional paresthesia associated with a rash over the left arm of 1 month’s duration. The patient suspected that the rash may have formed secondary to the bite of oak mites on the arms and chest while he was carrying milled wood. Further inquiry into the patient’s history revealed that he received the Ad26.COV2.S vaccine 2 weeks prior to the appearance of the rash. He denied mechanical trauma. His medical history included hypercholesterolemia and a mild COVID-19 infection 8 months prior to the appearance of the rash that did not require hospitalization. He denied fever or chills during the 2 weeks following vaccination. The pruritus was minimally relieved for short periods with over-the-counter calamine lotion. The patient’s medication regimen included daily pravastatin and loratadine at the time of the initial visit. He used acetaminophen as needed for knee pain.

A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.
FIGURE 1. A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.

Physical examination revealed palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula (Figure 1A), left anterolateral shoulder, left lateral volar forearm, and thenar eminence of the left hand (Figure 1B). Notably, the entire right arm, conjunctivae, tongue, lips, and bilateral fingernails were clear. Three 4-mm punch biopsies were performed at the initial presentation: 1 perilesional biopsy for direct immunofluorescence testing and 2 lesional biopsies for routine histologic evaluation. An extensive serologic workup failed to reveal abnormalities. An activated partial thromboplastin time, dilute Russell viper venom time, serum protein electrophoresis, and levels of rheumatoid factor and angiotensin-converting enzyme were within reference range. Anticardiolipin antibodies IgA, IgM, and IgG were negative. A cryoglobulin test was negative.

Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).
FIGURE 2. Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).

Histopathology revealed a proliferation of irregularly shaped vascular spaces with plump endothelium in the papillary dermis (Figure 2). Scattered leukocyte common antigen-positive lymphocytes were noted within lesions. The epidermis appeared normal, without evidence of spongiosis or alteration of the stratum corneum. Immunohistochemical studies of the perilesional skin biopsy revealed positivity for CD31 and D2-40 (Figure 3). Specimens were negative for CD20 and human herpesvirus 8. Direct immunofluorescence of the perilesional biopsy was negative.

Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).
FIGURE 3. Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).

A diagnosis of RAE was made based on clinical and histologic findings. Treatment with triamcinolone ointment 0.1% twice daily and oral cetirizine 10 mg twice daily was initiated. Re-evaluation 2 weeks later revealed notable improvement in the affected areas, including decreased edema, improvement of the purpura, and absence of pruritus. The patient noted no further spread or blister formation while the active areas were being treated with the topical steroid. The treatment regimen was modified to triamcinolone ointment 0.1% once daily, and cetirizine was discontinued. At 3-month follow-up, active areas had completely resolved (Figure 4) and triamcinolone was discontinued. To date, the patient has not had recurrence of symptoms and remains healthy.

At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.
FIGURE 4. A and B, At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.

 

 

Gottron and Nikolowski3 reported the first case of RAE in an adult patient who presented with purpuric patches secondary to skin infarction. Current definitions use the umbrella term cutaneous reactive angiomatosis to cover 3 major subtypes: reactive angioendotheliomatosis, diffuse dermal angioendotheliomatosis, and acroangiodermatitis (pseudo-Kaposi sarcoma [KS]). The manifestation of these subgroups is clinically similar, and they must be differentiated through histologic evaluation.4

Reactive angioendotheliomatosis has an unknown pathogenesis and is poorly defined clinically. The exact pathophysiology is unknown but likely is linked to vaso-occlusion and hypoxia.1 A PubMed search of articles indexed for MEDLINE, as well as a review of Science Direct, Google Scholar, and Cochrane Library, using the terms reactive angioendotheliomatosis, COVID, vaccine, Ad26.COV2.S, and RAE in any combination revealed no prior cases of RAE in association with Ad26.COV2.S vaccination.

By the late 1980s, systemic angioendotheliomatosis was segregated into 2 distinct entities: malignant and reactive.4 The differential diagnosis of malignant systemic angioendotheliomatosis includes KS and angiosarcoma; nonmalignant causes are the variants of cutaneous reactive angiomatosis. It is important to rule out KS because of its malignant and deceptive nature. It is unknown if KS originates in blood vessels or lymphatic endothelial cells; however, evidence is strongly in favor of blood vessel origin using CD31 and CD34 endothelial markers.5 CD34 positivity is more reliable than CD31 in diagnosing KS, but the absence of both markers does not offer enough evidence to rule out KS on its own.6

In our patient, histopathology revealed cells positive for CD31 and D2-40; the latter is a lymphatic endothelial cell marker that stains the endothelium of lymphatic channels but not blood vessels.7 Positive D2-40 can be indicative of KS and non-KS lesions, each with a distinct staining pattern. D2-40 staining on non-KS lesions is confined to lymphatic vessels, as it was in our patient; in contrast, spindle-shaped cells also will be stained in KS lesions.8

Another cell marker, CD20, is a B cell–specific protein that can be measured to help diagnose malignant diseases such as B-cell lymphoma and leukemia. Human herpesvirus 8 (also known as KS-associated herpesvirus) is the infectious cause of KS and traditionally has been detected using methods such as the polymerase chain reaction.9,10

Most cases of RAE are idiopathic and occur in association with systemic disease, which was not the case in our patient. We speculated that his reaction was most likely triggered by vascular transfection of endothelial cells secondary to Ad26.COV2.S vaccination. Alternatively, vaccination may have caused vascular occlusion, though the lack of cyanosis, nail changes, and route of inoculant make this less likely.

All approved COVID-19 vaccines are designed solely for intramuscular injection. In comparison to other types of tissue, muscles have superior vascularity, allowing for enhanced mobilization of compounds, which results in faster systemic circulation.11 Alternative methods of injection, including intravascular, subcutaneous, and intradermal, may lead to decreased efficacy or adverse events, or both.

 

 

Prior cases of RAE have been treated with laser therapy, topical or systemic corticosteroids, excisional removal, or topical β-blockers, such as timolol.12β-Blocking agents act on β-adrenergic receptors on endothelial cells to inhibit angiogenesis by reducing release of blood vessel growth-signaling molecules and triggering apoptosis. In this patient, topical steroids and oral antihistamines were sufficient treatment.

Vaccine-related adverse events have been reported but remain rare. The benefits of Ad26.COV2.S vaccination for protection against COVID-19 outweigh the extremely low risk for adverse events.13 For that reason, the Centers for Disease Control and Prevention recommends a booster for individuals who are eligible to maximize protection. Intramuscular injection of Ad26.COV2.S resulted in a lower incidence of moderate to severe COVID-19 cases in all age groups vs the placebo group. Hypersensitivity adverse events were reported in 0.4% of Ad26.COV2.S-vaccinated patients vs 0.4% of patients who received a placebo; the more common reactions were nonanaphylactic.13

There have been 12 reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, which sparked nationwide controversy over the safety of the Ad26.COV2.S vaccine.14 After further investigation into those reports, the US Food and Drug Administration and the Centers for Disease Control and Prevention concluded that the benefits of the Ad26.COV2.S vaccine outweigh the low risk for associated thrombosis.15

Although adverse reactions are rare, it is important that health care providers take proper safety measures before and while administering any COVID-19 vaccine. Patients should be screened for contraindications to the COVID-19 vaccine to mitigate adverse effects seen in the small percentage of patients who may need to take alternative precautions.

The broad tissue tropism and high transmissibility of SARS-CoV-2 are the main contributors to its infection having reached pandemic scale. The spike (S) protein on SARS-CoV-2 binds to ACE2, the most thoroughly studied SARS-CoV-2 receptor, which is found in a range of tissues, including arterial endothelial cells, leading to its transfection. Several studies have proposed that expression of the S protein causes endothelial dysfunction through cytokine release, activation of complement, and ultimately microvascular occlusion.16

Recent developments in the use of viral-like particles, such as vesicular stomatitis virus, may mitigate future cases of RAE that are associated with endothelial cell transfection. Vesicular stomatitis virus is a popular model virus for research applications due to its glycoprotein and matrix protein contributing to its broad tropism. Recent efforts to alter these proteins have successfully limited the broad tropism of vesicular stomatitis virus.17

The SARS-CoV-2 virus must be handled in a Biosafety Level 3 laboratory. Conversely, pseudoviruses can be handled in lower containment facilities due to their safe and efficacious nature, offering an avenue to expedite vaccine development against many viral outbreaks, including SARS-CoV-2.18

 

 

An increasing number of cutaneous manifestations have been associated with COVID-19 infection and vaccination. Eruptive pseudoangiomatosis, a rare self-limiting exanthem, has been reported in association with ­COVID-19 vaccination.19 Eruptive pseudoangiomatosis manifests as erythematous blanchable papules that resemble angiomas, typically in a widespread distribution. Eruptive pseudoangiomatosis has striking similarities to RAE histologically; both manifest as dilated dermal blood vessels with plump endothelial cells.

Our case is unique because of the vasculitic palpable nature of the lesions, which were localized to the left arm. Eruptive pseudoangiomatosis formation after COVID-19 infection or SARS-CoV-2 vaccination may suggest alteration of ACE2 by binding of S protein.20 Such alteration of the ACE2 pathway would lead to inflammation of angiotensin II, causing proliferation of endothelial cells in the formation of angiomalike lesions. This hypothesis suggests a paraviral eruption secondary to an immunologic reaction, not a classical virtual eruption from direct contact of the virus on blood vessels. Although EPA and RAE are harmless and self-limiting, these reports will spread awareness of the increasing number of skin manifestations related to COVID-19 and SARS-CoV-2 virus vaccination.

Acknowledgment—Thoughtful insights and comments on this manuscript were provided by Christine J. Ko, MD (New Haven, Connecticut); Christine L. Egan, MD (Glen Mills, Pennsylvania); Howard A. Bueller, MD (Delray Beach, Florida); and Juan Pablo Robles, PhD (Juriquilla, Mexico).

To the Editor:

Reactive angioendotheliomatosis (RAE) is a rare self-limited cutaneous vascular proliferation of endothelial cells within blood vessels that manifests clinically as infiltrated red-blue patches and plaques with purpura that can progress to occlude vascular lumina. The etiology of RAE is mostly idiopathic; however, the disorder typically occurs in association with a range of systemic diseases, including infection, cryoglobulinemia, leukemia, antiphospholipid syndrome, peripheral vascular disease, and arteriovenous fistula. Histopathologic examination of these lesions shows marked proliferation of endothelial cells, including occlusion of the lumen of blood vessels over wide areas.

After ruling out malignancy, treatment of RAE focuses on targeting the underlying cause or disease, if any is present; 75% of reported cases occur in association with systemic disease.1 Onset can occur at any age without predilection for sex. Reactive angioendotheliomatosis commonly manifests on the extremities but may occur on the head and neck in rare instances.2

The rarity of the condition and its poorly defined clinical characteristics make it difficult to develop a treatment plan. There are no standardized treatment guidelines for the reactive form of angiomatosis. We report a case of RAE that developed 2 weeks after vaccination with the Ad26.COV2.S vaccine (Johnson & Johnson Innovative Medicine [formerly Janssen Pharmaceutical Companies of Johnson & Johnson]) that improved following 2 weeks of treatment with a topical corticosteroid and an oral antihistamine.

A 58-year-old man presented to an outpatient dermatology clinic with pruritus and occasional paresthesia associated with a rash over the left arm of 1 month’s duration. The patient suspected that the rash may have formed secondary to the bite of oak mites on the arms and chest while he was carrying milled wood. Further inquiry into the patient’s history revealed that he received the Ad26.COV2.S vaccine 2 weeks prior to the appearance of the rash. He denied mechanical trauma. His medical history included hypercholesterolemia and a mild COVID-19 infection 8 months prior to the appearance of the rash that did not require hospitalization. He denied fever or chills during the 2 weeks following vaccination. The pruritus was minimally relieved for short periods with over-the-counter calamine lotion. The patient’s medication regimen included daily pravastatin and loratadine at the time of the initial visit. He used acetaminophen as needed for knee pain.

A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.
FIGURE 1. A, Reactive angioendotheliomatosis with palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula. B, Similar findings were seen on the thenar eminence of the left hand and left lateral volar forearm.

Physical examination revealed palpable purpura in a dermatomal distribution with nonpitting edema over the left scapula (Figure 1A), left anterolateral shoulder, left lateral volar forearm, and thenar eminence of the left hand (Figure 1B). Notably, the entire right arm, conjunctivae, tongue, lips, and bilateral fingernails were clear. Three 4-mm punch biopsies were performed at the initial presentation: 1 perilesional biopsy for direct immunofluorescence testing and 2 lesional biopsies for routine histologic evaluation. An extensive serologic workup failed to reveal abnormalities. An activated partial thromboplastin time, dilute Russell viper venom time, serum protein electrophoresis, and levels of rheumatoid factor and angiotensin-converting enzyme were within reference range. Anticardiolipin antibodies IgA, IgM, and IgG were negative. A cryoglobulin test was negative.

Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).
FIGURE 2. Histopathology revealed a papillary dermis containing a proliferation of irregularly shaped vascular spaces with plump endothelium (H&E, original magnification ×200).

Histopathology revealed a proliferation of irregularly shaped vascular spaces with plump endothelium in the papillary dermis (Figure 2). Scattered leukocyte common antigen-positive lymphocytes were noted within lesions. The epidermis appeared normal, without evidence of spongiosis or alteration of the stratum corneum. Immunohistochemical studies of the perilesional skin biopsy revealed positivity for CD31 and D2-40 (Figure 3). Specimens were negative for CD20 and human herpesvirus 8. Direct immunofluorescence of the perilesional biopsy was negative.

Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).
FIGURE 3. Positive direct immunofluorescence staining (brown pigment) of a punch biopsy specimen of endothelium confined to lymphatic vessels with D2-40 (original magnification ×200).

A diagnosis of RAE was made based on clinical and histologic findings. Treatment with triamcinolone ointment 0.1% twice daily and oral cetirizine 10 mg twice daily was initiated. Re-evaluation 2 weeks later revealed notable improvement in the affected areas, including decreased edema, improvement of the purpura, and absence of pruritus. The patient noted no further spread or blister formation while the active areas were being treated with the topical steroid. The treatment regimen was modified to triamcinolone ointment 0.1% once daily, and cetirizine was discontinued. At 3-month follow-up, active areas had completely resolved (Figure 4) and triamcinolone was discontinued. To date, the patient has not had recurrence of symptoms and remains healthy.

At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.
FIGURE 4. A and B, At 3-month follow-up, reactive angioendotheliomatosis on the left scapula and left thenar eminence, respectively, had completely resolved after treatment with triamcinolone ointment 0.1% and oral cetirizine.

 

 

Gottron and Nikolowski3 reported the first case of RAE in an adult patient who presented with purpuric patches secondary to skin infarction. Current definitions use the umbrella term cutaneous reactive angiomatosis to cover 3 major subtypes: reactive angioendotheliomatosis, diffuse dermal angioendotheliomatosis, and acroangiodermatitis (pseudo-Kaposi sarcoma [KS]). The manifestation of these subgroups is clinically similar, and they must be differentiated through histologic evaluation.4

Reactive angioendotheliomatosis has an unknown pathogenesis and is poorly defined clinically. The exact pathophysiology is unknown but likely is linked to vaso-occlusion and hypoxia.1 A PubMed search of articles indexed for MEDLINE, as well as a review of Science Direct, Google Scholar, and Cochrane Library, using the terms reactive angioendotheliomatosis, COVID, vaccine, Ad26.COV2.S, and RAE in any combination revealed no prior cases of RAE in association with Ad26.COV2.S vaccination.

By the late 1980s, systemic angioendotheliomatosis was segregated into 2 distinct entities: malignant and reactive.4 The differential diagnosis of malignant systemic angioendotheliomatosis includes KS and angiosarcoma; nonmalignant causes are the variants of cutaneous reactive angiomatosis. It is important to rule out KS because of its malignant and deceptive nature. It is unknown if KS originates in blood vessels or lymphatic endothelial cells; however, evidence is strongly in favor of blood vessel origin using CD31 and CD34 endothelial markers.5 CD34 positivity is more reliable than CD31 in diagnosing KS, but the absence of both markers does not offer enough evidence to rule out KS on its own.6

In our patient, histopathology revealed cells positive for CD31 and D2-40; the latter is a lymphatic endothelial cell marker that stains the endothelium of lymphatic channels but not blood vessels.7 Positive D2-40 can be indicative of KS and non-KS lesions, each with a distinct staining pattern. D2-40 staining on non-KS lesions is confined to lymphatic vessels, as it was in our patient; in contrast, spindle-shaped cells also will be stained in KS lesions.8

Another cell marker, CD20, is a B cell–specific protein that can be measured to help diagnose malignant diseases such as B-cell lymphoma and leukemia. Human herpesvirus 8 (also known as KS-associated herpesvirus) is the infectious cause of KS and traditionally has been detected using methods such as the polymerase chain reaction.9,10

Most cases of RAE are idiopathic and occur in association with systemic disease, which was not the case in our patient. We speculated that his reaction was most likely triggered by vascular transfection of endothelial cells secondary to Ad26.COV2.S vaccination. Alternatively, vaccination may have caused vascular occlusion, though the lack of cyanosis, nail changes, and route of inoculant make this less likely.

All approved COVID-19 vaccines are designed solely for intramuscular injection. In comparison to other types of tissue, muscles have superior vascularity, allowing for enhanced mobilization of compounds, which results in faster systemic circulation.11 Alternative methods of injection, including intravascular, subcutaneous, and intradermal, may lead to decreased efficacy or adverse events, or both.

 

 

Prior cases of RAE have been treated with laser therapy, topical or systemic corticosteroids, excisional removal, or topical β-blockers, such as timolol.12β-Blocking agents act on β-adrenergic receptors on endothelial cells to inhibit angiogenesis by reducing release of blood vessel growth-signaling molecules and triggering apoptosis. In this patient, topical steroids and oral antihistamines were sufficient treatment.

Vaccine-related adverse events have been reported but remain rare. The benefits of Ad26.COV2.S vaccination for protection against COVID-19 outweigh the extremely low risk for adverse events.13 For that reason, the Centers for Disease Control and Prevention recommends a booster for individuals who are eligible to maximize protection. Intramuscular injection of Ad26.COV2.S resulted in a lower incidence of moderate to severe COVID-19 cases in all age groups vs the placebo group. Hypersensitivity adverse events were reported in 0.4% of Ad26.COV2.S-vaccinated patients vs 0.4% of patients who received a placebo; the more common reactions were nonanaphylactic.13

There have been 12 reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, which sparked nationwide controversy over the safety of the Ad26.COV2.S vaccine.14 After further investigation into those reports, the US Food and Drug Administration and the Centers for Disease Control and Prevention concluded that the benefits of the Ad26.COV2.S vaccine outweigh the low risk for associated thrombosis.15

Although adverse reactions are rare, it is important that health care providers take proper safety measures before and while administering any COVID-19 vaccine. Patients should be screened for contraindications to the COVID-19 vaccine to mitigate adverse effects seen in the small percentage of patients who may need to take alternative precautions.

The broad tissue tropism and high transmissibility of SARS-CoV-2 are the main contributors to its infection having reached pandemic scale. The spike (S) protein on SARS-CoV-2 binds to ACE2, the most thoroughly studied SARS-CoV-2 receptor, which is found in a range of tissues, including arterial endothelial cells, leading to its transfection. Several studies have proposed that expression of the S protein causes endothelial dysfunction through cytokine release, activation of complement, and ultimately microvascular occlusion.16

Recent developments in the use of viral-like particles, such as vesicular stomatitis virus, may mitigate future cases of RAE that are associated with endothelial cell transfection. Vesicular stomatitis virus is a popular model virus for research applications due to its glycoprotein and matrix protein contributing to its broad tropism. Recent efforts to alter these proteins have successfully limited the broad tropism of vesicular stomatitis virus.17

The SARS-CoV-2 virus must be handled in a Biosafety Level 3 laboratory. Conversely, pseudoviruses can be handled in lower containment facilities due to their safe and efficacious nature, offering an avenue to expedite vaccine development against many viral outbreaks, including SARS-CoV-2.18

 

 

An increasing number of cutaneous manifestations have been associated with COVID-19 infection and vaccination. Eruptive pseudoangiomatosis, a rare self-limiting exanthem, has been reported in association with ­COVID-19 vaccination.19 Eruptive pseudoangiomatosis manifests as erythematous blanchable papules that resemble angiomas, typically in a widespread distribution. Eruptive pseudoangiomatosis has striking similarities to RAE histologically; both manifest as dilated dermal blood vessels with plump endothelial cells.

Our case is unique because of the vasculitic palpable nature of the lesions, which were localized to the left arm. Eruptive pseudoangiomatosis formation after COVID-19 infection or SARS-CoV-2 vaccination may suggest alteration of ACE2 by binding of S protein.20 Such alteration of the ACE2 pathway would lead to inflammation of angiotensin II, causing proliferation of endothelial cells in the formation of angiomalike lesions. This hypothesis suggests a paraviral eruption secondary to an immunologic reaction, not a classical virtual eruption from direct contact of the virus on blood vessels. Although EPA and RAE are harmless and self-limiting, these reports will spread awareness of the increasing number of skin manifestations related to COVID-19 and SARS-CoV-2 virus vaccination.

Acknowledgment—Thoughtful insights and comments on this manuscript were provided by Christine J. Ko, MD (New Haven, Connecticut); Christine L. Egan, MD (Glen Mills, Pennsylvania); Howard A. Bueller, MD (Delray Beach, Florida); and Juan Pablo Robles, PhD (Juriquilla, Mexico).

References
  1. McMenamin ME, Fletcher CDM. Reactive angioendotheliomatosis: a study of 15 cases demonstrating a wide clinicopathologic spectrum. Am J Surg Pathol. 2002;26:686-697. doi:10.1097/00000478-200206000-00001
  2. Khan S, Pujani M, Jetley S, et al. Angiomatosis: a rare vascular proliferation of head and neck region. J Cutan Aesthet Surg. 2015;8:108-110. doi:10.4103/0974-2077.158448
  3. Gottron HA, Nikolowski W. Extrarenal Lohlein focal nephritis of the skin in endocarditis. Arch Klin Exp Dermatol. 1958;207:156-176.
  4. Cooper PH. Angioendotheliomatosis: two separate diseases. J Cutan Pathol. 1988;15:259. doi:10.1111/j.1600-0560.1988.tb00556.x
  5. Cancian L, Hansen A, Boshoff C. Cellular origin of Kaposi’s sarcoma and Kaposi’s sarcoma-associated herpesvirus-induced cell reprogramming. Trends Cell Biol. Sep 2013;23:421-32. doi:10.1016/j.tcb.2013.04.001
  6. Russell Jones R, Orchard G, Zelger B, et al. Immunostaining for CD31 and CD34 in Kaposi sarcoma. J Clin Pathol. 1995;48:1011-1016. doi:10.1136/jcp.48.11.1011
  7. Kahn HJ, Bailey D, Marks A. Monoclonal antibody D2-40, a new marker of lymphatic endothelium, reacts with Kaposi’s sarcoma and a subset of angiosarcomas. Mod Pathol. 2002;15:434-440. doi:10.1038/modpathol.3880543
  8. Genedy RM, Hamza AM, Abdel Latef AA, et al. Sensitivity and specificity of D2-40 in differentiating Kaposi sarcoma from its mimickers. J Egyptian Womens Dermatolog Soc. 2021;18:67-74. doi:10.4103/jewd.jewd_61_20
  9. Mesri EA, Cesarman E, Boshoff C. Kaposi’s sarcoma and its associated herpesvirus. Nat Rev Cancer. 2010;10:707-719. doi:10.1038/nrc2888
  10. Patel RM, Goldblum JR, Hsi ED. Immunohistochemical detection of human herpes virus-8 latent nuclear antigen-1 is useful in the diagnosis of Kaposi sarcoma. Mod Pathol. 2004;17:456-460. doi:10.1038/modpathol.3800061
  11. Zuckerman JN. The importance of injecting vaccines into muscle. Different patients need different needle sizes. BMJ. 2000;321:1237-1238. doi:10.1136/bmj.321.7271.1237
  12. Bhatia R, Hazarika N, Chandrasekaran D, et al. Treatment of posttraumatic reactive angioendotheliomatosis with topical timolol maleate. JAMA Dermatol. 2021;157:1002-1004. doi:10.1001/jamadermatol.2021.1770
  13. Sadoff J, Gray G, Vandebosch A, et al; ENSEMBLE Study Group. Safety and efficacy of single-dose Ad26.COV2.S vaccine against Covid-19. N Engl J Med. 2021;384:2187-2201. doi:10.1056/NEJMoa2101544
  14. See I, Su JR, Lale A, et al. US case reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, March 2 to April 21, 2021. JAMA. 2021;325:2448-2456. doi:10.1001/jama.2021.7517
  15. Berry CT, Eliliwi M, Gallagher S, et al. Cutaneous small vessel vasculitis following single-dose Janssen Ad26.COV2.S vaccination. JAAD Case Rep. 2021;15:11-14. doi:10.1016/j.jdcr.2021.07.002
  16. Flaumenhaft R, Enjyoji K, Schmaier AA. Vasculopathy in COVID-19. Blood. 2022;140:222-235. doi:10.1182/blood.2021012250
  17. Hastie E, Cataldi M, Marriott I, et al. Understanding and altering cell tropism of vesicular stomatitis virus. Virus Res. 2013;176:16-32. doi:10.1016/j.virusres.2013.06.003
  18. Xiong H-L, Wu Y-T, Cao J-L, et al. Robust neutralization assay based on SARS-CoV-2 S-protein-bearing vesicular stomatitis virus (VSV) pseudovirus and ACE2-overexpressing BHK21 cells. Emerg Microbes Infect. 2020;9:2105-2113. doi:10.1080/22221751.2020.1815589
  19. Mohta A, Jain SK, Mehta RD, et al. Development of eruptive pseudoangiomatosis following COVID-19 immunization – apropos of 5 cases. J Eur Acad Dermatol Venereol. 2021;35:e722-e725. doi:10.1111/jdv.17499
  20. Angeli F, Spanevello A, Reboldi G, et al. SARS-CoV-2 vaccines: lights and shadows. Eur J Intern Med. 2021;88:1-8. doi:10.1016/j.ejim.2021.04.019
References
  1. McMenamin ME, Fletcher CDM. Reactive angioendotheliomatosis: a study of 15 cases demonstrating a wide clinicopathologic spectrum. Am J Surg Pathol. 2002;26:686-697. doi:10.1097/00000478-200206000-00001
  2. Khan S, Pujani M, Jetley S, et al. Angiomatosis: a rare vascular proliferation of head and neck region. J Cutan Aesthet Surg. 2015;8:108-110. doi:10.4103/0974-2077.158448
  3. Gottron HA, Nikolowski W. Extrarenal Lohlein focal nephritis of the skin in endocarditis. Arch Klin Exp Dermatol. 1958;207:156-176.
  4. Cooper PH. Angioendotheliomatosis: two separate diseases. J Cutan Pathol. 1988;15:259. doi:10.1111/j.1600-0560.1988.tb00556.x
  5. Cancian L, Hansen A, Boshoff C. Cellular origin of Kaposi’s sarcoma and Kaposi’s sarcoma-associated herpesvirus-induced cell reprogramming. Trends Cell Biol. Sep 2013;23:421-32. doi:10.1016/j.tcb.2013.04.001
  6. Russell Jones R, Orchard G, Zelger B, et al. Immunostaining for CD31 and CD34 in Kaposi sarcoma. J Clin Pathol. 1995;48:1011-1016. doi:10.1136/jcp.48.11.1011
  7. Kahn HJ, Bailey D, Marks A. Monoclonal antibody D2-40, a new marker of lymphatic endothelium, reacts with Kaposi’s sarcoma and a subset of angiosarcomas. Mod Pathol. 2002;15:434-440. doi:10.1038/modpathol.3880543
  8. Genedy RM, Hamza AM, Abdel Latef AA, et al. Sensitivity and specificity of D2-40 in differentiating Kaposi sarcoma from its mimickers. J Egyptian Womens Dermatolog Soc. 2021;18:67-74. doi:10.4103/jewd.jewd_61_20
  9. Mesri EA, Cesarman E, Boshoff C. Kaposi’s sarcoma and its associated herpesvirus. Nat Rev Cancer. 2010;10:707-719. doi:10.1038/nrc2888
  10. Patel RM, Goldblum JR, Hsi ED. Immunohistochemical detection of human herpes virus-8 latent nuclear antigen-1 is useful in the diagnosis of Kaposi sarcoma. Mod Pathol. 2004;17:456-460. doi:10.1038/modpathol.3800061
  11. Zuckerman JN. The importance of injecting vaccines into muscle. Different patients need different needle sizes. BMJ. 2000;321:1237-1238. doi:10.1136/bmj.321.7271.1237
  12. Bhatia R, Hazarika N, Chandrasekaran D, et al. Treatment of posttraumatic reactive angioendotheliomatosis with topical timolol maleate. JAMA Dermatol. 2021;157:1002-1004. doi:10.1001/jamadermatol.2021.1770
  13. Sadoff J, Gray G, Vandebosch A, et al; ENSEMBLE Study Group. Safety and efficacy of single-dose Ad26.COV2.S vaccine against Covid-19. N Engl J Med. 2021;384:2187-2201. doi:10.1056/NEJMoa2101544
  14. See I, Su JR, Lale A, et al. US case reports of cerebral venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, March 2 to April 21, 2021. JAMA. 2021;325:2448-2456. doi:10.1001/jama.2021.7517
  15. Berry CT, Eliliwi M, Gallagher S, et al. Cutaneous small vessel vasculitis following single-dose Janssen Ad26.COV2.S vaccination. JAAD Case Rep. 2021;15:11-14. doi:10.1016/j.jdcr.2021.07.002
  16. Flaumenhaft R, Enjyoji K, Schmaier AA. Vasculopathy in COVID-19. Blood. 2022;140:222-235. doi:10.1182/blood.2021012250
  17. Hastie E, Cataldi M, Marriott I, et al. Understanding and altering cell tropism of vesicular stomatitis virus. Virus Res. 2013;176:16-32. doi:10.1016/j.virusres.2013.06.003
  18. Xiong H-L, Wu Y-T, Cao J-L, et al. Robust neutralization assay based on SARS-CoV-2 S-protein-bearing vesicular stomatitis virus (VSV) pseudovirus and ACE2-overexpressing BHK21 cells. Emerg Microbes Infect. 2020;9:2105-2113. doi:10.1080/22221751.2020.1815589
  19. Mohta A, Jain SK, Mehta RD, et al. Development of eruptive pseudoangiomatosis following COVID-19 immunization – apropos of 5 cases. J Eur Acad Dermatol Venereol. 2021;35:e722-e725. doi:10.1111/jdv.17499
  20. Angeli F, Spanevello A, Reboldi G, et al. SARS-CoV-2 vaccines: lights and shadows. Eur J Intern Med. 2021;88:1-8. doi:10.1016/j.ejim.2021.04.019
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E20-E23
Page Number
E20-E23
Publications
Publications
Topics
Article Type
Display Headline
Reactive Angioendotheliomatosis Following Ad26.COV2.S Vaccination
Display Headline
Reactive Angioendotheliomatosis Following Ad26.COV2.S Vaccination
Sections
Inside the Article

Practice points

  • Reactive angioendotheliomatosis (RAE) is a rare benign vascular proliferation of endothelial cells lining blood vessels that clinically appears similar to Kaposi sarcoma and must be differentiated by microscopic evaluation.
  • An increasing number of reports link SARS-CoV-2 viral infection or vaccination against this virus with various cutaneous manifestations. Our case offers a link between RAE and Ad26.COV2.S vaccination.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Bilateral Burning Palmoplantar Lesions

Article Type
Changed
Wed, 02/28/2024 - 07:30
Display Headline
Bilateral Burning Palmoplantar Lesions

The Diagnosis: Lichen Sclerosus

Histopathology revealed a thin epidermis with homogenization of the upper dermal collagen. By contrast, the lower dermis was sclerotic with patchy chronic dermal infiltrate (Figure). Ultimately, the patient’s clinical presentation and histopathologic findings led to a diagnosis of lichen sclerosus (LS).

Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).
A and B, Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).

Lichen sclerosus is a rare chronic inflammatory skin condition that typically is characterized by porcelainwhite atrophic plaques on the skin, most often involving the external female genitalia including the vulva and perianal area.1 It is thought to be underdiagnosed and underreported.2 Extragenital manifestations may occur, though some cases are characterized by concomitant genital involvement.3,4 Our patient presented with palmoplantar distribution of plaques without genitalia involvement. Approximately 6% to 10% of patients with extragenital LS do not have genital involvement at the time of diagnosis.3,5 Furthermore, LS involving the palms and soles is exceedingly rare.2 Although extragenital LS may be asymptomatic, patients can experience debilitating pruritus; bullae with hemorrhage and erosion; plaque thickening with repeated excoriations; and painful fissuring, especially if lesions are in areas that are susceptible to friction or tension.3,6 New lesions on previously unaffected skin also may develop secondary to trauma through the Koebner phenomenon.1,6

Histologically, LS is characterized by epidermal hyperkeratosis accompanied by follicular plugging, epidermal atrophy with flattened rete ridges, vacuolization of the basal epidermis, marked edema in the superficial dermis (in early lesions) or homogenized collagen in the upper dermis (in established lesions), and a lymphohistiocytic infiltrate beneath the homogenized collagen. Although the pathogenesis of LS is unclear, purported etiologic factors from studies in genital disease include immune dysfunction, genetic predisposition, infection, and trauma.6 Lichen sclerosus is associated strongly with autoimmune diseases including alopecia areata, vitiligo, autoimmune thyroiditis, diabetes mellitus, and pernicious anemia, indicating its potential multifactorial etiology and linkage to T-lymphocyte dysfunction.1 Early LS lesions often appear as flat-topped and slightly scaly, hypopigmented, white or mildly erythematous, polygonal papules that coalesce to form larger plaques with peripheral erythema. With time, the inflammation subsides, and lesions become porcelain-white with varying degrees of palpable sclerosis, resembling thin paperlike wrinkles indicative of epidermal atrophy.6

The differential diagnosis of LS includes lichen planus (LP), morphea, discoid lupus erythematosus (DLE), and vitiligo.3 Lesions of LP commonly are described as flat-topped, polygonal, pink-purple papules localized mostly along the volar wrists, shins, presacral area, and hands.7 Lichen planus is considered to be more pruritic3 than LS and can be further distinguished by biopsy through identifying a well-formed granular layer and numerous cytoid bodies. Unlike LS, LP is not characterized by basement membrane thickening or epidermal atrophy.8

Skin lesions seen in morphea may resemble the classic atrophic white lesions of extragenital LS; however, it is unclear if the appearance of LS-like lesions with morphea is a simultaneous occurrence of 2 separate disorders or the development of clinical findings resembling LS in lesions of morphea.6 Furthermore, morphea involves deep inflammation and sclerosis of the dermis that may extend into subcutaneous fat without follicular plugging of the epidermis.3,9 In contrast, LS primarily affects the epidermis and dermis with the presence of epidermal follicular plugging.6

Lesions seen in DLE are characterized as well-defined, annular, erythematous patches and plaques followed by follicular hyperkeratosis with adherent scaling. Upon removal of the scale, follicle-sized keratotic spikes (carpet tacks) are present.10 Scaling of lesions and the carpet tack sign were absent in our patient. In addition, DLE typically reveals surrounding pigmentation and scarring over plaques,3 which were not observed in our patient.

Vitiligo commonly is associated with extragenital LS. As with LS, vitiligo can be explained by mechanisms of immune checkpoint inhibitor–induced cytotoxicity as well as perforin and granzyme-B expression.11 Although vitiligo resembles the late hypopigmented lesions of extragenital LS, there are no plaques or surface changes, and a larger, more generalized area of the skin typically is involved.3

References
  1. Chamli A, Souissi A. Lichen sclerosus. StatPearls [Internet]. StatPearls Publishing; 2022. http://www.ncbi.nlm.nih.gov/books/NBK538246/
  2. Gaddis KJ, Huang J, Haun PL. An atrophic and spiny eruption of the palms. JAMA Dermatol. 2018;154:1344-1345. doi:10.1001 /jamadermatol.2018.1265
  3. Arif T, Fatima R, Sami M. Extragenital lichen sclerosus: a comprehensive review [published online August 11, 2022]. Australas J Dermatol. doi:10.1111/ajd.13890
  4. Heibel HD, Styles AR, Cockerell CJ. A case of acral lichen sclerosus et atrophicus. JAAD Case Rep. 2020;8:26-27. doi:10.1016/j.jdcr.2020.12.008
  5. Seyffert J, Bibliowicz N, Harding T, et al. Palmar lichen sclerosus et atrophicus. JAAD Case Rep. 2020;6:697-699. doi:10.1016/j.jdcr.2020.06.005
  6. Jacobe H. Extragenital lichen sclerosus: clinical features and diagnosis. UpToDate. Updated July 11, 2023. Accessed December 14, 2023. https://www.uptodate.com/contents/extragenital-lichen-sclerosus?search=Lichen%20sclerosus&source =search_result&selectedTitle=2~66&usage_type=default&display_ rank=2
  7. Goldstein BG, Goldstein AO, Mostow E. Lichen planus. UpToDate. Updated October 25, 2021. Accessed December 14, 2023. https://www.uptodate.com/contents/lichen-planus?search=lichen%20 sclerosus&topicRef=15838&source=see_link
  8. Tallon B. Lichen sclerosus pathology. DermNet NZ website. Accessed December 5, 2023. https://dermnetnz.org/topics/lichen-sclerosus-pathology
  9. Jacobe H. Pathogenesis, clinical manifestations, and diagnosis of morphea (localized scleroderma) in adults. UpToDate. Updated November 15, 2021. Accessed December 14, 2023. https://medilib.ir/uptodate/show/13776
  10. McDaniel B, Sukumaran S, Koritala T, et al. Discoid lupus erythematosus. StatPearls [Internet]. StatPearls Publishing; 2022. Updated August 28, 2023. Accessed December 14, 2023. http://www.ncbi.nlm.nih.gov/books/NBK493145/
  11. Veronesi G, Scarfì F, Misciali C, et al. An unusual skin reaction in uveal melanoma during treatment with nivolumab: extragenital lichen sclerosus. Anticancer Drugs. 2019;30:969-972. doi:10.1097/ CAD.0000000000000819
Article PDF
Author and Disclosure Information

From the University of Florida College of Medicine, Gainesville. Drs. Gurnani and Montañez-Wiscovich are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Marjorie E. Montañez-Wiscovich, MD, PhD, 4037 NW 86 Terr, 4th Floor, Gainesville, FL 32606 (m.montanez@dermatology.med.ufl.edu).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E24-E26
Sections
Author and Disclosure Information

From the University of Florida College of Medicine, Gainesville. Drs. Gurnani and Montañez-Wiscovich are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Marjorie E. Montañez-Wiscovich, MD, PhD, 4037 NW 86 Terr, 4th Floor, Gainesville, FL 32606 (m.montanez@dermatology.med.ufl.edu).

Author and Disclosure Information

From the University of Florida College of Medicine, Gainesville. Drs. Gurnani and Montañez-Wiscovich are from the Department of Dermatology.

The authors report no conflict of interest.

Correspondence: Marjorie E. Montañez-Wiscovich, MD, PhD, 4037 NW 86 Terr, 4th Floor, Gainesville, FL 32606 (m.montanez@dermatology.med.ufl.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Lichen Sclerosus

Histopathology revealed a thin epidermis with homogenization of the upper dermal collagen. By contrast, the lower dermis was sclerotic with patchy chronic dermal infiltrate (Figure). Ultimately, the patient’s clinical presentation and histopathologic findings led to a diagnosis of lichen sclerosus (LS).

Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).
A and B, Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).

Lichen sclerosus is a rare chronic inflammatory skin condition that typically is characterized by porcelainwhite atrophic plaques on the skin, most often involving the external female genitalia including the vulva and perianal area.1 It is thought to be underdiagnosed and underreported.2 Extragenital manifestations may occur, though some cases are characterized by concomitant genital involvement.3,4 Our patient presented with palmoplantar distribution of plaques without genitalia involvement. Approximately 6% to 10% of patients with extragenital LS do not have genital involvement at the time of diagnosis.3,5 Furthermore, LS involving the palms and soles is exceedingly rare.2 Although extragenital LS may be asymptomatic, patients can experience debilitating pruritus; bullae with hemorrhage and erosion; plaque thickening with repeated excoriations; and painful fissuring, especially if lesions are in areas that are susceptible to friction or tension.3,6 New lesions on previously unaffected skin also may develop secondary to trauma through the Koebner phenomenon.1,6

Histologically, LS is characterized by epidermal hyperkeratosis accompanied by follicular plugging, epidermal atrophy with flattened rete ridges, vacuolization of the basal epidermis, marked edema in the superficial dermis (in early lesions) or homogenized collagen in the upper dermis (in established lesions), and a lymphohistiocytic infiltrate beneath the homogenized collagen. Although the pathogenesis of LS is unclear, purported etiologic factors from studies in genital disease include immune dysfunction, genetic predisposition, infection, and trauma.6 Lichen sclerosus is associated strongly with autoimmune diseases including alopecia areata, vitiligo, autoimmune thyroiditis, diabetes mellitus, and pernicious anemia, indicating its potential multifactorial etiology and linkage to T-lymphocyte dysfunction.1 Early LS lesions often appear as flat-topped and slightly scaly, hypopigmented, white or mildly erythematous, polygonal papules that coalesce to form larger plaques with peripheral erythema. With time, the inflammation subsides, and lesions become porcelain-white with varying degrees of palpable sclerosis, resembling thin paperlike wrinkles indicative of epidermal atrophy.6

The differential diagnosis of LS includes lichen planus (LP), morphea, discoid lupus erythematosus (DLE), and vitiligo.3 Lesions of LP commonly are described as flat-topped, polygonal, pink-purple papules localized mostly along the volar wrists, shins, presacral area, and hands.7 Lichen planus is considered to be more pruritic3 than LS and can be further distinguished by biopsy through identifying a well-formed granular layer and numerous cytoid bodies. Unlike LS, LP is not characterized by basement membrane thickening or epidermal atrophy.8

Skin lesions seen in morphea may resemble the classic atrophic white lesions of extragenital LS; however, it is unclear if the appearance of LS-like lesions with morphea is a simultaneous occurrence of 2 separate disorders or the development of clinical findings resembling LS in lesions of morphea.6 Furthermore, morphea involves deep inflammation and sclerosis of the dermis that may extend into subcutaneous fat without follicular plugging of the epidermis.3,9 In contrast, LS primarily affects the epidermis and dermis with the presence of epidermal follicular plugging.6

Lesions seen in DLE are characterized as well-defined, annular, erythematous patches and plaques followed by follicular hyperkeratosis with adherent scaling. Upon removal of the scale, follicle-sized keratotic spikes (carpet tacks) are present.10 Scaling of lesions and the carpet tack sign were absent in our patient. In addition, DLE typically reveals surrounding pigmentation and scarring over plaques,3 which were not observed in our patient.

Vitiligo commonly is associated with extragenital LS. As with LS, vitiligo can be explained by mechanisms of immune checkpoint inhibitor–induced cytotoxicity as well as perforin and granzyme-B expression.11 Although vitiligo resembles the late hypopigmented lesions of extragenital LS, there are no plaques or surface changes, and a larger, more generalized area of the skin typically is involved.3

The Diagnosis: Lichen Sclerosus

Histopathology revealed a thin epidermis with homogenization of the upper dermal collagen. By contrast, the lower dermis was sclerotic with patchy chronic dermal infiltrate (Figure). Ultimately, the patient’s clinical presentation and histopathologic findings led to a diagnosis of lichen sclerosus (LS).

Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).
A and B, Histopathology revealed a thin epidermis with homogenization of upper dermal collagen and a sclerotic dermis with a patchy chronic dermal infiltrate (H&E, original magnifications ×40 and ×100).

Lichen sclerosus is a rare chronic inflammatory skin condition that typically is characterized by porcelainwhite atrophic plaques on the skin, most often involving the external female genitalia including the vulva and perianal area.1 It is thought to be underdiagnosed and underreported.2 Extragenital manifestations may occur, though some cases are characterized by concomitant genital involvement.3,4 Our patient presented with palmoplantar distribution of plaques without genitalia involvement. Approximately 6% to 10% of patients with extragenital LS do not have genital involvement at the time of diagnosis.3,5 Furthermore, LS involving the palms and soles is exceedingly rare.2 Although extragenital LS may be asymptomatic, patients can experience debilitating pruritus; bullae with hemorrhage and erosion; plaque thickening with repeated excoriations; and painful fissuring, especially if lesions are in areas that are susceptible to friction or tension.3,6 New lesions on previously unaffected skin also may develop secondary to trauma through the Koebner phenomenon.1,6

Histologically, LS is characterized by epidermal hyperkeratosis accompanied by follicular plugging, epidermal atrophy with flattened rete ridges, vacuolization of the basal epidermis, marked edema in the superficial dermis (in early lesions) or homogenized collagen in the upper dermis (in established lesions), and a lymphohistiocytic infiltrate beneath the homogenized collagen. Although the pathogenesis of LS is unclear, purported etiologic factors from studies in genital disease include immune dysfunction, genetic predisposition, infection, and trauma.6 Lichen sclerosus is associated strongly with autoimmune diseases including alopecia areata, vitiligo, autoimmune thyroiditis, diabetes mellitus, and pernicious anemia, indicating its potential multifactorial etiology and linkage to T-lymphocyte dysfunction.1 Early LS lesions often appear as flat-topped and slightly scaly, hypopigmented, white or mildly erythematous, polygonal papules that coalesce to form larger plaques with peripheral erythema. With time, the inflammation subsides, and lesions become porcelain-white with varying degrees of palpable sclerosis, resembling thin paperlike wrinkles indicative of epidermal atrophy.6

The differential diagnosis of LS includes lichen planus (LP), morphea, discoid lupus erythematosus (DLE), and vitiligo.3 Lesions of LP commonly are described as flat-topped, polygonal, pink-purple papules localized mostly along the volar wrists, shins, presacral area, and hands.7 Lichen planus is considered to be more pruritic3 than LS and can be further distinguished by biopsy through identifying a well-formed granular layer and numerous cytoid bodies. Unlike LS, LP is not characterized by basement membrane thickening or epidermal atrophy.8

Skin lesions seen in morphea may resemble the classic atrophic white lesions of extragenital LS; however, it is unclear if the appearance of LS-like lesions with morphea is a simultaneous occurrence of 2 separate disorders or the development of clinical findings resembling LS in lesions of morphea.6 Furthermore, morphea involves deep inflammation and sclerosis of the dermis that may extend into subcutaneous fat without follicular plugging of the epidermis.3,9 In contrast, LS primarily affects the epidermis and dermis with the presence of epidermal follicular plugging.6

Lesions seen in DLE are characterized as well-defined, annular, erythematous patches and plaques followed by follicular hyperkeratosis with adherent scaling. Upon removal of the scale, follicle-sized keratotic spikes (carpet tacks) are present.10 Scaling of lesions and the carpet tack sign were absent in our patient. In addition, DLE typically reveals surrounding pigmentation and scarring over plaques,3 which were not observed in our patient.

Vitiligo commonly is associated with extragenital LS. As with LS, vitiligo can be explained by mechanisms of immune checkpoint inhibitor–induced cytotoxicity as well as perforin and granzyme-B expression.11 Although vitiligo resembles the late hypopigmented lesions of extragenital LS, there are no plaques or surface changes, and a larger, more generalized area of the skin typically is involved.3

References
  1. Chamli A, Souissi A. Lichen sclerosus. StatPearls [Internet]. StatPearls Publishing; 2022. http://www.ncbi.nlm.nih.gov/books/NBK538246/
  2. Gaddis KJ, Huang J, Haun PL. An atrophic and spiny eruption of the palms. JAMA Dermatol. 2018;154:1344-1345. doi:10.1001 /jamadermatol.2018.1265
  3. Arif T, Fatima R, Sami M. Extragenital lichen sclerosus: a comprehensive review [published online August 11, 2022]. Australas J Dermatol. doi:10.1111/ajd.13890
  4. Heibel HD, Styles AR, Cockerell CJ. A case of acral lichen sclerosus et atrophicus. JAAD Case Rep. 2020;8:26-27. doi:10.1016/j.jdcr.2020.12.008
  5. Seyffert J, Bibliowicz N, Harding T, et al. Palmar lichen sclerosus et atrophicus. JAAD Case Rep. 2020;6:697-699. doi:10.1016/j.jdcr.2020.06.005
  6. Jacobe H. Extragenital lichen sclerosus: clinical features and diagnosis. UpToDate. Updated July 11, 2023. Accessed December 14, 2023. https://www.uptodate.com/contents/extragenital-lichen-sclerosus?search=Lichen%20sclerosus&source =search_result&selectedTitle=2~66&usage_type=default&display_ rank=2
  7. Goldstein BG, Goldstein AO, Mostow E. Lichen planus. UpToDate. Updated October 25, 2021. Accessed December 14, 2023. https://www.uptodate.com/contents/lichen-planus?search=lichen%20 sclerosus&topicRef=15838&source=see_link
  8. Tallon B. Lichen sclerosus pathology. DermNet NZ website. Accessed December 5, 2023. https://dermnetnz.org/topics/lichen-sclerosus-pathology
  9. Jacobe H. Pathogenesis, clinical manifestations, and diagnosis of morphea (localized scleroderma) in adults. UpToDate. Updated November 15, 2021. Accessed December 14, 2023. https://medilib.ir/uptodate/show/13776
  10. McDaniel B, Sukumaran S, Koritala T, et al. Discoid lupus erythematosus. StatPearls [Internet]. StatPearls Publishing; 2022. Updated August 28, 2023. Accessed December 14, 2023. http://www.ncbi.nlm.nih.gov/books/NBK493145/
  11. Veronesi G, Scarfì F, Misciali C, et al. An unusual skin reaction in uveal melanoma during treatment with nivolumab: extragenital lichen sclerosus. Anticancer Drugs. 2019;30:969-972. doi:10.1097/ CAD.0000000000000819
References
  1. Chamli A, Souissi A. Lichen sclerosus. StatPearls [Internet]. StatPearls Publishing; 2022. http://www.ncbi.nlm.nih.gov/books/NBK538246/
  2. Gaddis KJ, Huang J, Haun PL. An atrophic and spiny eruption of the palms. JAMA Dermatol. 2018;154:1344-1345. doi:10.1001 /jamadermatol.2018.1265
  3. Arif T, Fatima R, Sami M. Extragenital lichen sclerosus: a comprehensive review [published online August 11, 2022]. Australas J Dermatol. doi:10.1111/ajd.13890
  4. Heibel HD, Styles AR, Cockerell CJ. A case of acral lichen sclerosus et atrophicus. JAAD Case Rep. 2020;8:26-27. doi:10.1016/j.jdcr.2020.12.008
  5. Seyffert J, Bibliowicz N, Harding T, et al. Palmar lichen sclerosus et atrophicus. JAAD Case Rep. 2020;6:697-699. doi:10.1016/j.jdcr.2020.06.005
  6. Jacobe H. Extragenital lichen sclerosus: clinical features and diagnosis. UpToDate. Updated July 11, 2023. Accessed December 14, 2023. https://www.uptodate.com/contents/extragenital-lichen-sclerosus?search=Lichen%20sclerosus&source =search_result&selectedTitle=2~66&usage_type=default&display_ rank=2
  7. Goldstein BG, Goldstein AO, Mostow E. Lichen planus. UpToDate. Updated October 25, 2021. Accessed December 14, 2023. https://www.uptodate.com/contents/lichen-planus?search=lichen%20 sclerosus&topicRef=15838&source=see_link
  8. Tallon B. Lichen sclerosus pathology. DermNet NZ website. Accessed December 5, 2023. https://dermnetnz.org/topics/lichen-sclerosus-pathology
  9. Jacobe H. Pathogenesis, clinical manifestations, and diagnosis of morphea (localized scleroderma) in adults. UpToDate. Updated November 15, 2021. Accessed December 14, 2023. https://medilib.ir/uptodate/show/13776
  10. McDaniel B, Sukumaran S, Koritala T, et al. Discoid lupus erythematosus. StatPearls [Internet]. StatPearls Publishing; 2022. Updated August 28, 2023. Accessed December 14, 2023. http://www.ncbi.nlm.nih.gov/books/NBK493145/
  11. Veronesi G, Scarfì F, Misciali C, et al. An unusual skin reaction in uveal melanoma during treatment with nivolumab: extragenital lichen sclerosus. Anticancer Drugs. 2019;30:969-972. doi:10.1097/ CAD.0000000000000819
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E24-E26
Page Number
E24-E26
Publications
Publications
Topics
Article Type
Display Headline
Bilateral Burning Palmoplantar Lesions
Display Headline
Bilateral Burning Palmoplantar Lesions
Sections
Questionnaire Body

A 59-year-old woman presented with atrophic, hypopigmented, ivory papules and plaques localized to the central palms and soles of 3 years’ duration. The lesions were associated with burning that was most notable after extended periods of ambulation. The lesions initially were diagnosed as plaque psoriasis by an external dermatology clinic. At the time of presentation to our clinic, treatment with several highpotency topical steroids and biologics approved for plaque psoriasis had failed. Her medical history and concurrent medical workup were notable for type 2 diabetes mellitus, liver dysfunction, thyroid nodules overseen by an endocrinologist, vitamin B12 and vitamin D deficiencies managed with supplementation, and diffuse androgenic alopecia with suspected telogen effluvium. Physical examination revealed no plaque fissuring, pruritus, or scaling. She had no history of radiation therapy or organ transplantation. A punch biopsy of the left palm was performed.

Bilateral burning palmoplantar lesions

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 12/19/2023 - 11:45
Un-Gate On Date
Tue, 12/19/2023 - 11:45
Use ProPublica
CFC Schedule Remove Status
Tue, 12/19/2023 - 11:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Large Indurated Plaque on the Chest With Ulceration and Necrosis

Article Type
Changed
Wed, 12/13/2023 - 10:34
Display Headline
Large Indurated Plaque on the Chest With Ulceration and Necrosis

The Diagnosis: Carcinoma en Cuirasse

Histopathology demonstrated a cellular infiltrate filling the dermis with sparing of the papillary and superficial reticular dermis (Figure 1A). The cells were arranged in strands and cords that infiltrated between sclerotic collagen bundles. Cytomorphologically, the cells ranged from epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (Figure 1B). Occasional mitotic figures were identified, and cells demonstrated diffuse nuclear positivity for GATA-3 (Figure 1C); 55% of the cells demonstrated estrogen receptor positivity, and immunohistochemistry of progesterone receptors was negative. These findings confirmed our patient’s diagnosis of breast carcinoma en cuirasse (CeC) as the primary manifestation of metastatic invasive ductal carcinoma. Our patient was treated with intravenous chemotherapy and tamoxifen.

A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboid
FIGURE 1. A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (H&E, original magnification ×400). C, The cells also demonstrated diffuse nuclear positivity for GATA-3 (original magnification ×40).

Histopathologic findings of morphea include thickened hyalinized collagen bundles and loss of adventitial fat.1 A diagnosis of chronic radiation dermatitis was inconsistent with our patient’s medical history and biopsy results, as pathology should reveal hyalinized collagen or stellate radiation fibroblasts.2,3 Nests of squamous epithelial cells with abundant eosinophilic cytoplasm and large vesicular nuclei were not seen, excluding squamous cell carcinoma as a possible diagnosis.4 Although sclerosing sweat duct carcinoma is characterized by infiltrating cords in sclerotic dermis, the cells were not arranged in ductlike structures 1– to 2–cell layers thick, excluding this diagnosis.5

Carcinoma en cuirasse—named for skin involvement that appears similar to the metal breastplate of a cuirassier—is a rare form of cutaneous metastasis that typically presents with extensive infiltrative plaques resulting in fibrosis of the skin and subcutaneous tissue.6,7 Carcinoma en cuirasse most commonly metastasizes from the breast but also may represent metastases from the lungs, gastrointestinal tract, or genitourinary systems.8 In the setting of a primary breast malignancy, metastatic plaques of CeC tend to represent tumor recurrence following a mastectomy procedure; however, in rare cases CeC can present as the primary manifestation of breast cancer or as a result of untreated malignancy.6,9 In our patient, CeC was the primary manifestation of metastatic invasive ductal carcinoma with additional paraneoplastic ichthyosis (Figure 2).

Ichthyotic plaques with brown scaling on the leg.
FIGURE 2. Ichthyotic plaques with brown scaling on the leg.

Carcinoma en cuirasse comprises 3% to 6% of cutaneous metastases originating from the breast.10,11 Breast cancer is the most common primary neoplasm displaying extracutaneous metastasis, comprising 70% of all cutaneous metastases in females.11 Cutaneous metastasis often indicates late stage of disease, portending a poor prognosis. In our patient, the cutaneous nodules were present for approximately 3 years prior to the diagnosis of stage IV invasive ductal cell carcinoma with metastasis to the skin and lungs. Prior to admission, she had not been diagnosed with breast cancer, thus no treatments had been administered. It is uncommon for CeC to present as the initial finding and without prior treatment of the underlying malignancy. The median length of survival after diagnosis of cutaneous metastasis from breast cancer is 13.8 months, with a 10-year survival rate of 3.1%.12

In addition to cutaneous metastasis, breast cancer also may present with paraneoplastic dermatoses such as ichthyosis.13 Ichthyosis is characterized by extreme dryness, flaking, thickening, and mild pruritus.14 It most commonly is an inherited condition, but it may be acquired due to malignancy. Acquired ichthyosis may manifest in systemic diseases including systemic lupus erythematosus, sarcoidosis, and hypothyroidism.15 Although acquired ichthyosis is rare, it has been reported in cases of internal malignancy, most commonly lymphoproliferative malignancies and less frequently carcinoma of the breasts, cervix, and lungs. Patients who acquire ichthyosis in association with malignancy usually present with late-stage disease.15 Our patient acquired ichthyosis 3 months prior to admission and had never experienced it previously. Although the exact mechanism for acquiring ichthyosis remains unknown, it is uncertain if ichthyosis associated with malignancy is paraneoplastic or a result of chemotherapy.14,16 In this case, the patient had not yet started chemotherapy at the time of the ichthyosis diagnosis, suggesting a paraneoplastic etiology.

Carcinoma en cuirasse and paraneoplastic ichthyosis individually are extremely rare manifestations of breast cancer. Thus, it is even rarer for these conditions to present concurrently. Treatment options for CeC include chemotherapy, radiotherapy, hormonal antagonists, and snake venom.11 Systemic chemotherapy targeting the histopathologic type of the primary tumor is the treatment of choice. Other treatment methods usually are chosen for late stages of disease progression.10 Paraneoplastic ichthyosis has been reported to show improvement with treatment of the underlying primary malignancy by surgical removal or chemotherapy.14,17 Tamoxifen less commonly is used for systemic treatment of CeC, but one case in the literature reported favorable outcomes.18

We describe 2 rare cutaneous manifestations of breast cancer occurring concomitantly: CeC and paraneoplastic ichthyosis. The combination of clinical and pathologic findings presented in this case solidified the diagnosis of metastatic invasive ductal carcinoma. We aim to improve recognition of paraneoplastic skin findings to accelerate the process of effective and efficient treatment.

References
  1. Walker D, Susa JS, Currimbhoy S, et al. Histopathological changes in morphea and their clinical correlates: results from the Morphea in Adults and Children Cohort V. J Am Acad Dermatol. 2017;76:1124-1130. https://doi.org/10.1016/j.jaad.2016.12.020
  2. Borrelli MR, Shen AH, Lee GK, et al. Radiation-induced skin fibrosis: pathogenesis, current treatment options, and emerging therapeutics. Ann Plast Surg. 2019;83(4 suppl 1):S59-S64. https://doi.org/10.1097/SAP.0000000000002098
  3. Boncher J, Bergfeld WF. Fluoroscopy-induced chronic radiation dermatitis: a report of two additional cases and a brief review of the literature. J Cutan Pathol. 2012;39:63-67. https://doi.org/10.1111/j .1600-0560.2011.01754.x
  4. Cassarino DS, Derienzo DP, Barr RJ. Cutaneous squamous cell carcinoma: a comprehensive clinicopathologic classification. part one. J Cutan Pathol. 2006;33:191-206. https://doi.org/10.1111 /j.0303-6987.2006.00516_1.x
  5. Harvey DT, Hu J, Long JA, et al. Sclerosing sweat duct carcinoma of the lower extremity treated with Mohs micrographic surgery. JAAD Case Rep. 2016;2:284-286. https://doi.org/10.1016/j.jdcr.2016.05.017
  6. Sharma V, Kumar A. Carcinoma en cuirasse. N Engl J Med. 2021;385:2562. doi:10.1056/NEJMicm2111669
  7. Oliveira GM, Zachetti DB, Barros HR, et al. Breast carcinoma en cuirasse—case report. An Bras Dermatol. 2013;88:608-610. doi:10.1590/abd1806-4841.20131926
  8. Alcaraz I, Cerroni L, Rütten A, et al. Cutaneous metastases from internal malignancies: a clinicopathologic and immunohistochemical review. Am J Dermatopathol. 2012;34:347-393. doi:10.1097 /DAD.0b013e31823069cf
  9. Glazebrook AJ, Tomaszewski W. Ichthyosiform atrophy of the skin in Hodgkin’s disease: report of a case, with reference to vitamin A metabolism. Arch Derm Syphilol. 1944;50:85-89. doi:10.1001 /archderm.1944.01510140008002
  10. Mordenti C, Concetta F, Cerroni M, et al. Cutaneous metastatic breast carcinoma: a study of 164 patients. Acta Dermatovenerol Alp Pannonica Adriat. 2000;9:143-148.
  11. Culver AL, Metter DM, Pippen JE Jr. Carcinoma en cuirasse. Proc (Bayl Univ Med Cent). 2019;32:263-265. doi:10.1080/08998280.2018.1564966
  12. Schoenlaub P, Sarraux A, Grosshans E, et al. Survival after cutaneous metastasis: a study of 200 cases [in French]. Ann Dermatol Venereol. 2001;128:1310-1315.
  13. Tan AR. Cutaneous manifestations of breast cancer. Semin Oncol. 2016;43:331-334. doi:10.1053/j.seminoncol.2016.02.030
  14. Song Y, Wu Y, Fan T. Dermatosis as the initial manifestation of malignant breast tumors: retrospective analysis of 4 cases. Breast Care. 2010;5:174-176. doi:10.1159/000314265
  15. Polisky RB, Bronson DM. Acquired ichthyosis in a patient with adenocarcinoma of the breast. Cutis. 1986;38:359-360.
  16. Haste AR. Acquired ichthyosis from breast cancer. Br Med J. 1967;4:96-98.
  17. Riesco Martínez MC, Muñoz Martín AJ, Zamberk Majlis P, et al. Acquired ichthyosis as a paraneoplastic syndrome in Hodgkin’s disease. Clin Transl Oncol. 2009;11:552-553. doi:10.1007/s12094-009-0402-2
  18. Siddiqui MA, Zaman MN. Primary carcinoma en cuirasse. J Am Geriatr Soc. 1996;44:221-222. doi:10.1111/j.1532-5415.1996.tb02455.xssss
Article PDF
Author and Disclosure Information

Caroline E. Roberts and Dr. Cascardo are from the William Beaumont School of Medicine, Oakland University, Rochester, Michigan. Dr. Cornell is from the Department of Dermatology, Trinity Health Ann Arbor, Ypsilanti, Michigan.

The authors report no conflict of interest.

Correspondence: Caroline E. Roberts, BM, 586 Pioneer Dr, Rochester, MI 48309 (carolineroberts@oakland.edu).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
E12-E14
Sections
Author and Disclosure Information

Caroline E. Roberts and Dr. Cascardo are from the William Beaumont School of Medicine, Oakland University, Rochester, Michigan. Dr. Cornell is from the Department of Dermatology, Trinity Health Ann Arbor, Ypsilanti, Michigan.

The authors report no conflict of interest.

Correspondence: Caroline E. Roberts, BM, 586 Pioneer Dr, Rochester, MI 48309 (carolineroberts@oakland.edu).

Author and Disclosure Information

Caroline E. Roberts and Dr. Cascardo are from the William Beaumont School of Medicine, Oakland University, Rochester, Michigan. Dr. Cornell is from the Department of Dermatology, Trinity Health Ann Arbor, Ypsilanti, Michigan.

The authors report no conflict of interest.

Correspondence: Caroline E. Roberts, BM, 586 Pioneer Dr, Rochester, MI 48309 (carolineroberts@oakland.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Carcinoma en Cuirasse

Histopathology demonstrated a cellular infiltrate filling the dermis with sparing of the papillary and superficial reticular dermis (Figure 1A). The cells were arranged in strands and cords that infiltrated between sclerotic collagen bundles. Cytomorphologically, the cells ranged from epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (Figure 1B). Occasional mitotic figures were identified, and cells demonstrated diffuse nuclear positivity for GATA-3 (Figure 1C); 55% of the cells demonstrated estrogen receptor positivity, and immunohistochemistry of progesterone receptors was negative. These findings confirmed our patient’s diagnosis of breast carcinoma en cuirasse (CeC) as the primary manifestation of metastatic invasive ductal carcinoma. Our patient was treated with intravenous chemotherapy and tamoxifen.

A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboid
FIGURE 1. A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (H&E, original magnification ×400). C, The cells also demonstrated diffuse nuclear positivity for GATA-3 (original magnification ×40).

Histopathologic findings of morphea include thickened hyalinized collagen bundles and loss of adventitial fat.1 A diagnosis of chronic radiation dermatitis was inconsistent with our patient’s medical history and biopsy results, as pathology should reveal hyalinized collagen or stellate radiation fibroblasts.2,3 Nests of squamous epithelial cells with abundant eosinophilic cytoplasm and large vesicular nuclei were not seen, excluding squamous cell carcinoma as a possible diagnosis.4 Although sclerosing sweat duct carcinoma is characterized by infiltrating cords in sclerotic dermis, the cells were not arranged in ductlike structures 1– to 2–cell layers thick, excluding this diagnosis.5

Carcinoma en cuirasse—named for skin involvement that appears similar to the metal breastplate of a cuirassier—is a rare form of cutaneous metastasis that typically presents with extensive infiltrative plaques resulting in fibrosis of the skin and subcutaneous tissue.6,7 Carcinoma en cuirasse most commonly metastasizes from the breast but also may represent metastases from the lungs, gastrointestinal tract, or genitourinary systems.8 In the setting of a primary breast malignancy, metastatic plaques of CeC tend to represent tumor recurrence following a mastectomy procedure; however, in rare cases CeC can present as the primary manifestation of breast cancer or as a result of untreated malignancy.6,9 In our patient, CeC was the primary manifestation of metastatic invasive ductal carcinoma with additional paraneoplastic ichthyosis (Figure 2).

Ichthyotic plaques with brown scaling on the leg.
FIGURE 2. Ichthyotic plaques with brown scaling on the leg.

Carcinoma en cuirasse comprises 3% to 6% of cutaneous metastases originating from the breast.10,11 Breast cancer is the most common primary neoplasm displaying extracutaneous metastasis, comprising 70% of all cutaneous metastases in females.11 Cutaneous metastasis often indicates late stage of disease, portending a poor prognosis. In our patient, the cutaneous nodules were present for approximately 3 years prior to the diagnosis of stage IV invasive ductal cell carcinoma with metastasis to the skin and lungs. Prior to admission, she had not been diagnosed with breast cancer, thus no treatments had been administered. It is uncommon for CeC to present as the initial finding and without prior treatment of the underlying malignancy. The median length of survival after diagnosis of cutaneous metastasis from breast cancer is 13.8 months, with a 10-year survival rate of 3.1%.12

In addition to cutaneous metastasis, breast cancer also may present with paraneoplastic dermatoses such as ichthyosis.13 Ichthyosis is characterized by extreme dryness, flaking, thickening, and mild pruritus.14 It most commonly is an inherited condition, but it may be acquired due to malignancy. Acquired ichthyosis may manifest in systemic diseases including systemic lupus erythematosus, sarcoidosis, and hypothyroidism.15 Although acquired ichthyosis is rare, it has been reported in cases of internal malignancy, most commonly lymphoproliferative malignancies and less frequently carcinoma of the breasts, cervix, and lungs. Patients who acquire ichthyosis in association with malignancy usually present with late-stage disease.15 Our patient acquired ichthyosis 3 months prior to admission and had never experienced it previously. Although the exact mechanism for acquiring ichthyosis remains unknown, it is uncertain if ichthyosis associated with malignancy is paraneoplastic or a result of chemotherapy.14,16 In this case, the patient had not yet started chemotherapy at the time of the ichthyosis diagnosis, suggesting a paraneoplastic etiology.

Carcinoma en cuirasse and paraneoplastic ichthyosis individually are extremely rare manifestations of breast cancer. Thus, it is even rarer for these conditions to present concurrently. Treatment options for CeC include chemotherapy, radiotherapy, hormonal antagonists, and snake venom.11 Systemic chemotherapy targeting the histopathologic type of the primary tumor is the treatment of choice. Other treatment methods usually are chosen for late stages of disease progression.10 Paraneoplastic ichthyosis has been reported to show improvement with treatment of the underlying primary malignancy by surgical removal or chemotherapy.14,17 Tamoxifen less commonly is used for systemic treatment of CeC, but one case in the literature reported favorable outcomes.18

We describe 2 rare cutaneous manifestations of breast cancer occurring concomitantly: CeC and paraneoplastic ichthyosis. The combination of clinical and pathologic findings presented in this case solidified the diagnosis of metastatic invasive ductal carcinoma. We aim to improve recognition of paraneoplastic skin findings to accelerate the process of effective and efficient treatment.

The Diagnosis: Carcinoma en Cuirasse

Histopathology demonstrated a cellular infiltrate filling the dermis with sparing of the papillary and superficial reticular dermis (Figure 1A). The cells were arranged in strands and cords that infiltrated between sclerotic collagen bundles. Cytomorphologically, the cells ranged from epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (Figure 1B). Occasional mitotic figures were identified, and cells demonstrated diffuse nuclear positivity for GATA-3 (Figure 1C); 55% of the cells demonstrated estrogen receptor positivity, and immunohistochemistry of progesterone receptors was negative. These findings confirmed our patient’s diagnosis of breast carcinoma en cuirasse (CeC) as the primary manifestation of metastatic invasive ductal carcinoma. Our patient was treated with intravenous chemotherapy and tamoxifen.

A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboid
FIGURE 1. A, Histopathology demonstrated a dermal cellular infiltrate arranged in cords that dissected through the sclerotic collagen bundles (H&E, original magnification ×40). B, Cells were epithelioid with large vesicular nuclei and prominent nucleoli to cuboidal with hyperchromatic nuclei with irregular contours and a high nuclear to cytoplasmic ratio (H&E, original magnification ×400). C, The cells also demonstrated diffuse nuclear positivity for GATA-3 (original magnification ×40).

Histopathologic findings of morphea include thickened hyalinized collagen bundles and loss of adventitial fat.1 A diagnosis of chronic radiation dermatitis was inconsistent with our patient’s medical history and biopsy results, as pathology should reveal hyalinized collagen or stellate radiation fibroblasts.2,3 Nests of squamous epithelial cells with abundant eosinophilic cytoplasm and large vesicular nuclei were not seen, excluding squamous cell carcinoma as a possible diagnosis.4 Although sclerosing sweat duct carcinoma is characterized by infiltrating cords in sclerotic dermis, the cells were not arranged in ductlike structures 1– to 2–cell layers thick, excluding this diagnosis.5

Carcinoma en cuirasse—named for skin involvement that appears similar to the metal breastplate of a cuirassier—is a rare form of cutaneous metastasis that typically presents with extensive infiltrative plaques resulting in fibrosis of the skin and subcutaneous tissue.6,7 Carcinoma en cuirasse most commonly metastasizes from the breast but also may represent metastases from the lungs, gastrointestinal tract, or genitourinary systems.8 In the setting of a primary breast malignancy, metastatic plaques of CeC tend to represent tumor recurrence following a mastectomy procedure; however, in rare cases CeC can present as the primary manifestation of breast cancer or as a result of untreated malignancy.6,9 In our patient, CeC was the primary manifestation of metastatic invasive ductal carcinoma with additional paraneoplastic ichthyosis (Figure 2).

Ichthyotic plaques with brown scaling on the leg.
FIGURE 2. Ichthyotic plaques with brown scaling on the leg.

Carcinoma en cuirasse comprises 3% to 6% of cutaneous metastases originating from the breast.10,11 Breast cancer is the most common primary neoplasm displaying extracutaneous metastasis, comprising 70% of all cutaneous metastases in females.11 Cutaneous metastasis often indicates late stage of disease, portending a poor prognosis. In our patient, the cutaneous nodules were present for approximately 3 years prior to the diagnosis of stage IV invasive ductal cell carcinoma with metastasis to the skin and lungs. Prior to admission, she had not been diagnosed with breast cancer, thus no treatments had been administered. It is uncommon for CeC to present as the initial finding and without prior treatment of the underlying malignancy. The median length of survival after diagnosis of cutaneous metastasis from breast cancer is 13.8 months, with a 10-year survival rate of 3.1%.12

In addition to cutaneous metastasis, breast cancer also may present with paraneoplastic dermatoses such as ichthyosis.13 Ichthyosis is characterized by extreme dryness, flaking, thickening, and mild pruritus.14 It most commonly is an inherited condition, but it may be acquired due to malignancy. Acquired ichthyosis may manifest in systemic diseases including systemic lupus erythematosus, sarcoidosis, and hypothyroidism.15 Although acquired ichthyosis is rare, it has been reported in cases of internal malignancy, most commonly lymphoproliferative malignancies and less frequently carcinoma of the breasts, cervix, and lungs. Patients who acquire ichthyosis in association with malignancy usually present with late-stage disease.15 Our patient acquired ichthyosis 3 months prior to admission and had never experienced it previously. Although the exact mechanism for acquiring ichthyosis remains unknown, it is uncertain if ichthyosis associated with malignancy is paraneoplastic or a result of chemotherapy.14,16 In this case, the patient had not yet started chemotherapy at the time of the ichthyosis diagnosis, suggesting a paraneoplastic etiology.

Carcinoma en cuirasse and paraneoplastic ichthyosis individually are extremely rare manifestations of breast cancer. Thus, it is even rarer for these conditions to present concurrently. Treatment options for CeC include chemotherapy, radiotherapy, hormonal antagonists, and snake venom.11 Systemic chemotherapy targeting the histopathologic type of the primary tumor is the treatment of choice. Other treatment methods usually are chosen for late stages of disease progression.10 Paraneoplastic ichthyosis has been reported to show improvement with treatment of the underlying primary malignancy by surgical removal or chemotherapy.14,17 Tamoxifen less commonly is used for systemic treatment of CeC, but one case in the literature reported favorable outcomes.18

We describe 2 rare cutaneous manifestations of breast cancer occurring concomitantly: CeC and paraneoplastic ichthyosis. The combination of clinical and pathologic findings presented in this case solidified the diagnosis of metastatic invasive ductal carcinoma. We aim to improve recognition of paraneoplastic skin findings to accelerate the process of effective and efficient treatment.

References
  1. Walker D, Susa JS, Currimbhoy S, et al. Histopathological changes in morphea and their clinical correlates: results from the Morphea in Adults and Children Cohort V. J Am Acad Dermatol. 2017;76:1124-1130. https://doi.org/10.1016/j.jaad.2016.12.020
  2. Borrelli MR, Shen AH, Lee GK, et al. Radiation-induced skin fibrosis: pathogenesis, current treatment options, and emerging therapeutics. Ann Plast Surg. 2019;83(4 suppl 1):S59-S64. https://doi.org/10.1097/SAP.0000000000002098
  3. Boncher J, Bergfeld WF. Fluoroscopy-induced chronic radiation dermatitis: a report of two additional cases and a brief review of the literature. J Cutan Pathol. 2012;39:63-67. https://doi.org/10.1111/j .1600-0560.2011.01754.x
  4. Cassarino DS, Derienzo DP, Barr RJ. Cutaneous squamous cell carcinoma: a comprehensive clinicopathologic classification. part one. J Cutan Pathol. 2006;33:191-206. https://doi.org/10.1111 /j.0303-6987.2006.00516_1.x
  5. Harvey DT, Hu J, Long JA, et al. Sclerosing sweat duct carcinoma of the lower extremity treated with Mohs micrographic surgery. JAAD Case Rep. 2016;2:284-286. https://doi.org/10.1016/j.jdcr.2016.05.017
  6. Sharma V, Kumar A. Carcinoma en cuirasse. N Engl J Med. 2021;385:2562. doi:10.1056/NEJMicm2111669
  7. Oliveira GM, Zachetti DB, Barros HR, et al. Breast carcinoma en cuirasse—case report. An Bras Dermatol. 2013;88:608-610. doi:10.1590/abd1806-4841.20131926
  8. Alcaraz I, Cerroni L, Rütten A, et al. Cutaneous metastases from internal malignancies: a clinicopathologic and immunohistochemical review. Am J Dermatopathol. 2012;34:347-393. doi:10.1097 /DAD.0b013e31823069cf
  9. Glazebrook AJ, Tomaszewski W. Ichthyosiform atrophy of the skin in Hodgkin’s disease: report of a case, with reference to vitamin A metabolism. Arch Derm Syphilol. 1944;50:85-89. doi:10.1001 /archderm.1944.01510140008002
  10. Mordenti C, Concetta F, Cerroni M, et al. Cutaneous metastatic breast carcinoma: a study of 164 patients. Acta Dermatovenerol Alp Pannonica Adriat. 2000;9:143-148.
  11. Culver AL, Metter DM, Pippen JE Jr. Carcinoma en cuirasse. Proc (Bayl Univ Med Cent). 2019;32:263-265. doi:10.1080/08998280.2018.1564966
  12. Schoenlaub P, Sarraux A, Grosshans E, et al. Survival after cutaneous metastasis: a study of 200 cases [in French]. Ann Dermatol Venereol. 2001;128:1310-1315.
  13. Tan AR. Cutaneous manifestations of breast cancer. Semin Oncol. 2016;43:331-334. doi:10.1053/j.seminoncol.2016.02.030
  14. Song Y, Wu Y, Fan T. Dermatosis as the initial manifestation of malignant breast tumors: retrospective analysis of 4 cases. Breast Care. 2010;5:174-176. doi:10.1159/000314265
  15. Polisky RB, Bronson DM. Acquired ichthyosis in a patient with adenocarcinoma of the breast. Cutis. 1986;38:359-360.
  16. Haste AR. Acquired ichthyosis from breast cancer. Br Med J. 1967;4:96-98.
  17. Riesco Martínez MC, Muñoz Martín AJ, Zamberk Majlis P, et al. Acquired ichthyosis as a paraneoplastic syndrome in Hodgkin’s disease. Clin Transl Oncol. 2009;11:552-553. doi:10.1007/s12094-009-0402-2
  18. Siddiqui MA, Zaman MN. Primary carcinoma en cuirasse. J Am Geriatr Soc. 1996;44:221-222. doi:10.1111/j.1532-5415.1996.tb02455.xssss
References
  1. Walker D, Susa JS, Currimbhoy S, et al. Histopathological changes in morphea and their clinical correlates: results from the Morphea in Adults and Children Cohort V. J Am Acad Dermatol. 2017;76:1124-1130. https://doi.org/10.1016/j.jaad.2016.12.020
  2. Borrelli MR, Shen AH, Lee GK, et al. Radiation-induced skin fibrosis: pathogenesis, current treatment options, and emerging therapeutics. Ann Plast Surg. 2019;83(4 suppl 1):S59-S64. https://doi.org/10.1097/SAP.0000000000002098
  3. Boncher J, Bergfeld WF. Fluoroscopy-induced chronic radiation dermatitis: a report of two additional cases and a brief review of the literature. J Cutan Pathol. 2012;39:63-67. https://doi.org/10.1111/j .1600-0560.2011.01754.x
  4. Cassarino DS, Derienzo DP, Barr RJ. Cutaneous squamous cell carcinoma: a comprehensive clinicopathologic classification. part one. J Cutan Pathol. 2006;33:191-206. https://doi.org/10.1111 /j.0303-6987.2006.00516_1.x
  5. Harvey DT, Hu J, Long JA, et al. Sclerosing sweat duct carcinoma of the lower extremity treated with Mohs micrographic surgery. JAAD Case Rep. 2016;2:284-286. https://doi.org/10.1016/j.jdcr.2016.05.017
  6. Sharma V, Kumar A. Carcinoma en cuirasse. N Engl J Med. 2021;385:2562. doi:10.1056/NEJMicm2111669
  7. Oliveira GM, Zachetti DB, Barros HR, et al. Breast carcinoma en cuirasse—case report. An Bras Dermatol. 2013;88:608-610. doi:10.1590/abd1806-4841.20131926
  8. Alcaraz I, Cerroni L, Rütten A, et al. Cutaneous metastases from internal malignancies: a clinicopathologic and immunohistochemical review. Am J Dermatopathol. 2012;34:347-393. doi:10.1097 /DAD.0b013e31823069cf
  9. Glazebrook AJ, Tomaszewski W. Ichthyosiform atrophy of the skin in Hodgkin’s disease: report of a case, with reference to vitamin A metabolism. Arch Derm Syphilol. 1944;50:85-89. doi:10.1001 /archderm.1944.01510140008002
  10. Mordenti C, Concetta F, Cerroni M, et al. Cutaneous metastatic breast carcinoma: a study of 164 patients. Acta Dermatovenerol Alp Pannonica Adriat. 2000;9:143-148.
  11. Culver AL, Metter DM, Pippen JE Jr. Carcinoma en cuirasse. Proc (Bayl Univ Med Cent). 2019;32:263-265. doi:10.1080/08998280.2018.1564966
  12. Schoenlaub P, Sarraux A, Grosshans E, et al. Survival after cutaneous metastasis: a study of 200 cases [in French]. Ann Dermatol Venereol. 2001;128:1310-1315.
  13. Tan AR. Cutaneous manifestations of breast cancer. Semin Oncol. 2016;43:331-334. doi:10.1053/j.seminoncol.2016.02.030
  14. Song Y, Wu Y, Fan T. Dermatosis as the initial manifestation of malignant breast tumors: retrospective analysis of 4 cases. Breast Care. 2010;5:174-176. doi:10.1159/000314265
  15. Polisky RB, Bronson DM. Acquired ichthyosis in a patient with adenocarcinoma of the breast. Cutis. 1986;38:359-360.
  16. Haste AR. Acquired ichthyosis from breast cancer. Br Med J. 1967;4:96-98.
  17. Riesco Martínez MC, Muñoz Martín AJ, Zamberk Majlis P, et al. Acquired ichthyosis as a paraneoplastic syndrome in Hodgkin’s disease. Clin Transl Oncol. 2009;11:552-553. doi:10.1007/s12094-009-0402-2
  18. Siddiqui MA, Zaman MN. Primary carcinoma en cuirasse. J Am Geriatr Soc. 1996;44:221-222. doi:10.1111/j.1532-5415.1996.tb02455.xssss
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
E12-E14
Page Number
E12-E14
Publications
Publications
Topics
Article Type
Display Headline
Large Indurated Plaque on the Chest With Ulceration and Necrosis
Display Headline
Large Indurated Plaque on the Chest With Ulceration and Necrosis
Sections
Questionnaire Body

A 47-year-old woman with no notable medical history presented to the emergency department with shortness of breath on simple exertion as well as a large lesion on the chest that had slowly increased in size over the last 3 years. The lesion was not painful or pruritic, and she had been treating it with topical emollients without substantial improvement. Physical examination revealed a large indurated plaque with areas of ulceration and necrosis spanning the mid to lateral chest. Additionally, ichthyotic brown scaling was present on the arms and legs. Upon further questioning, the patient reported that the scales on the extremities appeared in the last 3 months and were not previously noted. She had no recent routine cancer screenings, and her family history was notable for a brother with brain cancer. A punch biopsy of the chest plaque was performed.

Large indurated plaque on the chest with ulceration and necrosis

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 12/13/2023 - 08:30
Un-Gate On Date
Wed, 12/13/2023 - 08:30
Use ProPublica
CFC Schedule Remove Status
Wed, 12/13/2023 - 08:30
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

An 18-month-old male presents with a red mark on the forehead and nose

Article Type
Changed
Mon, 12/11/2023 - 16:12

Following the initial presentation, the lesion was initially considered an acquired port wine stain and the child was referred for laser treatment. Upon reassessment during laser treatment a few months later, the lesion had progressed to hyper- and hypopigmented plaques with associated tissue sclerosis and bone atrophy on the mid forehead, nose, and scalp. Patches of alopecia and atrophy were observed on the frontal scalp. The diagnosis was revised to linear morphea en coup de sabre and the child was referred to pediatric rheumatology and commenced treatment with methotrexate and oral corticosteroids.

Dr. Catalina Matiz, a pediatric dermatologist at Southern California Permanente Medical Group, San Diego
Dr. Catalina Matiz

Linear morphea, a rare connective tissue disorder, primarily affects girls in the first 2 decades of life. Lesions can initially present in many ways. Usually, they present as hypo- or hyperpigmented patches, but may also present as lichenoid uncolored or pink plaques resembling lichen striatus. There may also be erythematous patches mimicking a capillary malformation, as seen in our patient. A recent article reviewing the progression of the lesions from erythematous patches to sclerosis suggests it occurs between 3 and 7 months of age. Subsequent stages manifest as significant atrophy, hypo- and hyperpigmentation, and in severe cases, bone atrophy and deformity, often causing substantial cosmetic disfigurement and functional impairment.

Pathophysiologically, linear morphea involves a complex interplay of immunologic, vascular, and fibrotic processes. While the initial triggers remain elusive, dysregulated immune responses leading to endothelial injury, subsequent activation of fibroblasts and myofibroblasts, and excessive collagen deposition are implicated. Angiogenic disturbances exacerbate tissue ischemia, perpetuating the fibrotic cascade. Alterations in cytokine signaling pathways, particularly TGF-beta and interleukin-6, play pivotal roles in promoting fibrosis and modulating the inflammatory milieu.

Diagnosis of linear morphea en coup de sabre relies on clinical examination, imaging (ultrasonography, MRI, CT scan), and skin biopsy for histopathological analysis. Imaging helps evaluate tissue involvement, while histology reveals characteristic dermal sclerosis, collagen deposition, and inflammation. Early-stage histology may show telangiectatic changes, complicating its differentiation from capillary malformation.

Treatment aims to mitigate symptoms, halt disease progression, and improve cosmesis and functionality. This involves a multidisciplinary approach with systemic medications, phototherapy, physical therapy, and surgical interventions in severe cases. Early identification is crucial for systemic treatments such as methotrexate and systemic corticosteroids to arrest disease progression. Other adjunctive therapies include topical corticosteroids, calcineurin inhibitors, and phototherapy. Surgical procedures like tissue expansion or autologous fat grafting may address tissue atrophy and deformities.

Linear morphea en coup de sabre presents diagnostic and therapeutic challenges because of its rarity and variable clinical course. Collaborative efforts among dermatologists, rheumatologists, radiologists, and surgeons are essential for accurate diagnosis, evaluation, and tailored management. Continued research into pathogenesis and novel therapeutic agents is pivotal to enhance understanding and improve outcomes for those affected by this enigmatic dermatologic condition.

Dr. Matiz is a pediatric dermatologist at Southern California Permanente Medical Group, San Diego.

References

Gomez-Garcia LA et al. Pediatr Dermatol. 2022 Mar;39(2):275-80.

Ng SS, Tay YK. J Cosmet Laser Ther. 2015;17(5):277-80.

Nijhawan RI et al. J Am Acad Dermatol. 2011 Apr;64(4):779-82.

Publications
Topics
Sections

Following the initial presentation, the lesion was initially considered an acquired port wine stain and the child was referred for laser treatment. Upon reassessment during laser treatment a few months later, the lesion had progressed to hyper- and hypopigmented plaques with associated tissue sclerosis and bone atrophy on the mid forehead, nose, and scalp. Patches of alopecia and atrophy were observed on the frontal scalp. The diagnosis was revised to linear morphea en coup de sabre and the child was referred to pediatric rheumatology and commenced treatment with methotrexate and oral corticosteroids.

Dr. Catalina Matiz, a pediatric dermatologist at Southern California Permanente Medical Group, San Diego
Dr. Catalina Matiz

Linear morphea, a rare connective tissue disorder, primarily affects girls in the first 2 decades of life. Lesions can initially present in many ways. Usually, they present as hypo- or hyperpigmented patches, but may also present as lichenoid uncolored or pink plaques resembling lichen striatus. There may also be erythematous patches mimicking a capillary malformation, as seen in our patient. A recent article reviewing the progression of the lesions from erythematous patches to sclerosis suggests it occurs between 3 and 7 months of age. Subsequent stages manifest as significant atrophy, hypo- and hyperpigmentation, and in severe cases, bone atrophy and deformity, often causing substantial cosmetic disfigurement and functional impairment.

Pathophysiologically, linear morphea involves a complex interplay of immunologic, vascular, and fibrotic processes. While the initial triggers remain elusive, dysregulated immune responses leading to endothelial injury, subsequent activation of fibroblasts and myofibroblasts, and excessive collagen deposition are implicated. Angiogenic disturbances exacerbate tissue ischemia, perpetuating the fibrotic cascade. Alterations in cytokine signaling pathways, particularly TGF-beta and interleukin-6, play pivotal roles in promoting fibrosis and modulating the inflammatory milieu.

Diagnosis of linear morphea en coup de sabre relies on clinical examination, imaging (ultrasonography, MRI, CT scan), and skin biopsy for histopathological analysis. Imaging helps evaluate tissue involvement, while histology reveals characteristic dermal sclerosis, collagen deposition, and inflammation. Early-stage histology may show telangiectatic changes, complicating its differentiation from capillary malformation.

Treatment aims to mitigate symptoms, halt disease progression, and improve cosmesis and functionality. This involves a multidisciplinary approach with systemic medications, phototherapy, physical therapy, and surgical interventions in severe cases. Early identification is crucial for systemic treatments such as methotrexate and systemic corticosteroids to arrest disease progression. Other adjunctive therapies include topical corticosteroids, calcineurin inhibitors, and phototherapy. Surgical procedures like tissue expansion or autologous fat grafting may address tissue atrophy and deformities.

Linear morphea en coup de sabre presents diagnostic and therapeutic challenges because of its rarity and variable clinical course. Collaborative efforts among dermatologists, rheumatologists, radiologists, and surgeons are essential for accurate diagnosis, evaluation, and tailored management. Continued research into pathogenesis and novel therapeutic agents is pivotal to enhance understanding and improve outcomes for those affected by this enigmatic dermatologic condition.

Dr. Matiz is a pediatric dermatologist at Southern California Permanente Medical Group, San Diego.

References

Gomez-Garcia LA et al. Pediatr Dermatol. 2022 Mar;39(2):275-80.

Ng SS, Tay YK. J Cosmet Laser Ther. 2015;17(5):277-80.

Nijhawan RI et al. J Am Acad Dermatol. 2011 Apr;64(4):779-82.

Following the initial presentation, the lesion was initially considered an acquired port wine stain and the child was referred for laser treatment. Upon reassessment during laser treatment a few months later, the lesion had progressed to hyper- and hypopigmented plaques with associated tissue sclerosis and bone atrophy on the mid forehead, nose, and scalp. Patches of alopecia and atrophy were observed on the frontal scalp. The diagnosis was revised to linear morphea en coup de sabre and the child was referred to pediatric rheumatology and commenced treatment with methotrexate and oral corticosteroids.

Dr. Catalina Matiz, a pediatric dermatologist at Southern California Permanente Medical Group, San Diego
Dr. Catalina Matiz

Linear morphea, a rare connective tissue disorder, primarily affects girls in the first 2 decades of life. Lesions can initially present in many ways. Usually, they present as hypo- or hyperpigmented patches, but may also present as lichenoid uncolored or pink plaques resembling lichen striatus. There may also be erythematous patches mimicking a capillary malformation, as seen in our patient. A recent article reviewing the progression of the lesions from erythematous patches to sclerosis suggests it occurs between 3 and 7 months of age. Subsequent stages manifest as significant atrophy, hypo- and hyperpigmentation, and in severe cases, bone atrophy and deformity, often causing substantial cosmetic disfigurement and functional impairment.

Pathophysiologically, linear morphea involves a complex interplay of immunologic, vascular, and fibrotic processes. While the initial triggers remain elusive, dysregulated immune responses leading to endothelial injury, subsequent activation of fibroblasts and myofibroblasts, and excessive collagen deposition are implicated. Angiogenic disturbances exacerbate tissue ischemia, perpetuating the fibrotic cascade. Alterations in cytokine signaling pathways, particularly TGF-beta and interleukin-6, play pivotal roles in promoting fibrosis and modulating the inflammatory milieu.

Diagnosis of linear morphea en coup de sabre relies on clinical examination, imaging (ultrasonography, MRI, CT scan), and skin biopsy for histopathological analysis. Imaging helps evaluate tissue involvement, while histology reveals characteristic dermal sclerosis, collagen deposition, and inflammation. Early-stage histology may show telangiectatic changes, complicating its differentiation from capillary malformation.

Treatment aims to mitigate symptoms, halt disease progression, and improve cosmesis and functionality. This involves a multidisciplinary approach with systemic medications, phototherapy, physical therapy, and surgical interventions in severe cases. Early identification is crucial for systemic treatments such as methotrexate and systemic corticosteroids to arrest disease progression. Other adjunctive therapies include topical corticosteroids, calcineurin inhibitors, and phototherapy. Surgical procedures like tissue expansion or autologous fat grafting may address tissue atrophy and deformities.

Linear morphea en coup de sabre presents diagnostic and therapeutic challenges because of its rarity and variable clinical course. Collaborative efforts among dermatologists, rheumatologists, radiologists, and surgeons are essential for accurate diagnosis, evaluation, and tailored management. Continued research into pathogenesis and novel therapeutic agents is pivotal to enhance understanding and improve outcomes for those affected by this enigmatic dermatologic condition.

Dr. Matiz is a pediatric dermatologist at Southern California Permanente Medical Group, San Diego.

References

Gomez-Garcia LA et al. Pediatr Dermatol. 2022 Mar;39(2):275-80.

Ng SS, Tay YK. J Cosmet Laser Ther. 2015;17(5):277-80.

Nijhawan RI et al. J Am Acad Dermatol. 2011 Apr;64(4):779-82.

Publications
Publications
Topics
Article Type
Sections
Questionnaire Body

Hyperpigmented atrophic plaques on the nose left forehead and frontal scalp and left lateral cheek.
An 18-month-old male presented at the pediatric dermatology clinic with a 3-month history of a red mark on the left side of the forehead and nose. The mother noted a small mark at birth in that area, but it had intensified significantly over time. The lesion had become more pronounced in the past few weeks and did not exhibit variations throughout the day. There was no reported history of trauma to the area, and while the area had become pinker in the last few months, it did not cause discomfort to the child. In addition, the mother observed small areas of hair loss on the right frontal scalp. The child, delivered via normal vaginal delivery, showed normal growth and met developmental milestones. No pain, headaches, or history of seizures were reported, and the child had received no prior treatments. The mother had a history of alopecia areata. 
On examination, a faint pink patch was observed on the right forehead, frontal scalp, and nose. The lesion paled under pressure, with small areas of hair loss on the scalp. No atrophy was noted.

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Painful Growing Nodule on the Right Calf

Article Type
Changed
Tue, 12/05/2023 - 16:49
Display Headline
Painful Growing Nodule on the Right Calf

The Diagnosis: Merkel Cell Carcinoma

Multiple diagnoses should be considered for a small, round, blue cell neoplasm of the skin, including both primary and metastatic entities. In our patient, histopathology revealed sheets and nests of infiltrative neoplastic cells with dispersed chromatin, minimal cytoplasm, and multiple mitoses (quiz image 1).1 The lesional cells were in the dermis and superficial subcutaneous tissue but did not appear to be arising from the epidermis. Lymphovascular invasion also was evident on additional sections. Metastatic disease was identified in 3 sentinel lymph nodes from the right inguinal and right iliac regions. These features were compatible with a diagnosis of Merkel cell carcinoma (MCC).

Merkel cell carcinoma is a rare malignant neuroendocrine cutaneous tumor with a worldwide incidence of 0.1 to 1.6 cases per 100,000 individuals annually.2 The typical patient is older than 75 years with fair skin and a history of extensive sun exposure. Immunocompromised individuals are predisposed and more susceptible to infection with the Merkel cell polyomavirus, which promotes oncogenesis in the majority of MCCs. Our patient’s history of combined variable immunodeficiency likely explains her presentation at a younger age.

The prognosis in patients with MCC is poor, with 5-year survival rates of 51% for local disease, 35% for nodal disease, and 14% for systemic metastases. Survival also is reduced in cases with head/ neck primary tumors and polyomavirus-negative tumors, as well as in immunocompromised patients.2 Treatment of resectable MCC consists of Mohs micrographic surgery or wide local excision depending on the patient’s cosmetic concerns. Radiation therapy is recommended for cases with increased risk for recurrence or positive surgical margins, as well as when additional resection is impossible. A study investigating immunotherapy with nivolumab demonstrated complete pathologic response and radiographic tumor regression in nearly half of patients when given 4 weeks prior to surgery.3

Immunohistochemistry is essential in discerning MCC from other small blue cell tumors. Most MCC cases show positive expression of neuroendocrine markers such as synaptophysin, chromogranin, and insulinomaassociated protein 1. Perinuclear dotlike staining with cytokeratin (CK) 20 (quiz image 2) commonly is seen, but up to 15% of cases may be CK20 negative. Many of these CK20-negative cases also express CK7. This tumor also may stain with paired box 5 (PAX-5), CD99, terminal deoxynucleotidyl transferase, Ber-EP4, and CD1171,4; melanoma stains (ie, human melanoma black [HMB] 45, SRYrelated HMB-box 10 [SOX-10], S-100, melanoma antigen recognized by T-cells 1 [MART-1]) should be negative. However, PAX-5 expression may be a potential pitfall given that B-cell lymphomas also would express that marker and could mimic MCC histologically. Therefore, other universal lymphoid markers such as CD45 should be ordered to rule out this entity. Even with one or a few aberrant stains, a diagnosis of MCC still can be rendered using the histomorphology and the overall staining profile.4 Of prognostic significance, p63 expression is associated with more aggressive tumors, while Bcl-2 expression is favorable, as it offers an additional targeted treatment option.5,6

Basal cell carcinoma (BCC) is linked to excessive sun exposure and is the most common skin cancer. Similar to MCC, it typically is mitotically active and hyperchromatic; however, lymphovascular invasion or metastasis almost never is observed in BCC, whereas approximately one-third of MCC cases have metastasized by the time of diagnosis. Additionally, BCC lacks the perinuclear dotlike staining seen with CK20.2,7 Features present in BCC that are unusual for MCC include peripheral nuclear palisading, mucin, and retraction artifact on paraffin-embedded sections (Figure 1).7

Basal cell carcinoma
FIGURE 1. Basal cell carcinoma. Nodular growth with classic peripheral nuclear palisading, retraction, and focal mucin (H&E, original magnification ×200).

Leukemia cutis (or cutaneous infiltrates of leukemia) commonly displays a perivascular and periadnexal pattern in the dermis and subcutis. These infiltrates of neoplastic leukocytes can congregate into sheets, sometimes with an overlying Grenz zone, or form single-file infiltrates (Figure 2).1,4 The neoplastic cells can be monomorphic or atypical and commonly are susceptible to crush artifact.4 Although the immunohistochemical profile varies depending on the etiology of the underlying leukemia, broad hematologic markers such as CD43 and CD45 are helpful to discern these malignancies from MCC.4

Leukemia cutis
FIGURE 2. Leukemia cutis. Infiltration of metastatic leukemia cells in the dermis with a single-file infiltration pattern and atypical nuclei (H&E, original magnification ×400).

Being neuroendocrine in origin, metastatic small cell carcinoma (Figure 3) strongly mimics MCC histologically and usually stains with synaptophysin, chromogranin, and insulinoma-associated protein 1. Both tumor cells typically exhibit nuclear molding and high mitotic rates. Although small cell carcinoma is more likely to stain with high-molecular-weight cytokeratins (ie, CK7), it is not uncommon for these tumors to express lowmolecular- weight cytokeratins such as CK20. Because most cases originate from the lungs, these lesions should be positive for thyroid transcription factor 1 and negative for PAX-5, whereas MCC would show the reverse for those stains.1 Ultimately, however, clinical correlation with imaging results is the single best methodology for differentiation.

Metastatic small cell carcinoma
FIGURE 3. Metastatic small cell carcinoma. Sheets of infiltrative basophilic cells with fine chromatin, nuclear molding, and brisk mitoses (H&E, original magnification ×200).

Small cell melanoma, a variant of nevoid melanoma, can strongly resemble an MCC or a lymphoma. Usually located on the scalp or arising from a congenital nevus, small cell melanomas are aggressive and confer an unfavorable prognosis. Histologically, they consist of nests to sheets of atypical cells within the epidermis and dermis. These cells typically exhibit hyperchromatic nuclei, minimal cytoplasm, and frequent mitoses (Figure 4). Furthermore, the cells do not display maturation based on depth.8 These tumors usually are positive for HMB45, S-100, MART-1, SOX-10, and tyrosinase, all of which are extremely unlikely to stain an MCC.1

Small cell melanoma
FIGURE 4. Small cell melanoma. Infiltrative nests and individual cells involving the epidermis and dermis (H&E, original magnification ×400).

References
  1. Patterson JW, Hosler GA. Weedon’s Skin Pathology. 4th ed. Churchill Livingstone/Elsevier; 2016.
  2. Walsh NM, Cerroni L. Merkel cell carcinoma: a review. J Cutan Pathol. 2021;48:411-421.
  3. Topalian SL, Bhatia S, Amin A, et al. Neoadjuvant nivolumab for patients with resectable Merkel cell carcinoma in the CheckMate 358 Trial. J Clin Oncol. 2020;38:2476-2488.
  4. Rapini RP. Practical Dermatopathology. 3rd ed. Elsevier; 2021.
  5. Asioli S, Righi A, Volante M, et al. p63 expression as a new prognostic marker in Merkel cell carcinoma. Cancer. 2007;110:640-647.
  6. Verhaegen ME, Mangelberger D, Weick JW, et al. Merkel cell carcinoma dependence on Bcl-2 family members for survival. J Invest Dermatol. 2014;134:2241-2250.
  7. Le MD, O’Steen LH, Cassarino DS. A rare case of CK20/CK7 double negative Merkel cell carcinoma. Am J Dermatopathol. 2017;39:208-211.
  8. North JP, Bastian BC, Lazar AJ. Melanoma. In: Calonje E, Brenn T, Lazar AJ, et al, eds. McKee’s Pathology of the Skin With Clinical Correlations. 5th ed. Elsevier; 2020.
Article PDF
Author and Disclosure Information

From the Department of Pathology, San Antonio Military Medical Center, Fort Sam Houston, Texas.

The authors report no conflict of interest.

The views expressed herein are those of the authors and do not reflect the official policy or position of Brooke Army Medical Center, the US Army Medical Department, the US Army Office of the Surgeon General, the Department of the Army, the Department of the Air Force, or the Department of Defense of the US Government.

Correspondence: Jesse Lee Fitzgerald, DO, San Antonio Military Medical Center, 3551 Roger Brooke Dr, Fort Sam Houston, TX 78234 (jesse.l.fitzgerald3.mil@health.mil).

Issue
Cutis - 112(6)
Publications
Topics
Page Number
287,297-298
Sections
Author and Disclosure Information

From the Department of Pathology, San Antonio Military Medical Center, Fort Sam Houston, Texas.

The authors report no conflict of interest.

The views expressed herein are those of the authors and do not reflect the official policy or position of Brooke Army Medical Center, the US Army Medical Department, the US Army Office of the Surgeon General, the Department of the Army, the Department of the Air Force, or the Department of Defense of the US Government.

Correspondence: Jesse Lee Fitzgerald, DO, San Antonio Military Medical Center, 3551 Roger Brooke Dr, Fort Sam Houston, TX 78234 (jesse.l.fitzgerald3.mil@health.mil).

Author and Disclosure Information

From the Department of Pathology, San Antonio Military Medical Center, Fort Sam Houston, Texas.

The authors report no conflict of interest.

The views expressed herein are those of the authors and do not reflect the official policy or position of Brooke Army Medical Center, the US Army Medical Department, the US Army Office of the Surgeon General, the Department of the Army, the Department of the Air Force, or the Department of Defense of the US Government.

Correspondence: Jesse Lee Fitzgerald, DO, San Antonio Military Medical Center, 3551 Roger Brooke Dr, Fort Sam Houston, TX 78234 (jesse.l.fitzgerald3.mil@health.mil).

Article PDF
Article PDF
Related Articles

The Diagnosis: Merkel Cell Carcinoma

Multiple diagnoses should be considered for a small, round, blue cell neoplasm of the skin, including both primary and metastatic entities. In our patient, histopathology revealed sheets and nests of infiltrative neoplastic cells with dispersed chromatin, minimal cytoplasm, and multiple mitoses (quiz image 1).1 The lesional cells were in the dermis and superficial subcutaneous tissue but did not appear to be arising from the epidermis. Lymphovascular invasion also was evident on additional sections. Metastatic disease was identified in 3 sentinel lymph nodes from the right inguinal and right iliac regions. These features were compatible with a diagnosis of Merkel cell carcinoma (MCC).

Merkel cell carcinoma is a rare malignant neuroendocrine cutaneous tumor with a worldwide incidence of 0.1 to 1.6 cases per 100,000 individuals annually.2 The typical patient is older than 75 years with fair skin and a history of extensive sun exposure. Immunocompromised individuals are predisposed and more susceptible to infection with the Merkel cell polyomavirus, which promotes oncogenesis in the majority of MCCs. Our patient’s history of combined variable immunodeficiency likely explains her presentation at a younger age.

The prognosis in patients with MCC is poor, with 5-year survival rates of 51% for local disease, 35% for nodal disease, and 14% for systemic metastases. Survival also is reduced in cases with head/ neck primary tumors and polyomavirus-negative tumors, as well as in immunocompromised patients.2 Treatment of resectable MCC consists of Mohs micrographic surgery or wide local excision depending on the patient’s cosmetic concerns. Radiation therapy is recommended for cases with increased risk for recurrence or positive surgical margins, as well as when additional resection is impossible. A study investigating immunotherapy with nivolumab demonstrated complete pathologic response and radiographic tumor regression in nearly half of patients when given 4 weeks prior to surgery.3

Immunohistochemistry is essential in discerning MCC from other small blue cell tumors. Most MCC cases show positive expression of neuroendocrine markers such as synaptophysin, chromogranin, and insulinomaassociated protein 1. Perinuclear dotlike staining with cytokeratin (CK) 20 (quiz image 2) commonly is seen, but up to 15% of cases may be CK20 negative. Many of these CK20-negative cases also express CK7. This tumor also may stain with paired box 5 (PAX-5), CD99, terminal deoxynucleotidyl transferase, Ber-EP4, and CD1171,4; melanoma stains (ie, human melanoma black [HMB] 45, SRYrelated HMB-box 10 [SOX-10], S-100, melanoma antigen recognized by T-cells 1 [MART-1]) should be negative. However, PAX-5 expression may be a potential pitfall given that B-cell lymphomas also would express that marker and could mimic MCC histologically. Therefore, other universal lymphoid markers such as CD45 should be ordered to rule out this entity. Even with one or a few aberrant stains, a diagnosis of MCC still can be rendered using the histomorphology and the overall staining profile.4 Of prognostic significance, p63 expression is associated with more aggressive tumors, while Bcl-2 expression is favorable, as it offers an additional targeted treatment option.5,6

Basal cell carcinoma (BCC) is linked to excessive sun exposure and is the most common skin cancer. Similar to MCC, it typically is mitotically active and hyperchromatic; however, lymphovascular invasion or metastasis almost never is observed in BCC, whereas approximately one-third of MCC cases have metastasized by the time of diagnosis. Additionally, BCC lacks the perinuclear dotlike staining seen with CK20.2,7 Features present in BCC that are unusual for MCC include peripheral nuclear palisading, mucin, and retraction artifact on paraffin-embedded sections (Figure 1).7

Basal cell carcinoma
FIGURE 1. Basal cell carcinoma. Nodular growth with classic peripheral nuclear palisading, retraction, and focal mucin (H&E, original magnification ×200).

Leukemia cutis (or cutaneous infiltrates of leukemia) commonly displays a perivascular and periadnexal pattern in the dermis and subcutis. These infiltrates of neoplastic leukocytes can congregate into sheets, sometimes with an overlying Grenz zone, or form single-file infiltrates (Figure 2).1,4 The neoplastic cells can be monomorphic or atypical and commonly are susceptible to crush artifact.4 Although the immunohistochemical profile varies depending on the etiology of the underlying leukemia, broad hematologic markers such as CD43 and CD45 are helpful to discern these malignancies from MCC.4

Leukemia cutis
FIGURE 2. Leukemia cutis. Infiltration of metastatic leukemia cells in the dermis with a single-file infiltration pattern and atypical nuclei (H&E, original magnification ×400).

Being neuroendocrine in origin, metastatic small cell carcinoma (Figure 3) strongly mimics MCC histologically and usually stains with synaptophysin, chromogranin, and insulinoma-associated protein 1. Both tumor cells typically exhibit nuclear molding and high mitotic rates. Although small cell carcinoma is more likely to stain with high-molecular-weight cytokeratins (ie, CK7), it is not uncommon for these tumors to express lowmolecular- weight cytokeratins such as CK20. Because most cases originate from the lungs, these lesions should be positive for thyroid transcription factor 1 and negative for PAX-5, whereas MCC would show the reverse for those stains.1 Ultimately, however, clinical correlation with imaging results is the single best methodology for differentiation.

Metastatic small cell carcinoma
FIGURE 3. Metastatic small cell carcinoma. Sheets of infiltrative basophilic cells with fine chromatin, nuclear molding, and brisk mitoses (H&E, original magnification ×200).

Small cell melanoma, a variant of nevoid melanoma, can strongly resemble an MCC or a lymphoma. Usually located on the scalp or arising from a congenital nevus, small cell melanomas are aggressive and confer an unfavorable prognosis. Histologically, they consist of nests to sheets of atypical cells within the epidermis and dermis. These cells typically exhibit hyperchromatic nuclei, minimal cytoplasm, and frequent mitoses (Figure 4). Furthermore, the cells do not display maturation based on depth.8 These tumors usually are positive for HMB45, S-100, MART-1, SOX-10, and tyrosinase, all of which are extremely unlikely to stain an MCC.1

Small cell melanoma
FIGURE 4. Small cell melanoma. Infiltrative nests and individual cells involving the epidermis and dermis (H&E, original magnification ×400).

The Diagnosis: Merkel Cell Carcinoma

Multiple diagnoses should be considered for a small, round, blue cell neoplasm of the skin, including both primary and metastatic entities. In our patient, histopathology revealed sheets and nests of infiltrative neoplastic cells with dispersed chromatin, minimal cytoplasm, and multiple mitoses (quiz image 1).1 The lesional cells were in the dermis and superficial subcutaneous tissue but did not appear to be arising from the epidermis. Lymphovascular invasion also was evident on additional sections. Metastatic disease was identified in 3 sentinel lymph nodes from the right inguinal and right iliac regions. These features were compatible with a diagnosis of Merkel cell carcinoma (MCC).

Merkel cell carcinoma is a rare malignant neuroendocrine cutaneous tumor with a worldwide incidence of 0.1 to 1.6 cases per 100,000 individuals annually.2 The typical patient is older than 75 years with fair skin and a history of extensive sun exposure. Immunocompromised individuals are predisposed and more susceptible to infection with the Merkel cell polyomavirus, which promotes oncogenesis in the majority of MCCs. Our patient’s history of combined variable immunodeficiency likely explains her presentation at a younger age.

The prognosis in patients with MCC is poor, with 5-year survival rates of 51% for local disease, 35% for nodal disease, and 14% for systemic metastases. Survival also is reduced in cases with head/ neck primary tumors and polyomavirus-negative tumors, as well as in immunocompromised patients.2 Treatment of resectable MCC consists of Mohs micrographic surgery or wide local excision depending on the patient’s cosmetic concerns. Radiation therapy is recommended for cases with increased risk for recurrence or positive surgical margins, as well as when additional resection is impossible. A study investigating immunotherapy with nivolumab demonstrated complete pathologic response and radiographic tumor regression in nearly half of patients when given 4 weeks prior to surgery.3

Immunohistochemistry is essential in discerning MCC from other small blue cell tumors. Most MCC cases show positive expression of neuroendocrine markers such as synaptophysin, chromogranin, and insulinomaassociated protein 1. Perinuclear dotlike staining with cytokeratin (CK) 20 (quiz image 2) commonly is seen, but up to 15% of cases may be CK20 negative. Many of these CK20-negative cases also express CK7. This tumor also may stain with paired box 5 (PAX-5), CD99, terminal deoxynucleotidyl transferase, Ber-EP4, and CD1171,4; melanoma stains (ie, human melanoma black [HMB] 45, SRYrelated HMB-box 10 [SOX-10], S-100, melanoma antigen recognized by T-cells 1 [MART-1]) should be negative. However, PAX-5 expression may be a potential pitfall given that B-cell lymphomas also would express that marker and could mimic MCC histologically. Therefore, other universal lymphoid markers such as CD45 should be ordered to rule out this entity. Even with one or a few aberrant stains, a diagnosis of MCC still can be rendered using the histomorphology and the overall staining profile.4 Of prognostic significance, p63 expression is associated with more aggressive tumors, while Bcl-2 expression is favorable, as it offers an additional targeted treatment option.5,6

Basal cell carcinoma (BCC) is linked to excessive sun exposure and is the most common skin cancer. Similar to MCC, it typically is mitotically active and hyperchromatic; however, lymphovascular invasion or metastasis almost never is observed in BCC, whereas approximately one-third of MCC cases have metastasized by the time of diagnosis. Additionally, BCC lacks the perinuclear dotlike staining seen with CK20.2,7 Features present in BCC that are unusual for MCC include peripheral nuclear palisading, mucin, and retraction artifact on paraffin-embedded sections (Figure 1).7

Basal cell carcinoma
FIGURE 1. Basal cell carcinoma. Nodular growth with classic peripheral nuclear palisading, retraction, and focal mucin (H&E, original magnification ×200).

Leukemia cutis (or cutaneous infiltrates of leukemia) commonly displays a perivascular and periadnexal pattern in the dermis and subcutis. These infiltrates of neoplastic leukocytes can congregate into sheets, sometimes with an overlying Grenz zone, or form single-file infiltrates (Figure 2).1,4 The neoplastic cells can be monomorphic or atypical and commonly are susceptible to crush artifact.4 Although the immunohistochemical profile varies depending on the etiology of the underlying leukemia, broad hematologic markers such as CD43 and CD45 are helpful to discern these malignancies from MCC.4

Leukemia cutis
FIGURE 2. Leukemia cutis. Infiltration of metastatic leukemia cells in the dermis with a single-file infiltration pattern and atypical nuclei (H&E, original magnification ×400).

Being neuroendocrine in origin, metastatic small cell carcinoma (Figure 3) strongly mimics MCC histologically and usually stains with synaptophysin, chromogranin, and insulinoma-associated protein 1. Both tumor cells typically exhibit nuclear molding and high mitotic rates. Although small cell carcinoma is more likely to stain with high-molecular-weight cytokeratins (ie, CK7), it is not uncommon for these tumors to express lowmolecular- weight cytokeratins such as CK20. Because most cases originate from the lungs, these lesions should be positive for thyroid transcription factor 1 and negative for PAX-5, whereas MCC would show the reverse for those stains.1 Ultimately, however, clinical correlation with imaging results is the single best methodology for differentiation.

Metastatic small cell carcinoma
FIGURE 3. Metastatic small cell carcinoma. Sheets of infiltrative basophilic cells with fine chromatin, nuclear molding, and brisk mitoses (H&E, original magnification ×200).

Small cell melanoma, a variant of nevoid melanoma, can strongly resemble an MCC or a lymphoma. Usually located on the scalp or arising from a congenital nevus, small cell melanomas are aggressive and confer an unfavorable prognosis. Histologically, they consist of nests to sheets of atypical cells within the epidermis and dermis. These cells typically exhibit hyperchromatic nuclei, minimal cytoplasm, and frequent mitoses (Figure 4). Furthermore, the cells do not display maturation based on depth.8 These tumors usually are positive for HMB45, S-100, MART-1, SOX-10, and tyrosinase, all of which are extremely unlikely to stain an MCC.1

Small cell melanoma
FIGURE 4. Small cell melanoma. Infiltrative nests and individual cells involving the epidermis and dermis (H&E, original magnification ×400).

References
  1. Patterson JW, Hosler GA. Weedon’s Skin Pathology. 4th ed. Churchill Livingstone/Elsevier; 2016.
  2. Walsh NM, Cerroni L. Merkel cell carcinoma: a review. J Cutan Pathol. 2021;48:411-421.
  3. Topalian SL, Bhatia S, Amin A, et al. Neoadjuvant nivolumab for patients with resectable Merkel cell carcinoma in the CheckMate 358 Trial. J Clin Oncol. 2020;38:2476-2488.
  4. Rapini RP. Practical Dermatopathology. 3rd ed. Elsevier; 2021.
  5. Asioli S, Righi A, Volante M, et al. p63 expression as a new prognostic marker in Merkel cell carcinoma. Cancer. 2007;110:640-647.
  6. Verhaegen ME, Mangelberger D, Weick JW, et al. Merkel cell carcinoma dependence on Bcl-2 family members for survival. J Invest Dermatol. 2014;134:2241-2250.
  7. Le MD, O’Steen LH, Cassarino DS. A rare case of CK20/CK7 double negative Merkel cell carcinoma. Am J Dermatopathol. 2017;39:208-211.
  8. North JP, Bastian BC, Lazar AJ. Melanoma. In: Calonje E, Brenn T, Lazar AJ, et al, eds. McKee’s Pathology of the Skin With Clinical Correlations. 5th ed. Elsevier; 2020.
References
  1. Patterson JW, Hosler GA. Weedon’s Skin Pathology. 4th ed. Churchill Livingstone/Elsevier; 2016.
  2. Walsh NM, Cerroni L. Merkel cell carcinoma: a review. J Cutan Pathol. 2021;48:411-421.
  3. Topalian SL, Bhatia S, Amin A, et al. Neoadjuvant nivolumab for patients with resectable Merkel cell carcinoma in the CheckMate 358 Trial. J Clin Oncol. 2020;38:2476-2488.
  4. Rapini RP. Practical Dermatopathology. 3rd ed. Elsevier; 2021.
  5. Asioli S, Righi A, Volante M, et al. p63 expression as a new prognostic marker in Merkel cell carcinoma. Cancer. 2007;110:640-647.
  6. Verhaegen ME, Mangelberger D, Weick JW, et al. Merkel cell carcinoma dependence on Bcl-2 family members for survival. J Invest Dermatol. 2014;134:2241-2250.
  7. Le MD, O’Steen LH, Cassarino DS. A rare case of CK20/CK7 double negative Merkel cell carcinoma. Am J Dermatopathol. 2017;39:208-211.
  8. North JP, Bastian BC, Lazar AJ. Melanoma. In: Calonje E, Brenn T, Lazar AJ, et al, eds. McKee’s Pathology of the Skin With Clinical Correlations. 5th ed. Elsevier; 2020.
Issue
Cutis - 112(6)
Issue
Cutis - 112(6)
Page Number
287,297-298
Page Number
287,297-298
Publications
Publications
Topics
Article Type
Display Headline
Painful Growing Nodule on the Right Calf
Display Headline
Painful Growing Nodule on the Right Calf
Sections
Questionnaire Body

A 47-year-old woman with a history of combined variable immunodeficiency presented with a 2.6×2.4-cm nodule on the lateral aspect of the right calf that was first noticed 2 years prior as a smaller nodule. It increased in size and became painful to touch over the last 3 to 4 months. Following diagnostic biopsy, the nodule was removed by wide local excision and was tan-brown on gross dissection. The lesion showed dotlike perinuclear positivity with cytokeratin 20 immunostaining. Positron emission tomography–computed tomography showed no evidence of lung lesions. A complete blood cell count was within reference range.

Painful growing nodule on the right calf

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 12/05/2023 - 08:00
Un-Gate On Date
Tue, 12/05/2023 - 08:00
Use ProPublica
CFC Schedule Remove Status
Tue, 12/05/2023 - 08:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media