Superpage
Skin nonmelanocytic tumor

Authors: Ossama Abbas, M.D., Maryam Aghighi, M.D., Aadil Ahmed, M.D., Raniah Al Amri, M.D., Said Albahra, M.D., Borislav A. Alexiev, M.D., Yazan Alhalaseh, M.D., Ohoud Aljarbou, M.D., Nima Mesbah Ardakani, M.D., Sepideh Nikki Asadbeigi, M.D., Masoud Asgari, M.D., Aribah Atiq, M.B.B.S., Phyu Aung, M.D., Ph.D., Anshu Bandhlish, M.D., Laura Battle, M.D., Colleen J. Beatty, M.D., Monika Bhatt, M.D., Bernadette M. Boac, M.D., Elizabeth Boswell, M.D., Iva Brčić, M.D., Ph.D., Candice E. Brem, M.D., Richard A. Carr, M.B.Ch.B., Oliver Hsinju Chang, M.D., Jennifer Chapman, M.D., Alcides Chaux, M.D., Sheng Chen, M.D., Ph.D., Wei-Shen Chen, M.D., Ph.D., Woo Cheal Cho, M.D., Matthias Choschzick, M.D., Qurratulain Chundriger, M.B.B.S., Michael R. Clay, M.D., Jarish Cohen, M.D., Ph.D., Chico J. Collie, M.B.B.S., Carli Cox, M.D., Antonio L. Cubilla, M.D., Christopher Cullison, M.D., Mary Dick, M.D., Brendan C. Dickson, M.D., M.Sc., Hillary Rose Elwood, M.D., Na’im Fanaian, M.D., Negin Farsi, M.D., Saira Fatima, M.B.B.S., Julie Feldstein, M.D., María-Teresa Fernández-Figueras, Ph.D., Maria C. Ferrufino-Schmidt, M.D., Jerad M. Gardner, M.D., Nohra Ghaoui, M.D., Kathryn Gibbons, M.D., Pavandeep Gill, M.D., Christopher S. Hale, M.D., Lamiaa Hamie, M.D., M.Sc., Mowafak Hamodat, M.B.Ch.B., M.Sc., Mugahed Hamza, M.B.B.S., Jonathan D. Ho, M.B.B.S., D.Sc., Anjelica Hodgson, M.D., Haya Homsi, M.D., M.P.H., Gregory A. Hosler, M.D., Ph.D. , Amanda Ireland, M.B.B.S., Christine E. Jabcuga, M.D., Antonina Kalmykova, M.D., Viktoryia Kozlouskaya, M.D., Ph.D., Mahsa Khanlari, M.D., Mahyar Khazaeli, M.D., Joseph Khoury, M.D., Ronan Knittel, M.D., Elaine Kunzler, M.D., Kerri-Ann Latchmansingh, M.D., Robert E. LeBlanc, M.D., Philip LeBoit, M.D., Bonnie A. Lee, M.D., Bernadette Liegl-Atzwanger, M.D., Yi Ariel Liu, M.D., Yen-Chun Liu, M.D., Ph.D., Dragos C. Luca, M.D., Thai Yen Ly, M.D., Vasudevan D. Mahalingam, D.O., M.S., Jose G. Mantilla, M.D., Mario L. Marques-Piubelli, M.D., Anthony Martinez, M.D., Claire Mazahery, M.D., Ph.D., Phillip H. McKee, M.D., Jennifer M. McNiff, M.D., Doniya Milani, M.S., Roberto N. Miranda, M.D., Ata Moshiri, M.D., M.P.H. , Kiran Motaparthi, M.D., Carlos A. Murga-Zamalloa, M.D., Cuong Nguyen, M.D., Alexander Nirenberg, M.B.B.S., Haruto Nishida, M.D., Ph.D., Farres Obeidin, M.D., Erdener Özer, M.D., Ph.D., Noelia Pérez Muñoz, M.D., Nat Pernick, M.D., Anamarija M. Perry, M.D., Ashbita Pokharel, M.B.B.S., Olga Pozdnyakova, M.D., Ph.D., Dinesh Pradhan, M.D., Hong Qu, M.D., Aishwarya Ravindran, M.D., Muhammad Raza, M.B.B.S., Karen L. Rech, M.D., Aayushma Regmi, M.B.B.S., Nicole D. Riddle, M.D., Bethany R. Rohr, M.D., Shira Ronen, M.D., Cecilia Rosales, M.D., Simon F. Roy, M.D., Eleanor Russell-Goldman, M.D., Ph.D., Zaid Saeed Kamil, M.B.Ch.B., Jasmine Saleh, M.D., M.P.H., Christian Salib, M.D., Omar P. Sangueza, M.D., Sara C. Shalin, M.D., Ph.D., Vijay Shankar, M.D., Vignesh Shanmugam, M.D., Poonam Sharma, M.B.B.S., Lauren Smith, M.D., Jodi Speiser, M.D., Aravindhan Sriharan, M.D., Lauren N. Stuart, M.D., M.B.A., Narittee Sukswai, M.D., Saul Suster, M.D., Beenu Thakral, M.D., Joel Tjarks, M.D., Carlos A. Torres-Cabala, M.D., Tien-Anh Tran, M.D., Ghassan A. Tranesh, M.D., Calvin Tseng, M.D., Nicholas Turnbull, M.B.Ch.B., Nasir Ud Din, M.B.B.S., Caroline I.M. Underwood, M.D., V. Claire Vaughan, M.D., Noreen M. Walsh, M.D., Grace Y. Wang, M.D., Elizabeth Warbasse, M.D., Michella Whisman, M.D., Joshua Wisell, M.D., Bitania Wondimu, M.D., Shaofeng Yan, M.D., Ph.D., Sherehan Zada, M.D., Brandon Zelman, D.O., Nicholas A. Zoumberos, M.D.
Resident / Fellow Advisory Boards: Josephine K. Dermawan, M.D., Ph.D., Farres Obeidin, M.D., Caroline I.M. Underwood, M.D.
Board of reviewers: Caroline I.M. Underwood, M.D.
Editorial Board Members: Elizabeth Courville, M.D., Hillary Rose Elwood, M.D., Jonathan D. Ho, M.B.B.S., D.Sc., Viktoryia Kozlouskaya, M.D., Ph.D., Robert E. LeBlanc, M.D., Kiran Motaparthi, M.D., Farres Obeidin, M.D., Bethany R. Rohr, M.D., Sara C. Shalin, M.D., Ph.D., Patricia Tsang, M.D., M.B.A., Brandon Umphress, M.D., Debra L. Zynger, M.D.
Deputy Editors-in-Chief: Genevieve M. Crane, M.D., Ph.D., Jonathan D. Ho, M.B.B.S., D.Sc., Debra L. Zynger, M.D.
Editor-in-Chief: Debra L. Zynger, M.D.

Copyright: 2001-2024, PathologyOutlines.com, Inc.

DermPath related: Jobs, Fellowships, Conferences, Cases, CME, Board Review, What's New

Related chapters: Skin melanocytic tumor, Skin nontumor, Soft tissue

Editorial Board oversight: Jonathan D. Ho, M.B.B.S., D.Sc. (last revised April 2022), Viktoryia Kazlouskaya, M.D., Ph.D. (last revised January 2022)
Page views in 2024 to date: 16

Acanthoma fissuratum (pending)
[Pending]

Acquired digital fibrokeratoma
Definition / general
  • Acquired benign lesion of acral (limb or other extremity) skin
Terminology
  • Also known as acral fibrokeratoma and acquired periungual fibrokeratoma
Epidemiology
  • Middle aged adults
  • Men > women
Sites
  • Classically located on fingers and toes but can occur elsewhere on acral skin
Etiology
  • Etiology unknown
  • Trauma has been implicated but no studies have substantiated that hypothesis
Clinical features
  • Skin colored, slow growing firm nodule, from a few millimeters to over 1 cm in size
  • Rare giant variants have been described (Ann Dermatol 2011;23:64)
  • Hyperkeratotic collarete at base is characteristic
  • May have prominent verruciform surface resembling a verruca or cutaneous horn
Case reports
Treatment
  • Benign, although appearance or discomfort may prompt treatment
  • Excision is curative
Clinical images

Images hosted on other servers:
Left great toe

Left great toe

Middle finger

Middle finger

Collarette of epidermis surrounds nodules

Collarette of epidermis surrounds nodules


Solitary, projecting keratotic mass Solitary, projecting keratotic mass

Solitary, projecting keratotic mass

Skin colored, pedunculated firm nodule protruding from right heel toward sole Skin colored, pedunculated firm nodule protruding from right heel toward sole Skin colored, pedunculated firm nodule protruding from right heel toward sole

Skin colored, pedunculated firm nodule protruding from right heel toward sole

Gross description
  • Pedunculated to dome shaped nodule on acral skin
Microscopic (histologic) description
  • Polypoid lesion with variably hyperplastic epidermis covering a dermal proliferation composed of dense collagen fibers and variable amounts of mature fibroblasts, small blood vessels and elastic tissue (J Am Acad Dermatol 1985;12:816)
  • Thickened collagen in dermis is oriented predominantly in the vertical direction
  • Stellate stromal cells may be present
  • Covered by variably acanthotic epidermis with hyperkeratotic orthokeratosis
  • Lesion merges with adjacent normal dermis
  • Neural structures are absent or inconspicious
  • Lacks adnexal structures
Microscopic (histologic) images

Contributed by Hillary Rose Elwood, M.D.
Polypoid dermal proliferation

Polypoid dermal proliferation

Prominent dense collagen

Prominent dense collagen

Vertically oriented collagen fibers

Vertically oriented collagen fibers



Images hosted on other servers:
Dome shaped tumor

Dome shaped tumor

Collagen fibers perpendicularly arranged Collagen fibers perpendicularly arranged

Collagen fibers perpendicularly arranged

Acanthosis and massive orthokeratosis

Acanthosis and massive orthokeratosis

Thickened collagen Thickened collagen

Thickened collagen


Hyperkeratosis and acanthosis

Hyperkeratosis and acanthosis

Proliferating fibroblasts and capillaries Proliferating fibroblasts and capillaries

Proliferating fibroblasts and capillaries

Various images Various images

Various images

Negative stains
  • Noncontributory
Differential diagnosis
  • Acrochordon: non acral location, pedunculated, less hyperkeratotic, less dense connective tissue
  • Hypertrophic scar: normal or atrophic epidermis, dermal band of fibroblasts and dense collagen, blood vessels oriented perpendicular to epidermis
  • Periungual fibroma (Koenen tumor): similar/identical histology, distinction is predominantly based on clinical findings (i.e. location, multiple lesions, patient with tuberous sclerosis); some have noted that periungual fibromas can have prominent stellate atypical myofibroblasts, may have a more prominent vascular component, and may lack the epidermal changes of digital fibrokeratoma (Arch Dermatol 1995;131:1465)
  • Supernumerary digit: has prominent neural structures (i.e. peripheral nerves or tactile corpuscles), sometimes cartilage/bone is present; most are related to the fifth digit

Acquired elastotic hemangioma (pending)
[Pending]

Actinic keratosis
Definition / general
  • Intraepidermal keratinocytic lesion secondary to solar damage
  • Most common precursor of cutaneous squamous cell carcinoma (SCC)
Essential features
  • Most common precursor of cutaneous squamous cell carcinoma
  • Secondary to solar damage
  • Epidermal dysplasia that is not full thickness
  • Parakeratosis, dermal solar elastosis and mild dermal lymphocytic infiltrate
Terminology
  • Solar keratosis, senile keratosis, keratinocytic intraepithelial neoplasia
ICD coding
  • ICD-10: L57.0 - actinic keratosis
  • ICD-11: EK90 - actinic keratosis and other discrete epidermal dysplasias
  • ICD-O: see SNOMED
  • SNOMED CT: 201101007
Epidemiology
Sites
Pathophysiology
  • Cumulative solar damage causes a high mutation burden in the germinative (basal) layer of keratinocytes
Etiology
  • Chronic sun damage (ultraviolet UVB radiation and UVA to a lesser extent)
  • Chronic immunosuppression
  • Arsenic exposure
  • PUVA therapy
  • Chronic cutaneous inflammation
  • βHPV (Viruses 2017;9:187)
    • High prevalence rates and viral loads in actinic keratoses
    • Virus, when combined with UV exposure, may play a role in progression to SCC
Diagrams / tables

Contributed by María-Teresa Fernández-Figueras, Ph.D.

Pathways of actinic keratosis progression

Clinical features
  • Single or multiple erythematous and hyperkeratotic macules or papules
  • Usually < 1 cm in diameter
  • Sometimes pigmented or ulcerated
  • Chronic sun damage of background skin
  • Reference: J Drugs Dermatol 2010;9:1125
Diagnosis
  • Usually by clinical examination
  • Dermoscopy shows erythematous to brown networks, irregular hyperpigmented follicular openings, grey dots and lineal or circular structures; surface scales are common (Curr Probl Dermatol 2015;46:70)
  • In vivo reflectance confocal microscopy shows a disarranged or mildly atypical honeycomb pattern (Eur J Dermatol 2016;26:549)
Prognostic factors
  • Can remain stable for long periods or regress
  • Rate of transformation to SCC: closer to 1/100 (from 1/10 to 1/1000 in different studies)
    • Proliferative variant has higher risk of invasion
  • 2 main pathways for progression to SCC (J Eur Acad Dermatol Venereol 2018;32:581):
    • Classical: atypical basal keratinocytes in an actinic keratosis invade the mid to upper epidermis and transform to invasive SCC
    • Differentiated: invasive SCC develops directly from actinic keratosis with dysplasia limited to the basal layer
Case reports
  • 54 year old man with a dark lesion on his nasal tip; biopsy revealed pigmented actinic keratosis (Cureus 2019;11:e4721)
  • 68 year old woman with non small cell lung adenocarcinoma with eruptive inflamed actinic keratosis consequent to systemic chemotherapy (Dermatol Online J 2017;23:13030/qt77s2r0p9)
  • 72 year old woman developed an acute inflammation of her actinic keratosis while on oral capecitabine chemotherapy for rectal adenocarcinoma (J Cutan Med Surg 2012;16:298)
  • 77 year old woman with seborrheic keratosis that transformed into bowenoid actinic keratosis (Case Rep Dermatol 2020;12:19)
Treatment
Clinical images

Contributed by Dr. Victoria Amat-Samaranch
Actinic keratosis

Clinical appearance of actinic keratosis

Dermoscopy of the previous case

Dermoscopy of the previous case

Gross description
Gross images
Frozen section description
  • Frozen section not performed as lesion is precancerous
Microscopic (histologic) description
  • Atypia of basal keratinocytes with loss of polarization, crowding and overlapping
    • Can progress to involve the mid to upper epidermal layers
    • Can extend along adnexal structures; typically limited to infundibular extension
    • Never full thickness
  • Typically, mild acanthosis with occasional downward buds; tangential sectioning of these buds can prompt overdiagnosis of superficially invasive SCC but these do not represent actual squamous pearls
  • Focal parakeratosis
    • Flag sign: occasional columns of grey loose keratin above preserved acrosyringiums (known as Freudenthal funnels) alternate vertically with eosinophilic compact hyperkeratosis
  • Lost / attenuated granular layer
  • Dermal changes: solar elastosis, mild inflammatory infiltrate (usually lymphocytes; plasma cells present in longstanding lesions)
  • Variants:
    • Hypertrophic / hyperplastic: prominent irregular acanthosis, parakeratosis and orthokeratosis
    • Atrophic: thin epidermis
    • Lichenoid: band-like lymphocytic infiltrate of papillary dermis, basal keratinocytes with scattered apoptosis and vacuolar change
    • Acantholytic: suprabasal clefting between atypical keratinocytes
    • Pigmented: increased melanin in the basal keratinocytes and melanocytes; melanophages in the papillary dermis
    • Proliferative: dermal projections of nested atypical keratinocytes; dense dermal inflammation
    • Bowenoid: near full thickness atypia, usually focal
    • Pagetoid: pale cells
Microscopic (histologic) images

Contributed by María-Teresa Fernández-Figueras, Ph.D.

Early lesion

Keratinocyte atypia and lymphocytic infiltrate

Bowenoid actinic keratosis

Spared acrosyringium

Pigmented atrophic


Hypertrophic

Freudenthal funnels

Acantholytic

Acantholysis and eccrine involvement

Lichenoid


Proliferative

Pagetoid

Regression

Relapse

p53

Virtual slides

Images hosted on other servers:

Atrophic with infundibular involvement

Hypertrophic

Positive stains
Negative stains
Molecular / cytogenetics description
Videos

Tutorial on actinic keratosis histopathology

Sample pathology report
  • Skin, forehead, curettage:
    • Fragments of hypertrophic actinic keratosis (see comment)
    • Comment: Due to fragmentation, small areas of in situ or invasive squamous cell carcinoma cannot be ruled out.
Differential diagnosis
  • Actinic cheilitis:
  • Squamous cell carcinoma:
    • In situ: full thickness epidermal dysplasia
    • Invasive:
      • Isolated dermal squamous cells or pearls
      • Dermal desmoplasia
    • Pseudomaturation with large keratinized cells close to the dermal collagen
    • Finger-like projections of proliferative actinic keratosis are challenging to differentiate from invasion, especially when tangentially sectioned
  • Bowen disease (classical):
  • Benign lichenoid keratosis:
    • Also known as lichen planus-like keratosis: resembles a lichen planus and often shows hypergranulosis (Dermatol Reports 2011;3:e25)
    • Sharply demarcated
    • Mild reactive keratinocyte atypia
  • Dysmaturatative drug eruptions:
    • In patients receiving chemotherapeutic drugs (J Eur Acad Dermatol Venereol 2016;30:638)
    • Atypical starburst-like or ring-like mitoses are common
    • Dyskeratosis of basal and suprabasal layers of the epidermis
    • Occasional squamous syringometaplasia
  • Grover disease (transient acantholytic dermatosis):
    • Lesions show dismaturation and lentiginous silhouette (Am J Dermatopathol 2010;32:541)
    • Areas of acantholysis or dyskeratosis can be present in actinic keratosis but the phenomenon of acantholytic dyskeratosis favors the diagnosis of Grover disease
Board review style question #1

Which of the following is true about the entity shown in the photo above?

  1. Clinically ranges from small papules to pustulous plaques or erythroderma
  2. It is frequently located in the genital area
  3. It is the most common precursor of cutaneous invasive squamous cell carcinoma
  4. Nodular growth pattern is considered a low risk variant
  5. p16 is positive in high risk cases
Board review style answer #1
C. The image depicts actinic keratosis, which is the most common precursor of cutaneous invasive squamous cell carcinoma. Answer C is an essential feature of actinic keratosis as it has well established its link in the development of squamous cell carcinoma either in the classical or differentiated pathways. Erythroderma is not a clinical feature of actinic keratosis. The genital area, which is not typically sun exposed, is not a frequent location. Actinic keratosis does not typically occur in a nodular growth pattern. p16 positivity is seen in high risk HPV related lesions, whereas βHPV associated with actinic keratosis is not considered one of these strains.

Comment Here

Reference: Actinic keratosis
Board review style question #2
Positivity of which of the following these immunohistochemical stains may be helpful in the diagnosis of actinic keratosis?

  1. BerEP4
  2. EMA
  3. MelanA
  4. p16
  5. p53
Board review style answer #2
E. p53. p53 protein is overexpressed in the basal layers of the epithelium in 50% of actinic keratosis, which may aid in the diagnosis of difficult cases. BerEP4 is positive in basal cell carcinoma but negative in actinic keratosis. EMA is widely expressed on epithelial cells and has no use in solar damaged skin. MelanA might show a slight melanocytic hyperplasia but is not specific for actinic keratosis. Finally, strong p16 positivity is seen in lesions caused by high risk HPV, whereas βHPV associated with actinic keratosis is not considered one of these strains.

Comment Here

Reference: Actinic keratosis
Board review style question #3
Which immunohistochemical positive staining in actinic keratosis can be the cause of a diagnostic error?

  1. CD20
  2. HMB45
  3. MelanA
  4. Sox10
  5. Vimentin
Board review style answer #3
C. MelanA. The presence of MelanA positive cells and pseudomelanocytic nests is a well known pitfall in the histological and immunohistochemical diagnosis of actinic keratosis. These cells are typically negative for other melanocytic markers, such as SOX10 or HMB45. CD20 is a marker of B lymphocytes and vimentin is negative in squamous epithelium.

Comment Here

Reference: Actinic keratosis

Adenoid cystic carcinoma (primary cutaneous)
Definition / general
  • Rare, slow growing cutaneous adnexal tumor with 3 major histologic patterns: tubular, cribriform and solid
  • Morphologically indistinguishable from adenoid cystic carcinoma at other sites; must rule out cutaneous extension from salivary gland malignancy or metastasis from other locations
  • More indolent than salivary gland counterpart (Am J Surg Pathol 2013;37:1603)
Essential features
  • Primary cutaneous adenoid cystic carcinoma (PCACC) is a rare adnexal tumor with different histopathologic growth patterns, including cribriform, tubular and solid variants
  • Perineural invasion is a common feature and may explain the high recurrence level
  • Diagnosis is confirmed only after ruling out metastatic disease through thorough clinical history and imaging studies
  • 60% of PCACC cases exhibit MYB overexpression, which is mediated by MYB::NFIB fusion or other MYB chromosomal abnormalities
ICD coding
  • ICD-O: 8200/3 - adenoid cystic carcinoma
  • ICD-11: 2C33 & XH4302 - adnexal carcinoma of skin & adenoid cystic carcinoma
Epidemiology
Sites
  • Head and neck (particularly the scalp) is the most frequently involved site, followed by the trunk, upper limbs / limb girdle and lower limb / limb gridle (Histopathology 2022;80:407)
Pathophysiology
Etiology
  • Unknown
Clinical features
Diagnosis
  • Essential
    • Histology with 2 types of lumina, including true small bilayered ducts (with or without secretion) and pseudocysts containing basophilic mucinous material in multinodular neoplasm
    • 2 main cell types: ductal and myoepithelial cells
  • Desirable
    • Cylindroma-like appearance
    • Perineural invasion
    • Coexpression of MYB and CD117
    • Demonstration of MYB or MYBL1 rearrangements in selected cases
Prognostic factors
  • Metastasis is rare (typically to lymph nodes or lungs) and associated with high grade transformation (Histopathology 2022;80:407)
  • Recurrence is very common, primarily attributed to perineural invasion (Am J Surg Pathol 2013;37:1603)
  • Vulva and perigenital sites may correlate with more aggressive disease / metastases, possibly due to the abundance of nerve tissue in this area; however, larger studies are needed to validate this supposition (Am J Surg Pathol 2013;37:1603)
  • Factors that may reflect a more biologically aggressive tumor (Histopathology 2022;80:407)
    • Longest diameter of the lesion (≥ 1 cm)
    • Involvement of subcutaneous fat tissue and widely infiltrative border
    • Grade III / high grade transformation
  • In multivariate analysis of 451 cases, more recent year of diagnosis, advanced patient age and advanced stage were associated with poorer outcomes (JAAD 2021;85:245)
Case reports
Treatment
  • Wide surgical excision to prevent recurrence
  • Mohs surgery has been performed for better control of surgical margins (JAMA Dermatol 2013;149:1343)
Clinical images

Images hosted on other servers:
Nodule with slight erythema

Nodule with slight erythema

Scalp lesion with alopecia

Scalp lesion with alopecia

Erythematous patch on chest

Erythematous patch on chest

Gross description
  • Smooth surface with a firm consistency and a tan-gray color
Gross images

Images hosted on other servers:
Lesion between dermis and hypodermis

Lesion between dermis and hypodermis

Frozen section description
  • Frozen sections are only utilized to confirm diagnosis or verify that the surgical margins were negative
  • Shows the characteristic histologic findings of PCACC: basaloid cells within dermis, arranged in cords and islands forming tubular structures and cribriform patterns
Frozen section images

Images hosted on other servers:
Ductal and cystic structures in dermis

Ductal and cystic structures in dermis

Cribriform pattern

Cribriform pattern

Secretion within cysts

Secretion within cysts

Microscopic (histologic) description
  • Poorly circumscribed, dermal based basaloid neoplasm with numerous ductular and cystic spaces
  • Often with subcutaneous extension; epidermal involvement unusual but has been reported (Am J Dermatopathol 2019;41:619)
    • An essential diagnostic feature is the coexistence of true small bilayered ducts and pseudocysts (Histopathology 2022;80:407)
    • Sparse ductal structures composed of inner epithelial cells surrounded by an outer layer of basal / myoepithelial cells; intraluminal secretion may be present
    • Cystic spaces (larger than ducts) contain abundant basophilic mucinous material or hyalinized eosinophilic material
    • Deposition of hyaline basement membrane material on the intraluminal aspect of cystic spaces
  • Mixture of cribriform, tubular and solid patterns (Am J Surg Pathol 2013;37:1603)
    • Cells are typically small and monomorphic appearing with hyperchromatic nuclei and scant cytoplasm
  • Mitotic activity is typically low
  • Perineural invasion is a common finding

Grading criteria (Histopathology 2022;80:407)
Grade I Grade II Grade III
Histologic features
  • Tubular and cribriform patterns
  • Few or no mitotic figures
  • Pure cribriform or mixed pattern
  • Few or no mitotic figures
  • Necrosis
  • Increased mitotic figures
Solid area None < 30% > 30%
Cytologic features No cellular atypia Atypia > grade I but < grade III Significant cellular atypia

  • High grade transformation (HGT), previously known as dedifferentiation
    • Distinct population of anaplastic cells with loss of the biphasic ductal - myoepithelial differentiation
      • Loss of myoepithelial cell differentiation is more usual; however, 1 PCACC case with HGT showed loss of epithelial cell differentiation and was diagnosed as myoepithelial carcinoma
    • Nuclear enlargement, higher mitotic counts and necrosis are commonly seen
  • HGT should be differentiated from solid type adenoid cystic carcinoma (ACC)
    • With HGT, solid transformed areas are
      • Separated from the tubular / cribriform component
      • Have higher p53 and Ki67 expression
    • Solid type ACC
      • Solid cell nests are intermixed with cribriform and tubular structures throughout the tumor
Microscopic (histologic) images

Contributed by Haya Homsi, M.D., M.P.H. and Shira Ronen, M.D.
Dermal basaloid neoplasm

Dermal basaloid neoplasm

Multiple ductular and cystic structures

Multiple ductular and cystic structures

Cribriform pattern

Cribriform pattern

Eosinophilic basement membrane Eosinophilic basement membrane

Eosinophilic basement membrane


Basophilic intraluminal mucin Basophilic intraluminal mucin

Basophilic intraluminal mucin

Perineural invasion

Perineural invasion

Solid area

Solid area

Small cells with round hyperchromatic nuclei

Small cells with round hyperchromatic nuclei


Infiltrative nests and cords

Infiltrative nests and cords

EMA

EMA

CD117

CD117

Calponin

Calponin

S100

S100

Positive stains
Negative stains
Molecular / cytogenetics description
  • ~60% of PCACC cases have been found to harbor MYB gene activations, either through MYB chromosomal abnormalities or MYB::NFIB fusion (J Cutan Pathol 2017;44:201)
  • Less commonly, MYBL1 gene alterations have also been reported (Am J Dermatopathol 2018;40:721)
  • According to one study, detection of MYB gene alterations does not necessarily correlate with MYB protein expression (i.e., MYB immunohistochemistry staining) (Histopathology 2022;80:407)
Molecular / cytogenetics images

Images hosted on other servers:
MYB rearrangements on FISH

MYB rearrangements on FISH

Videos

Primary cutaneous adenoid cystic carcinoma

Sample pathology report
  • Skin, forehead, excision
    • Adenoid cystic carcinoma (see comment)
    • Comment: Sections demonstrate an excision specimen containing multiple foci of basaloid nodules with a cribriform architecture and duct formation. Lumen spaces are filled with blue mucin and eosinophilic basement membrane-like material. Cytologically, the tumor cells show scant to moderate pale eosinophilic cytoplasm and oval nuclei with vesicular chromatin. Significant pleomorphism and mitotic activity is not appreciated. Solid growth is not identified. Perineural invasion is present. The margins are negative for tumor.
    • To further characterize this case, immunohistochemical stains with SMMS1, CD117 and SOX10 were performed and compared with appropriate controls. The tumor is positive for SOX10, CD117 and SMMS1.
    • Molecular studies detected MYB gene rearrangement.
    • The histologic and immunohistochemical findings support the diagnosis of adenoid cystic carcinoma. This could be a primary cutaneous lesion but a metastasis from a salivary gland tumor cannot be excluded. An appropriate clinical evaluation to rule out that possibility is recommended.
Differential diagnosis
  • Metastatic adenoid cystic carcinoma:
    • Can only exclude based on clinical history and imaging studies
    • CK15 and vimentin may have some discriminatory value in differentiating between primary cutaneous and salivary gland ACCs; one study found that CK15 and vimentin showed diffuse positivity in 36% and 57% of PCACCs, respectively, whereas all salivary ACCs were negative or only focally positive for either CK15 or vimentin (Am J Surg Path 2015;39:1347)
  • Basal cell carcinoma (adenoid type):
    • Peripheral palisading
    • Retraction artifact
    • Mucinous stroma
    • Attachment to epidermis often present
    • Lack of true ductal structures with myoepithelial cells
    • Focal areas of conventional basal cell carcinoma
  • Cutaneous secretory carcinoma (Am J Surg Pathol 2017;41:62):
    • Back to back tubules and microcysts with intraluminal secretions
    • Positive for S100, mammaglobin, STAT5A and NTRK
  • Primary cutaneous mucinous carcinoma:
    • Less prominent cribriform pattern
    • Mucin present in stroma
    • Usually positive for ER, PR, GCDFP-15 and neuroendocrine markers
  • Primary cutaneous cribriform tumor (J Cutan Pathol 2005;32:577):
    • Interconnected cribriform lesion that varies in size and shape
    • Focal solid areas
    • More eosinophilic cells
    • Lacks myoeithelial cells
    • Perineural invasion not a common feature
  • Cylindroma / spiradenoma:
    • Cylindroma: absence of prominent cystic / ductal spaces with mucin
    • Spiradenoma: presence of dilated vascular spaces and numerous lymphocytes
    • Lack perineural invasion
    • Recognition of the typical areas
      • Cylindroma: well circumscribed, symmetrical lesion composed of multiple aggregates of basaloid cells surrounded by a prominent basement membrane that appear to fit together in jigsaw puzzle arrangement
      • Spiradenoma: regular nodules of small basaloid cells admixed with lymphocytes and basement membrane material
Board review style question #1

Which of the following is true about the primary cutaneous lesion shown above?

  1. Metastasis is frequent
  2. Most commonly involves the extremities
  3. Perineural invasion is a common finding
  4. Typically presents as multiple nodules
Board review style answer #1
C. Perineural invasion is a common finding. Perineural invasion is very common in adenoid cystic carcinoma (ACC). Answer A is incorrect because metastasis from primary cutaneous ACC is rare. Answer B is incorrect because the head and neck, followed by the trunk, are the most common sites. Answer D is incorrect because it appears as a solitary nodule.

Comment Here

Reference: Adenoid cystic carcinoma (primary cutaneous)
Board review style question #2
A 70 year old man presented with a scalp nodule. Microscopic examination showed cribriform nests and columns of bland cells arranged concentrically around gland-like spaces filled with homogenous basophilic material. Testing for mutation in which gene should be recommended for this patient?

  1. CYLD
  2. BHD
  3. EGFR
  4. MYB
Board review style answer #2
D. MYB. MYB and MYBL1 gene alterations, including MYB::NFIB or MYBL1::NFIB fusions, are frequently detected in adenoid cystic carcinoma (ACC). Answer A is incorrect because CYLD gene alterations are seen in cylindromas, spiradenomas and trichoepitheliomas. Answer B is incorrect because pathogenic BHD mutations are seen in Birt-Hogg-Dubé syndrome. Answer C is incorrect because EGFR gene alternations are not seen in ACC.

Comment Here

Reference: Adenoid cystic carcinoma (primary cutaneous)

Alveolar rhabdomyosarcoma

Angiofibroma / fibrous papule
Definition / general
  • Benign fibrohistiocytic tumor consisting of dermal dendritic cells
Essential features
  • Common benign lesion composed of stellate, factor XIIIa positive stromal cells
  • Most common variant referred to as fibrous papule when present on the central face
Terminology
  • Solitary angiofibroma on the central face referred to as fibrous papule
  • Multiple angiofibromatous lesions in tuberous sclerosis referred to as adenoma sebaceum
  • On the penis, referred to as pearly penile papules
ICD coding
  • ICD-O: 9160/0 - angiofibroma, NOS
  • ICD-10: D21.9 - benign neoplasm of connective and other soft tissue, unspecified
  • ICD-10: D23.30 - other benign neoplasm of skin of unspecified part of face
Epidemiology
Sites
  • Central face, particularly on the nose
Pathophysiology
  • Unknown at this time
Etiology
Clinical features
  • Dome shaped, skin colored papules measuring a few millimeters
  • Asymptomatic
Diagnosis
Case reports
Treatment
  • For cosmetic purposes, can be removed by excisional / shave biopsy or electrosurgery
Clinical images

Images hosted on other servers:
Fibrous papule of the nose Various images

Fibrous papule of the nose

Tuberous sclerosis angiofibromas

Tuberous sclerosis angiofibromas

Microscopic (histologic) description
  • Slightly raised dermal lesion composed of collagenous stroma with increased vasculature
  • Increased stromal cells with varying morphology
    • Cells can be plump, spindle shaped, stellate or multinucleate
  • Mitotic figures are rare
  • Epidermis uninvolved but can appear flattened or atrophic
  • Can have overlying junctional melanocytic hyperplasia
    • Useful clue in partially / limited biopsies
    • However, overdiagnosis (atypical junction melanocytic hyperplasia or melanoma in situ) is a pitfall that should be avoided
  • Histologic variants exist:
Microscopic (histologic) images

Contributed by Gregory A. Hosler, M.D., Ph.D.
Fibrous papule, superficial shave

Fibrous papule, superficial shave

Fibrous papule, classic Fibrous papule, classic

Fibrous papule, classic

Hypercellular variant Hypercellular variant

Hypercellular variant


Giant hypercellular variant Giant hypercellular variant

Giant hypercellular variant

Clear cell variant Clear cell variant

Clear cell variant

Pigmented variant Pigmented variant

Pigmented variant



Contributed by Hillary Rose Elwood, M.D.
Fibrous papule with multinucleate cells

Fibrous papule with multinucleate cells

Fibrous papule, classic Fibrous papule, classic

Fibrous papule, classic

Clear cell variant

Clear cell variant

Virtual slides

Images hosted on other servers:
Cutaneous angiofibroma

Cutaneous angiofibroma

Positive stains
Negative stains
Electron microscopy description
Videos

Fibrous papule (angiofibroma)
by Dr. Gardner

Sample pathology report
  • Skin, nasal tip, shave biopsy:
    • Fibrous papule
Differential diagnosis
  • Adenoma sebaceum (angiofibroma of tuberous sclerosis):
    • Fewer bizarre dermal stromal cells than in fibrous papule
    • Vessels are smaller and likely to show more concentric fibrosis
  • Pearly penile papules:
    • Absence of pilosebaceous follicles
    • Multiple in number and located on the penis
  • Angioma:
    • Proliferation of ectatic vessels
    • Lacks stellate stromal cells
  • Fibrosing / sclerotic nevus:
    • Lacks stellate cells and cellular stroma
    • S100 positive
  • Pleomorphic fibroma:
    • Pleomorphic cells can resemble those occasionally present in fibrous papule
    • More commonly located on the truck and extremities
    • Lacks vascularity
  • Scar:
    • Horizontal orientation of fibroblasts with vertically oriented blood vessels
Board review style question #1

Which of the following is most commonly expressed in this lesion from the nasal ala of a 45 year old woman?

  1. Cytokeratin AE1 / AE3
  2. Factor XIIIa
  3. MART1
  4. NKI-C3
  5. S100
Board review style answer #1
B. Factor XIIIa. The spindle cells of fibrous papule express factor XIIIa. Although NKI-C3 may be positive in the clear cell variant of fibrous papule, it is negative in classic variant shown here. S100, MART1 and cytokeratins are negative.

Comment Here

Reference: Angiofibroma / fibrous papule
Board review style question #2
Multiple facial fibrous papules are associated with what hereditary condition?

  1. Brooke-Spiegler syndrome
  2. Cowden syndrome
  3. Gardner syndrome
  4. Reed syndrome
  5. Tuberous sclerosis
Board review style answer #2
E. Tuberous sclerosis. Multiple facial fibrous papules are classically associated with the autosomal dominant neurocutaneous syndrome of tuberous sclerosis. Angiofibromatous lesions found in patients with this syndrome are referred to as adenoma sebaceum (a misnomer, as there is no adenomatous proliferation of sebaceous glands). Other hereditary conditions also associated with multiple facial fibrous papules include multiple endocrine neoplasia type I (MEN 1), neurofibromatosis II and Birt-Hogg-Dubé.

Comment Here

Reference: Angiofibroma / fibrous papule

Angiokeratoma
Definition / general
  • Benign vascular lesion characterized by superficial vascular ectasia and overlying epidermal hyperplasia (acanthosis or hyperkeratosis)
  • Lesions may be solitary or multiple / diffuse

  • Five types with similar histology:
    • Angiokeratoma of Mibelli: seen in children and adolescents on dorsum of toes and fingers
    • Angiokeratoma of Fordyce: scrotal skin of elderly
    • Angiokeratoma corporis diffusum: clustered papules in a bathing suit distribution; associated with Anderson-Fabry disease (X-linked recessive lysosomal storage disease)
    • Angiokeratoma circumscriptum: least common type, usually congenital, associated with nevus flammeus, cavernous hemangioma
    • Idiopathic solitary or multiple angiokeratomas
Essential features
  • Benign vascular lesion
  • Characterized by superficial vascular ectasia and overlying epidermal hyperplasia
  • May occur in a variety of clinical settings
  • Associated with Anderson-Fabry disease (X-linked recessive lysosomal storage disease)
Case reports
Gross description
  • Small red to brown / black papule or nodule with verrucous surface, can be clustered
Gross images

Images hosted on other servers:

Various images

Microscopic (histologic) description
  • Vascular ectasia of the papillary dermis which may appear to extend into the epidermis
  • Overlying epidermal hyperplasia characterized by acanthosis, elongation of the rete and hyperkeratosis, with the epidermis encircling the dilated vascular spaces
  • Often thrombosis within the vascular ectasia

Microscopic (histologic) images

Contributed by Sabrina C. Sopha, M.D, Joel Tjarks, M.D., Angel Fernandez-Flores, M.D., Ph.D. and @RaulSGonzalezMD on Twitter

43 year old man, scrotal lesion

Angiokeratoma

Angiokeratoma, vulva

Various images

Angiokeratoma Angiokeratoma

Angiokeratoma

Electron microscopy description
  • In patients with Anderson-Fabry disease, see lipid bodies and lamellar inclusions in endothelial cells, pericytes and smooth muscle cells in angiokeratomas
Differential diagnosis
  • Hemangioma (verrucous, lobular capillary, etc.)
  • Lymphangioma
  • Venous lake
  • Clinical differential diagnosis may include pigmented / melanocytic lesions due to thrombosis

Angioleiomyoma

Angiolymphoid hyperplasia with eosinophilia
Definition / general
  • Angiolymphoid hyperplasia with eosinophilia (ALHE) is a rare, benign vascular neoplasm characterized by proliferation of blood vessels along with inflammatory infiltrate rich in eosinophils
Essential features
  • Nonmalignant vasoproliferative lesion
  • Predominant epithelioid or histiocytoid endothelial cells
  • Papules or nodules
  • Mixed inflammatory infiltrate composed of lymphocytes, plasma cells and eosinophils
Terminology
  • Angiolymphoid hyperplasia with eosinophilia
  • Epithelioid hemangioma
  • Obsolete terms
    • Histiocytoid hemangioma
    • Angiomatous nodule
    • Pseudopyogenic granuloma
    • Inflammatory angiomatous nodule
ICD coding
  • ICD-10: L98.8 - other specified disorders of the skin and subcutaneous tissue
Epidemiology
  • Most common in middle aged adults
  • Slight female preponderance
  • More frequent in Asian populations, particularly in individuals of Japanese and Chinese descent (J Am Acad Dermatol 2016;74:506)
Sites
  • Most common sites are skin and subcutaneous tissue of head and neck, particularly the face, scalp and ear lobes (Int J Surg Pathol 2016;24:59)
  • Less common sites are limbs, oral mucosa and genitalia
Pathophysiology
  • Pathophysiology of ALHE is likely multifactorial, involving a complex interplay of angiogenesis, immune dysregulation and possibly genetic and environmental factors (Dermatol Pract Concept 2018;8:28)
    • Initial inflammatory event or injury to the blood vessels triggers a cascade of events leading to the proliferation of blood vessels known as angiogenesis
    • Eosinophils are known to have a role in allergic and inflammatory response
    • Presence of eosinophils is very much suggestive of a dysregulated immune response
    • Inflammatory mediators released by eosinophils may be a contributing factor to the vascular changes seen in ALHE
    • Other possible contributing factors are
      • Arteriovenous shunting
      • Local trauma
      • Elevated serum estrogen levels
Etiology
  • Idiopathic condition
  • Theories include vascular abnormalities, immunologic factors, inflammatory response, infectious agents, genetic factors, hormonal factors (Open Access Maced J Med Sci 2017;5:423)
Clinical features
  • Slow growing cutaneous lesions
  • Can be single or multiple
  • Firm, red to violaceous (purple) nodules or papules (J Am Acad Dermatol 2016;75:e19)
  • Size ranges from a few millimeters to centimeters
  • Sometimes can be pruritic and occasionally ulcerate (Dermatol Pract Concept 2021;11:e2021003)
  • Rarely associated with tenderness / pain
  • Some patients may experience regional lymphadenopathy near the affected area
  • Systemic involvement is rare
  • Can be clinically mistaken for hemangioma or other vascular lesions
  • Peripheral eosinophilia has been reported in few cases (Indian J Dermatol 2020;65:556)
Diagnosis
  • Definite diagnosis requires a biopsy and histopathological examination
  • Imaging modalities can help distinguish ALHE from other vascular or soft tissue lesions (J Am Acad Dermatol 2016;74:506)
  • Additional diagnostic tests can be performed to rule out the other causes of eosinophilia, if clinically indicated (J Am Acad Dermatol 2016;74:506)
Laboratory
Radiology description
Prognostic factors
  • Benign localized disease with indolent course
  • Does not regress without intervention
  • Recurrence is a common feature; can appear at the same site or in different locations (Indian Dermatol Online J 2017;8:267)
Case reports
Treatment
  • Total surgical excision is the current treatment of choice
  • Other treatment options include laser therapy and corticosteroid injections (J Am Acad Dermatol 2013;68:e48)
Clinical images

Images hosted on other servers:
Angiomatous subcutaneous plaque on scalp

Angiomatous subcutaneous plaque on scalp

Erythematous nodule on skin

Erythematous nodule on skin

Erythematous nodule on right cheek

Erythematous nodule on right cheek

Erythematous nodules in occipital region

Erythematous nodules in occipital region

Coalescing nodules

Coalescing nodules

Gross description
  • Single or multiple, dome shaped, light pink to red brown papules or subcutaneous masses with no specific distinguishing surface features (J Am Acad Dermatol 2016;74:506)
  • Size may range from few millimeters to a centimeter
  • There might be erosion or crust formation of the surface (J Am Acad Dermatol 2016;74:506)
Gross images

Images hosted on other servers:
Excised lesion

Excised lesion

Microscopic (histologic) description
  • Dermal based lesion with extension into the subcutaneous tissue
  • Proliferation of vascular channels as nests and cords of endothelial cell proliferations with admixed lymphocytes, plasma cells and eosinophils, accompanied by hemorrhage and proliferation of thick and thin walled blood vessels (Dermatol Pract Concept 2018;8:28)
  • Endothelial cells show large vesicular nuclei with acidophilic and sometimes vacuolated cytoplasm, imparting a hobnail appearance (Clin Colorectal Cancer 2010;9:179)
  • Mitoses can be seen but lack atypical features and anaplasia
  • Eosinophilia is usually a striking feature
  • Stroma may show varying degrees of fibrosis, which can be a contributing factor to the firmness observed clinically (StatPearls: Angiolymphoid Hyperplasia With Eosinophilia [Accessed 19 January 2024])
Microscopic (histologic) images

Contributed by Aribah Atiq, M.B.B.S.
Prominent lymphoid follicles

Prominent lymphoid follicles

Vascular proliferation with lymphoid infiltrate

Vascular proliferation with lymphoid infiltrate

ALHE involving salivary gland

ALHE involving salivary gland

Vascular proliferation

Vascular proliferation

Fibrosis with inflammation

Fibrosis with inflammation

Eosinophilia

Eosinophilia

Virtual slides

Images hosted on other servers:
Angiolymphoid hyperplasia with eosinophilia

Angiolymphoid hyperplasia with eosinophilia

Cytology description
  • Plump polygonal endothelial cells with vesicular nuclei and deeply eosinophilic cytoplasm (Diagn Cytopathol 2021;49:E7)
  • Background shows eosinophils and lymphocytes
Positive stains
Negative stains
Electron microscopy description
  • Electron microscopy of ALHE reveals features that support the endothelial origin (Ann Pathol 1985;5:271)
Molecular / cytogenetics description
  • Molecular basis of ALHE is limited and further studies are needed to be done to understand the underlying genetic and molecular alterations driving this condition
Videos

Angiolymphoid hyperplasia with eosinophilia

Sample pathology report
  • Right cheek, excision:
    • Angiolymphoid hyperplasia with eosinophilia (see comment)
    • Comment: Histologic sections consist of skin with a dermal based lesion composed of proliferation of vascular channels accompanied by inflammatory infiltrate. Endothelial cells are showing epithelioid morphology with hobnail nuclei. The inflammatory infiltrate is composed of lymphocytes, plasma cells and eosinophils. The lesion appears to be completely excised and excision is curative.
Differential diagnosis
Additional references
Board review style question #1
A 35 year old man presented with skin lesions and was diagnosed with angiolymphoid hyperplasia with eosinophilia (ALHE) on biopsy. Which of the following is true about ALHE?

  1. Common in head and neck area
  2. HHV8 is usually positive
  3. Inflammatory infiltrate in the lesion is composed of eosinophils only
  4. Presence of mitosis is suggestive of malignancy
  5. Shows strong positivity with all vascular markers
Board review style answer #1
A. Common in head and neck area. ALHE is most common in head and neck area. Answer B is incorrect because HHV8 is negative in almost all cases. Answer C is incorrect because the inflammatory infiltrate in the lesion is usually a mixture of lymphocytes, plasma cells and eosinophils. Answer D is incorrect because ALHE can show occasional mitosis but lacks anaplasia, so presence of mitosis does not warrant a diagnosis of malignancy. Answer E is incorrect because ALHE can show stronger staining with CD31 as compared to other vascular markers.

Comment Here

Reference: Angiolymphoid hyperplasia with eosinophilia
Board review style question #2

A 23 year old woman presented with multiple reddish papules or nodules on the neck. The photograph above shows one of the lesions. What is the most likely diagnosis?

  1. Angiolymphoid hyperplasia with eosinophilia
  2. Angiosarcoma
  3. Bacillary angiomatosis
  4. Glomeruloid hemangioma
  5. Masson tumor
Board review style answer #2
A. Angiolymphoid hyperplasia with eosinophilia. Proliferating blood vessels with histiocytoid-like endothelial cells is a feature of angiolymphoid hyperplasia with eosinophilia. Answer B is incorrect because cells in angiosarcoma show marked atypia. Answer C is incorrect because bacillary angiomatosis shows proliferation of small blood vessels and is characterized by the presence of small bacilli. Answer D is incorrect because glomeruloid hemangioma is a hemangioma with glomeruloid structures associated with POEMS (polyneuropathy, organomegaly, endocrinopathy, M protein, skin changes). Answer E is incorrect because Masson tumor is an intraluminal papillary growth due to an organized thrombus.

Comment Here

Reference: Angiolymphoid hyperplasia with eosinophilia

Angiosarcoma
Definition / general
  • Malignant neoplasm with vascular differentiation
Essential features
  • Infiltrative vascular neoplasm with broad histologic profile ranging from a well differentiated neoplasm with frank vascular differentiation to a poorly differentiated tumor with epithelioid or spindled cells
  • May mimic poorly differentiated carcinoma, inflammatory process, lymphoma or melanoma
Terminology
  • Also known as hemangiosarcoma
Epidemiology
  • Classically arises in one of three scenarios:
    • Head and neck of the elderly
    • Chronic lymphedema
    • Postradiation (usually in the setting of breast cancer)
Sites
  • Sun exposed skin of the elderly (head and neck); breast with history of lymphedema or radiation therapy
Clinical features
  • Wide age range (most common in adults)
  • Presents as purple nodules or plaques
  • Highly aggressive
  • Frequent recurrence and metastasis
Prognostic factors
  • Poor prognosis - high mortality
  • Epithelioid tumors are often more aggressive
Case reports
Treatment
  • Surgical resection with negative margins
  • Chemotherapy is occasionally used
Clinical images

Images hosted on other servers:

Scalp, neck and breast lesions

Gross description
  • Violet elevated nodules with ill defined margins
Microscopic (histologic) description
  • Infiltrating, freely anastomosing channels lined by spindled to epithelioid endothelial cells with variable atypia, surrounding adnexae and dissecting dermal collagen
  • Endothelial cells may have multilayered appearance
  • May have free floating intraluminal endothelial cells (“fish in the creek”)
Microscopic (histologic) images

Contributed by Hillary Rose Elwood, M.D. and Joel Tjarks, M.D.

High grade atypical vascular tumor

Atypical vascular tumor involving dermis and subcutis

Vascular proliferation dissecting throughout the dermal collagen

Missing Image

Angiosarcoma

Positive stains
Negative stains
Molecular / cytogenetics description
  • MYC (8q24) amplification seen in great majority radiation / lymphedema associated tumors
Differential diagnosis

Apocrine tubular adenoma
Definition / general
  • Benign dermal adnexal neoplasm of apocrine derivation
  • Most common location is scalp, typically in women (M:F ratio is 1:2)
  • Also called apocrine adenoma, tubular adenoma, tubulopapillary hidradenoma, papillary tubular adenoma
  • Associated with organoid nevus, nevus sebaceus of Jadassohn and syringocystadenoma papilliferum (SCAP)
  • Rarely occurs in nose, eyelid, leg, trunk, axilla, chest, external auditory meatus, cheek, vulva
  • Clinically asymptomatic, sometimes smooth, sometimes irregular, well-defined nodule
  • Usually < 2 cm but reported up to 7 cm
Case reports
Treatment
  • Complete excision is curative
  • Malignant transformation is rare
Clinical images

Images hosted on other servers:
Mimics basal cell carcinoma

Mimics basal cell carcinoma

Gross description
  • Firm, slow growing, dermal or cutaneous skin colored nodule
Microscopic (histologic) description
  • Well circumscribed dermal neoplasm that may extend into subcutis
  • Lobular pattern of dermal and subcutaneous tubular apocrine structures often encased by a fibrous, sometimes hyalinized stroma
  • Lobules have dilated, variably sized tubules lined by two layers of epithelial cells
  • Pseudopapillae are common, but true papillae are more often associated with SCAP
  • Decapitation secretion by apical layer
  • Cuboidal to columnar cells with eosinophilic cytoplasm and round bland nuclei
  • Often hyaline and clear cell change
  • May show cyst formation with papillae or pseudopapillae protruding into the lumen
  • Variable overlyng epidermal hyperplasia
  • Rare connection with overlying epidermis
Microscopic (histologic) images

Images hosted on other servers:
Intradermal nodule

Intradermal nodule

Positive stains
  • EMA, CAM5.2 (luminal surface of tubules), CEA (luminal surface of tubules)
  • SMA (outer tubules), GCDFP-15 (focal diffuse cytoplasmic), CK7
  • S100 (myoepithelial layer)
Electron microscopy description
  • Tall columnar cells on basal lamina forming acini
  • Cells lining tubules have luminal villi and apical pinching
  • Conspicuous mitochondria, prominent golgi
  • Lipid rich cytoplasmic secretory vacuoles
  • Decapitation secretion (J Am Acad Dermatol 1984;11:639)
Differential diagnosis
  • Apocrine cystadenoma: more dilated, cystic spaces rather than tubules
  • Hidradenoma papilliferum: often has complex arborizing papillae, with more closely arranged tumor cells and glands
    • Limited to female genital region
  • Papillary apocrine carcinoma: more cytologic atypia, irregular nuclear contours and a higher mitotic rate
  • Papillary eccrine adenoma: classically has features of eccrine rather than apocrine derivation; lacks decapitation secretion; different clinical presentation and distribution
  • Syringocystadenoma papilliferum:
    • Usually connects to epidermis
    • Fibrovascular cores within papillary structures
    • Plasma cells within stroma
    • Tubular apocrine adenoma may be a variant

Apocrine tubular adenoma
Definition / general
  • Benign dermal adnexal neoplasm of apocrine derivation
  • Associated with organoid nevus, nevus sebaceus of Jadassohn and syringocystadenoma papilliferum (SCAP)
  • In most cases shows an apocrine differentiation but eccrine differentiation may be present as well
  • Reference: Int J Mol Sci 2021;22:5077
Essential features
  • Rare, benign adnexal neoplasm
  • Most common location is the scalp but can occur on other sites (Hum Pathol 2018;73:59)
  • Microscopically, it is a well circumscribed intradermal tumor composed of tubules lined by 2 cell layers or more in a fibrous, sometimes hyalinized stroma
Terminology
ICD coding
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
Epidemiology
  • Age distribution of apocrine tubular adenoma is very wide, ranging from 28 to 85 years according to one study (Hum Pathol 2018;73:59)
Sites
  • Most common location is scalp
  • Rarely occurs in the nose, eyelid, leg, trunk, axilla, chest, external auditory meatus, cheek, vulva
Pathophysiology
Etiology
Repetitive with pathophysiology
Diagrams / tables
Not relevant to this topic
Clinical features
  • Clinically asymptomatic, well defined nodule
  • Usually < 2 cm but reported up to 7 cm
Diagnosis
  • Skin biopsy
Laboratory
Not relevant to this topic
Radiology description
Not relevant to this topic
Radiology images
Not relevant to this topic
Prognostic factors
  • Lesion is benign and recurrence following excision is uncommon
Case reports
Treatment
  • Complete excision is curative
  • Malignant transformation is rare
Clinical images

Images hosted on other servers:
Mimics basal cell carcinoma

Mimics basal cell carcinoma

Gross description
  • Firm, slow growing, dermal or cutaneous, skin colored nodule
Gross images
Not relevant to this topic
Frozen section description
Not relevant to this topic
Frozen section images
Not relevant to this topic
Microscopic (histologic) description
  • May arise in the dermis without any epidermal connection
  • Well circumscribed dermal neoplasm that may extend into subcutis
  • Lobular pattern of dermal and subcutaneous tubular apocrine structures often encased by a fibrous, sometimes hyalinized stroma
  • Lobules have dilated, variably sized, well formed tubules lined by 2 layers of epithelial cells
  • Pseudopapillae are common but true papillae are more often associated with SCAP
  • Decapitation secretion by apical layer and flattened outer myoepithelial layer
  • Cuboidal to columnar cells with eosinophilic cytoplasm and round bland nuclei
  • Often hyaline and clear cell change
  • May show cyst formation with papillae or pseudopapillae protruding into the lumen
  • Variable overlying epidermal hyperplasia
  • Rare connection with overlying epidermis
  • References: J Cutan Pathol 1987;14:114, Saudi J Ophthalmol 2019;33:304, Int J Mol Sci 2021;22:5077
Microscopic (histologic) images

Contributed by Mugahed Hamza, M.B.B.S. and Sara C. Shalin, M.D., Ph.D.
Well circumscribed tumor, pseudopapillae Well circumscribed tumor, pseudopapillae Well circumscribed tumor, pseudopapillae

Well circumscribed tumor, pseudopapillae

Well formed tubules Well formed tubules

Well formed tubules


Well formed tubules

Well formed tubules

Lobular pattern, decapitation secretion Lobular pattern, decapitation secretion Lobular pattern, decapitation secretion

Lobular pattern, decapitation secretion

Virtual slides
Image blocked by login
Cytology description
Not relevant to this topic
Cytology images
Not relevant to this topic
Immunofluorescence description
Not relevant to this topic
Immunofluorescence images
Not relevant to this topic
Positive stains
Electron microscopy description
  • Tall columnar cells on basal lamina forming acini
  • Cells lining tubules have luminal microvilli and apical pinching
  • Conspicuous mitochondria, prominent Golgi
  • Lipid rich cytoplasmic secretory vacuoles
  • Decapitation secretion (J Am Acad Dermatol 1984;11:639)
Electron microscopy images
Not available
Molecular / cytogenetics description
Molecular / cytogenetics images
Not available
Videos
Not available
Sample pathology report
  • Skin, scalp, shave biopsy:
    • Apocrine tubular adenoma
Differential diagnosis
  • Apocrine cystadenoma:
    • More dilated, cystic spaces rather than tubules
  • Hidradenoma papilliferum:
    • Often has complex arborizing papillae, with more closely arranged tumor cells and glands
    • Limited to female genital region
  • Papillary apocrine carcinoma:
    • More cytologic atypia, irregular nuclear contours and a higher mitotic rate along with infiltrative growth
  • Papillary eccrine adenoma:
    • Classically has features of eccrine rather than apocrine derivation
    • Lacks decapitation secretion
    • Different clinical presentation and distribution
  • Syringocystadenoma papilliferum:
    • Usually connects to epidermis
    • Fibrovascular cores within papillary structures
    • Plasma cells within stroma
    • Tubular apocrine adenoma may be a variant
Additional references
Not relevant to this topic
Board review style question #1
Which of the following is true regarding apocrine tubular adenoma?

  1. Associated with Cowden syndrome
  2. Associated with mucinous carcinoma
  3. Most common in the extremities
  4. Sometimes associated with syringocystadenoma papilliferum
Board review style answer #1
D. Sometimes associated with syringocystadenoma papilliferum. Apocrine tubular adenoma presents on the scalp, often arising in a background of nevus sebaceus and is sometimes associated with syringocystadenoma papilliferum. Answer A is incorrect because while Cowden syndrome is associated with other cutaneous adnexal tumors, it is not associated with apocrine tubular adenoma. Answer B is incorrect because it is not associated with mucinous carcinoma. Answer C is incorrect because the most common location is on the scalp, not the extremities.

Comment Here

Reference: Apocrine tubular adenoma
Board review style question #2


A 30 year old man presents with a 1.2 cm nodule on the scalp. Sections from the tumor are shown in the photos above. This tumor can be associated with which of the following conditions?

  1. Cowden syndrome
  2. Mucinous carcinoma
  3. Muir-Torre syndrome
  4. Nevus sebaceus
Board review style answer #2
D. Nevus sebaceus. The tumor is an apocrine tubular adenoma, which often arises in a background of nevus sebaceus and is sometimes associated with syringocystadenoma papilliferum. Answer A is incorrect because while Cowden syndrome is associated with other cutaneous adnexal tumors, it is not associated with apocrine tubular adenoma. Answer B is incorrect because it is also not associated with mucinous carcinoma. Answer C is incorrect because while Muir-Torre syndrome is associated with various sebaceous cutaneous neoplasms, it is not associated with apocrine tubular adenoma.

Comment Here

Reference: Apocrine tubular adenoma

Arsenical keratosis
Definition / general
  • Arsenic is a well water contaminant, used in industrial, mining, agricultural (pesticide) and medicinal (chemotherapy) substances (Toxicol Sci 2011;123:305)
  • Often causes hyperkeratotic lesions of skin called arsenical keratoses
  • Risk factor for Bowen disease, squamous cell carcinoma, basal cell carcinoma and carcinomas of lung, bladder and kidney
  • Skin related problems are rare in U.S.
Clinical features
  • Acute arsenical dermatitis or long term sequelae as a diffuse erythematous papular or pustular bullous dermatosis that can progress to exfoliative dermatitis
  • "Rain drops on a dusty road": hyperpigmented macules with small foci of hypopigmentation and darker hyperpigmentation in trunk, areola and flexural
  • Transverse white nail striations
  • Palmar and plantar keratoses 2+ years after exposure; may transform to Bowen’s disease, squamous cell carcinoma and superficial basal cell carcinoma
Microscopic (histologic) description
Microscopic (histologic) images

Images hosted on other servers:

Left: normal;
right: hyperkeratotic
skin due to arsenic


Arteriovenous malformation (pending)

Atypical fibroxanthoma
Definition / general
Essential features
  • Low grade malignant cutaneous neoplasm
  • Usually presents on the sun exposed skin (e.g. head and neck) of elderly patients with a slight male predominance
  • Histologically, a dermal based, well circumscribed tumor composed of pleomorphic, irregularly arranged, spindled to epithelioid cells with numerous mitotic figures
  • Diagnosis of exclusion that requires the evaluation of multiple immunohistochemical studies to rule out other differential diagnoses
  • Size, substantial involvement of the subcutis or beyond, perineural invasion, lymphovascular invasion or necrosis may suggest a diagnosis of pleomorphic dermal sarcoma instead
Terminology
  • Dermal variant of superficial undifferentiated pleomorphic sarcoma / malignant fibrous histiocytoma
ICD coding
  • ICD-O: 8830/1 - atypical fibroxanthoma
Epidemiology
  • Elderly patients with a slight male predominance (Am J Dermatopathol 2010;32:533)
  • Rarely, young patients with Li-Fraumeni syndrome or xeroderma pigmentosum
Sites
  • Sun exposed skin (usually head and neck)
Pathophysiology
  • Unclear but likely associated with ultraviolet induced damage
Etiology
  • Associations with ultraviolet radiation exposure and ultraviolet radiation signature mutations in TP53, Li-Fraumeni syndrome, xeroderma pigmentosum, radiotherapy, immunosuppression, organ transplantation
Clinical features
Diagnosis
  • Diagnosis of exclusion
  • Requires use of ancillary techniques (i.e. immunohistochemistry panels) to rule out other differential diagnoses
  • Rendering a definitive diagnosis on superficial biopsies alone may be difficult, given the differential diagnosis of pleomorphic dermal sarcoma
Prognostic factors
  • Low grade tumor with an almost always benign clinical course
  • Recurrence may occur due to incomplete surgical removal
  • Metastasis is rare (Am J Dermatopathol 2015;37:455)
Case reports
Treatment
Clinical images

Images hosted on other servers:

Lesion on ear

Gross description
  • Single red to pink nodule or polypoid tumor, usually less than 2 cm in diameter and may be ulcerated and covered with serum crust
Microscopic (histologic) description
  • Dermal based, well circumscribed tumor
  • Composed of irregularly arranged spindled to epithelioid cells with no involvement of the subcutis
  • Necrosis, lymphovascular invasion and perineural invasion should not be present
  • Cytomorphologically, lesional cells are highly bizarre and atypical, with marked pleomorphism in size and shape, abundant eosinophilic cytoplasm, occasional multinucleation and numerous mitotic figures, including atypical forms
  • Epidermal collarette, ulceration and actinic changes, including prominent solar elastosis, may be present
  • No connection to the overlying epidermis and no in situ component
  • Histologic variants (Am J Dermatopathol 2010;32:533):
  • May have forms with osteoclast giant cells, osteoid and chondroid formation (Am J Dermatopathol 2010;32:533)
Microscopic (histologic) images

Contributed by Pavandeep Gill, M.D. and Phyu Aung, M.D., Ph.D.
Dermal based tumor

Dermal based tumor

Highly atypical cells

Highly atypical cells

Solar elastosis and actinic changes

Solar elastosis and actinic changes

CD68

CD68

MiTF

MITF

Negative stains
Molecular / cytogenetics description
Videos

Atypical fibroxanthoma and mimics

Atypical fibroxanthoma pearls

Sample pathology report
  • Vertex scalp, skin shave:
    • Malignant spindle cell neoplasm predominantly in dermis, consistent with atypical fibroxanthoma, present at tissue edges (see comment)
    • Comment: Sections show a dermal, poorly differentiated pleomorphic neoplasm, characterized by large and bizarre spindled to epithelioid cells with abundant eosinophilic cytoplasm, occasional multinucleation and numerous mitotic figures. Adjacent dermis contains solar elastosis. The overlying epidermis does not have a connection to the tumor. Immunohistochemical studies show that lesional cells are positive for CD68 and negative for CAM5.2, AE1 / AE3, CD31 and S100. Overall, these features support the above interpretation.
Differential diagnosis
  • Pleomorphic dermal sarcoma:
    • Histologically similar to atypical fibroxanthoma but larger and shows substantial involvement of the subcutis or beyond, perineural invasion, lymphovascular invasion or necrosis
    • Behaves in a more aggressive manner than atypical fibroxanthoma
  • Melanoma:
    • Usually associated with an in situ component in the overlying epidermis
    • Melanin pigment, lymphovascular invasion, perineural invasion, involvement of the subcutis and microsatellites may also be present
    • Lesional cells are positive for melanocytic immunohistochemical markers (e.g. SOX10, S100, MelanA, HMB45, tyrosinase and MITF), although reactivity to melanocytic markers other than S100 and SOX10 may be lost in spindle cell / desmoplastic variants
  • Squamous cell carcinoma:
    • Usually associated with a connection to the overlying epidermis and an in situ component
    • Keratin, intercellular bridging, invasion of the subcutis, perineural invasion and lymphovascular invasion may be present
    • Lesional cells are positive for p40 (Am J Surg Pathol 2014;38:1102)
  • Angiosarcoma:
    • Usually positive for vascular immunohistochemical markers, such as CD31, CD34 and ERG
  • Atypical fibrous histiocytoma:
    • Usually presents in younger patients, lacks prominent actinic changes and may have areas of classic dermatofibroma in the periphery of the tumor
  • Leiomyosarcoma:
    • Usually positive for smooth muscle markers, such as smooth muscle myosin and desmin
  • Metastatic carcinoma:
    • Should be considered in the differential diagnosis for patients with a known history of carcinoma
    • Lesional cells are usually positive for pancytokeratin and other immunohistochemical markers similar to the primary tumor
  • Superficial CD34+ fibroblastic tumor:
    • May overlap with histologic variants of atypical fibroxanthoma
    • Bizarre atypical nuclei but typically only rare mitoses
    • Diffuse CD34 and frequent keratin positivity
Board review style question #1

The finding of which of the following additional histologic features in the lesion shown above would most likely favor a diagnosis of melanoma over atypical fibroxanthoma?

  1. Presence of an in situ component
  2. Presence of numerous mitotic figures, including atypical forms
  3. Presence of pigment
  4. Reactivity for MITF
Board review style answer #1
A. Presence of an in situ component. Choice B is incorrect because numerous mitotic figures, including atypical forms, are seen in both melanoma and AFX. Choice C is incorrect because pigment can be seen in both AFX (hemosiderin) and melanoma (melanin). Choice D is incorrect because reactivity for anti-MiTF may be seen in both melanoma and AFX.

Comment Here

Reference: Atypical fibroxanthoma
Board review style question #2
Assuming all other listed markers are negative, a positive immunohistochemical study with which marker is most consistent with a diagnosis of atypical fibroxanthoma?

  1. CD10
  2. p40
  3. S100
  4. Smooth muscle myosin
Board review style answer #2
A. CD10. Choice B is incorrect because p40 positivity suggests a diagnosis of squamous cell carcinoma. Choice C is incorrect because S100 positivity suggests a diagnosis of melanoma or neural tumor. Choice D is incorrect because smooth muscle myosin positivity suggests a diagnosis of smooth muscle tumor.

Comment Here

Reference: Atypical fibroxanthoma

Atypical smooth muscle tumor
Definition / general
  • Superficial dermal based smooth muscle tumor which is biologically different from deep leiomyosarcoma
  • Typically larger than leiomyomas with vascular invasion
  • Recurs, but only rarely metastasizes
  • May be associated with HIV infection
Case reports
Microscopic (histologic) description
  • Cellular lesions of smooth muscle type cells with atypia, necrosis and mitotic activity
  • May have prominent vascular pattern, clear cell features, desmoplasia
Microscopic (histologic) images

Images hosted on other servers:

Subcutaneous spindle cell neoplasm

Spindle cells and osteoclast-like giant cells

SMA+ and CD68+

Positive stains

Atypical vascular lesion post radiation

Basal cell carcinoma
Definition / general
  • Basal cell carcinoma (BCC) arises from the interfollicular or follicular epithelium
  • Most common malignant tumor type in humans
  • Local aggressive course
  • Low disease associated death rate; metastases to lung and bone exceptionally rare
  • When multiple, associated with a number of genetic conditions, including basal cell nevus (Gorlin), Bazex-Dupré-Christol, Rombo syndromes and xeroderma pigmentosum
Essential features
  • Nests of basaloid cells with peripheral palisading associated with a fibromyxoid stroma
Terminology
  • Basal cell epithelioma
  • Rodent ulcer
  • Basalioma
  • Fibroepithelioma of Pinkus
ICD coding
  • ICD-O:
    • 8090/3 - Basal cell carcinoma, NOS
    • 8097/3 - Nodular basal cell carcinoma
    • 8091/3 - Superficial basal cell carcinoma
    • 8097/3 - Micronodular basal cell carcinoma
    • 8092/3 - Infiltrating basal cell carcinoma
    • 8092/3 - Morpheaform basal cell carcinoma
    • 8094/3 - Basosquamous carcinoma
    • 8090/3 - Pigmented basal cell carcinoma
    • 8092/3 - Basal cell carcinoma with sarcomatoid differentiation (metaplastic BCC)
    • 8090/3 - Basal cell carcinoma with adnexal differentiation
    • 8093/3 - Fibroepithelial basal cell carcinoma
    • N/A - Basomelanocytic tumor
Epidemiology
Sites
Pathophysiology
Etiology
Clinical features
  • Nodular variant usually presents as a pearly pink or flesh colored papule or nodule with arborizing and branching vessels (Br J Dermatol 2002;147:41)
  • In the past, ulceration of a nodular BCC gave rise to the term rodent ulcer
  • Tumor with ulceration has characteristic rolled borders
  • Superficial variant presents with scaly macules, patches or plaques with an erythematous surface
  • Pigmented variant resembles a nodular or superficial BCC but has a pigmented surface and can be clinically mistaken for a melanoma
  • Neglected tumors may result in massive skin, soft tissue and bone destruction with severe disfiguration
Diagnosis
  • Clinical features
  • Dermatoscopy
  • Histological features
Prognostic factors
  • Histological type associated with risk of local recurrence
    • Lower risk: nodular, superficial, pigmented, infundibulocystic, fibroepithelial
    • Higher risk: basosquamous, sclerosing / morpheaform, keloidal, infiltrating, BCC with sarcomatoid differentiation and micronodular variants
  • Circumscription of tumor margins
  • Presence of perineural and lymphovascular invasion
  • Status of surgical margins
  • Tumor size
  • Localization (part of skin affected, e.g. head and neck, trunk, etc.)
  • History of nonmelanoma skin cancer
  • Radiation therapy
  • PUVA therapy
  • Immunosuppression
  • Reference: NCCN: Basal Cell Skin Cancer Evidence Blocks [Accessed 15 December 2020]
Case reports
Treatment
  • Surgery
  • Mohs micrographic surgery
  • Curettage and electrodesiccation for tumors with lower risk of local recurrence
  • Radiation therapy
  • Topical treatment (5-fluorouracil, imiquimod)
  • Photodynamic therapy with aminolevulinic acid or porfimer sodium
  • Nicotinamide (can be used for prophylactic treatment)
  • Systemic therapy (sonidegib and vismodegib are hedgehog pathway inhibitors [HhIs])
  • Reference: NCCN: Basal Cell Skin Cancer Evidence Blocks [Accessed 15 December 2020]
Clinical images

Contributed by Sergiy Vasylenko, M.D., Bohdan Lytvynenko, M.D. and Arghya Banerjee, M.D.
Nodular BCC Nodular BCC

Nodular BCC

Ulcerative BCC

Ulcerative BCC

Pigmented, ulcerated BCC

Pigmented, ulcerated BCC

Superficial BCC Superficial BCC

Superficial BCC


Pigmented BCC Pigmented BCC

Pigmented BCC

Infiltrative BCC Infiltrative BCC

Infiltrative BCC

Gross description
  • Variable, mirroring the clinical variants described above
Frozen section description
Microscopic (histologic) description
Common variants (Calonje: McKee's Pathology of the Skin, 5th Edition, 2019)
  • Nodular and nodulocystic BCC
    • Relatively circumscribed mass
    • Epidermal or follicular attachment variably present
    • Large basaloid lobules with peripheral nuclear palisade
    • Lobules may be solid or show central cyst formation due to excessive mucin production
    • Fibromyxoid stroma
    • Cleft formation between tumor lobules and stroma
    • Pleomorphism is generally mild
    • Variable mitotic activity and apoptosis
    • Sometimes necrosis en masse
  • Adenoid BCC
    • Reticulate pseudoglandular pattern of basaloid cells
    • Mucinous stroma
    • Can mimic true gland formation, resulting in diagnostic confusion with a sweat gland adenocarcinoma, e.g. adenoid cystic carcinoma
  • Micronodular BCC
    • Small basaloid nests
    • Peripheral palisading less prominent
    • Retraction artifact usually absent
    • Can diffusely infiltrate the dermis and extend into the subcutis
  • Infiltrative BCC
    • Small irregular clumps of basaloid cells
    • Limited peripheral palisading
    • Stroma loose and mucin may be prominent
    • Extensive spread
    • Perineural invasion can be seen
  • Morpheaform (sclerosing, morphoeic) BCC
    • Thin strands and nests of basaloid cells
    • Limited peripheral palisading
    • Stroma is dense and sclerotic
    • Extensive spread
    • Perineural invasion can be seen
  • Keratotic BCC
    • Horn cyst formation
  • Basosquamous (metatypical) BCC
    • Biphasic tumor
    • Foci of neoplastic squamous differentiation
  • Pigmented BCC
    • Nodular and superficial variants can be pigmented
    • Colonization of tumor's complexes with melanocytes
    • Stromal melanophages
  • Superficial BCC
    • Isolated basaloid lobules projecting from the lower margin of the epidermis
  • Ulcerative BCC
    • Ulceration
    • Highly infiltrative growth pattern
    • Less commonly nodulocystic variant with ulceration
  • BCC, fibroepitheliomatous type (fibroepithelioma of Pinkus)
    • Anastomosing strands and cords of basaloid cells connected to the epidermis
    • Peripheral nuclear palisading with a formation of follicular germ-like structures
    • Rarely, isthmic differentiation is present
    • Fibrotic stroma that can differentiate towards follicular papillae in the areas of germ-like structure formation

Rare variants
  • Pleomorphic (giant cell, BCC with monster cells) BCC
    • Mitotic activity
    • Apoptosis
    • Сellular pleomorphism and giant cell formation
    • Atypia has no prognostic significance
  • Clear cell BCC
    • Focal or total clear cell change with clear to finely granular eosinophilic cytoplasm due to lysosomal degeneration
    • Peripheral palisading is usually preserved
  • Signet ring cell BCC
    • Tumor cells with laterally displaced nuclei
    • May show positivity for S100 protein, glial fibrillary acidic protein and smooth muscle actin, suggesting myoepithelial differentiation
    • Referred to as BCC with myoepithelial differentiation
  • Granular BCC
    • Tumor cells with abundant granular eosinophilic cytoplasm
  • BCC with differentiation towards adnexal structures
    • Tumor complexes can show differentiation towards sebaceous, follicular, eccrine or apocrine structures
  • Infundibulocystic BCC
    • Synonym: BCC with follicular differentiation
    • Superficial dermal proliferation of basaloid cells arranged in anastomosing cords and strands
    • Peripheral palisading is present
    • Stroma is typically scanty
    • Infundibular cysts are present
  • Metaplastic BCC
    • Stromal malignant metaplastic features (carcinosarcoma)
    • Chondroid, osteoid and smooth muscle differentiation may be seen
  • BCC with matricial differentiation (shadow cell BCC)
    • Tumor cells show differentiation towards matricial cells of the hair follicle displaying shadow cells and trichohyaline granules
  • Keloidal BCC
    • Stroma shows thick, sclerotic collagen bundles
  • BCC with thickened basement membrane
    • Tumor complexes surround by thickened basement membrane
  • Basomelanocytic tumor
    • Combined and intermixed basal cell and melanomatous elements
  • BCC with neuroid type nuclear palisading
    • Central nuclear palisading reminiscent of that seen in schwannoma
Microscopic (histologic) images

Contributed by Antonina Kalmykova, M.D., Phillip H. McKee, M.D., Sate Hamza, M.D., Eduardo Calonje, M.D.,
Wayne Grayson, M.B.Ch.B., Ph.D., James Sampson, M.B.B.S., M.Sc. and Assia Bassarova, M.D., Ph.D.

Nodular BCC Nodular BCC

Nodular BCC

Nodular BCC Nodular BCC

Nodular BCC

Nodulocystic BCC

Nodulocystic BCC

Pigmented BCC

Pigmented BCC


Ulcerative BCC

Ulcerative BCC

Superficial BCC

Superficial BCC

Adenoid BCC

Adenoid BCC

Infiltrative BCC

Infiltrative BCC

Morpheaform BCC

Morpheaform BCC

Keloidal BCC

Keloidal BCC


Micronodular BCC

Micronodular BCC

Keratotic BCC

Keratotic BCC

Granular cell BCC

Granular cell BCC

BCC with matricial differentiation

BCC with matricial differentiation

Clear cell BCC

Clear cell BCC

Basosquamous carcinoma

Basosquamous carcinoma


BCC with thickened basement membrane

BCC with thickened basement membrane

Basomelanocytic tumor

Basomelanocytic tumor

Basomelanocytic tumor: AE1 / AE3

Basomelanocytic tumor: AE1 / AE3

Basomelanocytic tumor: melanA

Basomelanocytic tumor: melanA

Metaplastic BCC Metaplastic BCC

Metaplastic BCC


Metaplastic BCC

Metaplastic BCC

Metaplastic BCC: BerEP4

Metaplastic BCC: BerEP4

Metaplastic BCC: SMA

Metaplastic BCC: SMA

Fibroepithelial BCC (Pinkus tumor) Fibroepithelial BCC (Pinkus tumor)

Fibroepithelial BCC (Pinkus tumor)


Rippled pattern BCC

Rippled pattern BCC

Rippled pattern BCC: p63

Rippled pattern BCC: p63

Rippled pattern BCC: CK8/18

Rippled pattern BCC: CK8/18

Rippled pattern BCC: BerEP4

Rippled pattern BCC: BerEP4

Cytology description
  • Sensitivity and specificity of exfoliative cytology estimate 97.5% (95% CI 94.5% to 98.9%) and 90.1% (95% CI 81.1% to 95.1%) respectively and can result in wrong or false positive result (Cochrane Database Syst Rev 2018;12:CD013187)
  • Complexes of tightly packed basaloid cells with palisading in the periphery
  • High nuclear/cytoplasmic ratio, minimal atypia and mitotic activity
Positive stains
Molecular / cytogenetics description
  • Chromosomal gains at 6p (47%), 6q (20%), 9p (20%), 7 (13%) and X (13%)
  • Regional loss on 9q in 33% of tested tumors encompassing 9q22.3 to which the Patched gene has been mapped (J Invest Dermatol 2001;117:683)
Videos

BCC discussed by Dr. Jerad Gardner

Malignant keratinocytic neoplasms discussed by Dr. Clay J. Cockerell

Sample pathology report
  • Face, excision:
    • Nodular basal cell carcinoma (LVI0 Pn0 R0) (ICD-O code 8097/3) (see synoptic report)
    • Synoptic report:
      • Tumor site: face
      • Procedure: excision
      • Histological type: nodular basal cell carcinoma
      • Depth of invasion: 2.5 mm
      • Tumor size (greatest dimension): 7 mm
      • Anatomic level (Clark level): IV
      • Lymphovascular invasion (LVI): 0
      • Perineural invasion (Pn): 0
      • Peripheral margins: uninvolved
      • Deep margin: uninvolved
  • Reference: NCCN: Basal Cell Skin Cancer Evidence Blocks [Accessed 15 December 2020]
Differential diagnosis
  • Follicular induction (also known as follicular basal cell hyperplasia, epidermal basaloid cell hyperplasia and basaloid epidermal proliferation) versus superficial BCC:
    • Usually seen above dermatofibroma, less common above nevi
    • Follicular differentiation (germinative buds and papillary mesenchymal bodies) shows clear cell hyperplasia, epidermal hyperplasia between sites of follicular induction
    • No atypical nuclei, increased mitotic figures or apoptotic bodies
    • CK20+ colonizing Merkel cells
  • Trichoepithelioma / trichoblastoma versus nodular BCC:
    • Delicate perifollicular-like stroma with small spindled cells
    • Papillary mesenchymal bodies
    • No prominent cleft retraction, myxoinflammatory stroma, significant mitotic activity (is sometimes marked in trichoblastoma and should not be misconstrued as implying malignant trichoblastoma), cytologic atypia or tumor necrosis
  • Desmoplastic trichoepithelioma versus morpheaform BCC:
    • Does not extend into the deep dermis
    • Architectural symmetry, horn cysts with calcification, granulomatous inflammation and follicular / sebaceous / infundibular differentiation
    • No mitotic figures, ductal differentiation, cytologic atypia, tumor to stroma clefting or perineural infiltration
    • CK20+ colonizing Merkel cells, androgen receptor-
    • PHLDA1+ (highly specific for trichoepithelioma / trichoblastoma, although expression in micronodular BCC has been reported)
    • Note: no single IHC marker can differentiate trichoepithelioma and BCC
  • Basaloid follicular hamartoma versus infundibulocystic BCC:
    • Cystic structures (uncommon in BCC, except infundibulocystic type)
    • No tumor necrosis, mitotic activity, cytologic atypia, myxoinflammatory stroma or peripheral clefting
    • CD34+ (outlines tumor islands)
    • CK20+ (Merkel cells): utility is uncertain for distinction from infundibulocystic BCC
  • Tumor of follicular infundibulum versus superficial BCC:
    • Cytoplasmic pallor (due to glycogen) and conspicuous basement membrane
    • No cytologic atypia, mitotic activity, no myxoid inflammatory stroma or stroma to tumor clefting
    • Colonizing CK20+ Merkel cells, BerEP4-
    • Elastin may demonstrate an elastin fiber network at the base of the lesion
  • Syringoma versus morpheaform BCC:
    • Well circumscribed, superficial
    • Ductal differentiation (reminiscent of tadpoles)
    • No tumor necrosis, mitotic activity, cytologic atypia, myxoinflammatory stroma, peripheral clefting or perineural invasion
    • CD200+, claudin 4+, EMA+, CEA+ (highlight lumina)
  • Squamous cell carcinoma (basaloid, clear cell, sarcomatoid):
    • Shows an intraepidermal component, pagetoid spread
    • No peripheral palisading, clefting or myxoinflammatory stroma
    • Areas with conventional features like keratinization, keratin pearls
    • BerEP4- (also absent in squamous areas of basosquamous carcinoma)
    • EMA+ (may be expressed in squamous areas of BCC), CD44+
  • Microcystic adnexal carcinoma versus morpheaform BCC:
    • Superficial keratocysts (infundibular or isthmic), ductal differentiation with eosinophilic luminal secretions
    • Frequently invades nerves (can't discriminate)
    • No peripheral palisading, mitotic activity, tumor to stroma retraction or myxoinflammatory stroma
    • CEA+, EMA+ in ductal structures
    • CK15+, BerEP4 variable (mixed reports)
    • p63 highlights ductal myoepithelial cells
  • Sebaceous carcinoma versus BCC (nodular, clear cell):
    • Intraepidermal pagetoid spread
    • No peripheral clefting
    • May have evidence of sebaceous, lobular architecture
    • Often relatively greater cytologic atypia and less basaloid morphology
    • Diffuse androgen receptor+ (versus focally positive in BCC)
    • Low molecular weight CK+, EMA+
    • BerEP4-, adipophilin+, perilipin+
  • Merkel cell carcinoma:
    • Rarely may show intraepidermal with pagetoid spread
    • No / focal peripheral palisading
    • Homogenous salt and pepper chromatin
    • Nuclear molding
    • Abundant mitotic activity and necrosis
    • MCPyV 70 - 80% (highly specific marker)
    • CK20+ (paranuclear, dot-like pattern)
    • Neuroendocrine markers expressed but rarely also positive in BCC
  • Adenoid cystic carcinoma versus adenoid BCC:
    • Well defined cribriform patterns
    • No peripheral palisading, continuity with the epidermis or adjacent hair follicle or retraction artifacts between the tumor islands and stroma
    • Typically shows hyaline material lining the lumina of the ducts and sometimes also present in the stroma
    • CD117+ (20% of BCC CD117+), CD43+ (40%), CK7+, CEA+, EMA+ (in ductal structures)
    • p63+, SMA+, calponin+ (myoepithelial cell population at periphery of tumor islands)
  • Clear cell melanoma (balloon cell melanoma) versus clear cell BCC:
    • Intraepidermal pagetoid spread, variable melanin pigment
    • No peripheral palisading, stroma to tumor clefting or myxoinflammatory stroma
    • SOX10+, S100+, HMB45+, MelanA+, tyrosinase+, BerEP4-
  • Clear cell eccrine porocarcinoma versus clear cell BCC:
    • No myxoinflammatory stroma (sometimes may be present), peripheral clefting or conventional poroma-like areas
    • CEA+, EMA+, BerEP4-
  • Clear cell hidradenocarcinoma versus clear cell BCC:
    • No peripheral palisading, clefting or myxoinflammatory stroma
    • CEA+, S100+, BerEP4-
  • Trichilemmal carcinoma versus clear cell BCC:
    • No conventional areas of BCC
    • Contains abundant glycogen (PAS+), CEA-, EMA-
  • Pilomatrix carcinoma versus BCC with matricial differentiation:
    • No conventional areas of BCC
  • Pigmented reticulated seborrheic keratosis versus BCC with follicular differentiation:
    • No peripheral palisading, myxoinflammatory stroma, peripheral clefting, atypical nuclei, increased mitotic figures or apoptotic bodies
    • CK20+ colonizing Merkel cells may be present
  • Cylindroma versus BCC with thick basement membrane:
    • No conventional areas of BCC
    • CEA+, S100+
Board review style question #1

This image comes from a tumor on the face of an elderly patient. What is the most likely diagnosis?

  1. Sclerosing sweat ductal carcinoma
  2. Microcystic adnexal carcinoma
  3. Basal cell carcinoma with ductal differentiation
  4. Secretory carcinoma
  5. Metastatic adenocarcinoma
Board review style answer #1
C. Basal cell carcinoma with ductal differentiation

Comment Here

Reference: Basal cell carcinoma
Board review style question #2

This image comes from the head of an elderly patient. What is the most likely diagnosis?

  1. Sebaceoma
  2. Sebaceous carcinoma
  3. Sebaceous adenoma
  4. Basal cell carcinoma with sebaceous differentiation
  5. Porocarcinoma
Board review style answer #2
D. Basal cell carcinoma with sebaceous differentiation

Comment Here

Reference: Basal cell carcinoma

Basaloid follicular hamartoma
Definition / general
  • Rare, benign cutaneous lesion associated with inherited syndromes
  • Confused histologically with infundibulocystic basal cell carcinoma
Essential features
  • Benign cutaneous hair follicle hamartoma
  • Can occur as papule(s) or plaque
  • May be solitary, linear or diffuse in distribution (Indian J Dermatol Venereol Leprol 2019;85:60)
  • Confused histologically with basal cell carcinoma or other benign basaloid dermal growths
  • May occur spontaneously or as the result of inherited disorders
Terminology
  • Basaloid follicular hamartoma
  • Generalized basaloid follicular hamartoma syndrome: autosomal dominant variant
  • Linear unilateral basal cell nevus with comedones: childhood form occurring in a linear distribution, often along the lines of Blaschko and may have associated comedones
  • Generalized hair follicle hamartoma: multiple basaloid follicular hamartomas and coexisting alopecia and myasthenia gravis (Patterson: Weedon's Skin Pathology, 4th Edition, 2015, Chapter 33)
ICD coding
  • ICD-10: Q82.5 - congenital nonneoplastic nevus
Epidemiology
  • Associated with certain autoimmune diseases: myasthenia gravis, alopecia, systemic lupus erythematosus
  • Happle-Tinschert syndrome (Dermatology 2009;218:221)
  • Bazex syndrome, aka Bazex-Dupre-Christol syndrome (X-linked dominant)
  • Nevoid basal cell carcinoma syndrome, aka Gorlin syndrome, aka basal cell nevus syndrome
  • Generalized basaloid follicular hamartoma (autosomal dominant mutation) (J Cutan Pathol 2008;35:477)
  • Brown-Crounse syndrome
Sites
  • Skin of the face, trunk, limbs, scalp, axilla and pubic area
  • Distribution may be localized or generalized depending on the variant (Arch Pathol Lab Med 2010;134:1215)
Pathophysiology
  • Can occur sporadically or in association with genetic mutations
  • Mutated PTCH (protein patched homolog) gene leads to disrupted tumor suppressor function
  • PTCH abnormally binds sonic hedgehog (Shh), a protein signal involved in embryogenesis
  • Constitutive activation of downstream signaling in transcription factors like Gli-1 and abnormal growth ensues (Arch Pathol Lab Med 2010;134:1215)
  • Associated with autoimmune disease
Clinical features
  • May occur as solitary or multiple hypopigmented to hyperpigmented papules, commonly over the central face
  • May occur as a plaque or coalescing papules
  • Lesions may be associated with alopecia, hypertrichosis or open comedones (J Cutan Pathol 2008;35:477)
  • Unilateral variants may follow lines of Blaschko and can be associated with underlying neurological findings (Korean J Radiol 2014;15:534)
Diagnosis
  • Diagnosis relies on recognizing the specific pathologic features
  • Correlation with clinical findings is required
  • Some authors have discussed the utility of immunohistochemical studies in distinguishing basaloid follicular hamartoma from basal cell carcinoma (Arch Pathol Lab Med 2017;141:1490)
  • Significant morphologic overlap between other benign tumors of the follicular infundibulum as well as infundibulocystic basal cell carcinoma exists; some have argued that infundibulocystic basal cell carcinoma and basaloid follicular hamartoma are the same entity (J Cutan Pathol 2014;41:916)
Case reports
Treatment
  • Not officially considered a premalignant lesion
  • Close follow up to monitor for progression to basal cell carcinoma should be considered
  • Some patients may be at greater risk for development of malignancy such as those with basal cell nevus syndrome
  • Those that arise in the setting of autoimmune disease may resolve with treatment of the corresponding autoimmune disorder
  • Surgical excision with carbon oxide laser therapy and dermabrasion have been successfully used in a 14 year old boy with unilateral facial basaloid follicular hamartoma with no recurrence at 11 years (J Dermatol 2017;44:e278)
  • Pulsed dye laser therapy was used with some success in the treatment of acquired basaloid follicular hamartomas on the face of a 68 year old man (J Clin Aesthet Dermatol 2018;11:39)
  • Three children with basal cell nevus syndrome who had extensive basal cell carcinomas and basaloid follicular hamartomas were treated with 5-aminolevulenic acid photodynamic therapy with satisfactory cosmetic results lasting at least 6 years (Arch Dermatol 2005;141:60)
  • Oral and topical retinoids have been used with some efficacy in an isolated case (J Am Acad Dermatol 2003;49:1067)
Clinical images

Images hosted on other servers:
Missing Image

Hyperpigmented papules on antecubital fossa

Missing Image

Hyperpigmented papules behind ear and on nose

Missing Image

Dark brown papules over the lateral face

Frozen section description
  • Basaloid follicular hamartomas may be seen on frozen section, especially during Mohs surgery
  • The same diagnostic criteria apply for frozen section diagnosis as for the evaluation of paraffin embedded tissue
  • Distinguishing between basaloid follicular hamartoma and basal cell carcinoma on frozen section is challenging
Microscopic (histologic) description
  • Anastomosing strands of basaloid and squamoid cells with connection to the overlying epithelium
  • Should only arise where normal hair follicles are present
  • Bland cells without atypia, mitoses or apoptotic bodies
  • Loose, cellular or myxoid stroma
  • Horns, cysts and pigmentation may be present
  • If peripheral palisading and retraction are present, they should be focal and less than the amount typically seen within basal cell carcinoma (Arch Pathol Lab Med 2010;134:1215)
Microscopic (histologic) images

Contributed by the University of Colorado
Missing Image

Basaloid islands

Missing Image

Arborizing

Missing Image

Bland cells

Missing Image

Altered stroma

Missing Image

Epidermal connection


Missing Image

Bland

Missing Image

Branching and fenestrations

Missing Image

Folliculocentric

Missing Image

Peripheral palisading

Missing Image

Cellular stroma


Missing Image

No clefting

Missing Image

Multiple nests

Missing Image

Focal cystic dilation

Missing Image

Basaloid cords

Missing Image

Irregularly shaped

Positive stains
  • BCL2: stains peripheral basaloid cells; a diffuse staining pattern is observed in all basaloid cells in basal cell carcinoma
  • CD34: stains the stroma around basaloid cells; the stroma in basal cell carcinoma may only rarely stain with CD34 (J Cutan Pathol 2001;28:538)
  • CD10: stains the stroma around basaloid cells; both the stroma and basaloid cells in basal cell carcinoma may stain with CD10
  • P40: strongly positive in basaloid cells
  • CK20: focal positivity has been reported (J Cutan Pathol 2014;41:916)
  • There is no specific immunohistochemical study for basaloid follicular hamartoma and studies analyzing sensitivity and specificity of markers are limited by the availability of this rare tumor
Negative stains
  • Immunohistochemistry has not been widely studied in basaloid follicular hamartoma
Molecular / cytogenetics description
  • Targeted next generation sequencing may be helpful in the diagnosis of patients with syndromic phenotypes
  • The first family described with generalized basaloid follicular hamartoma was in North Carolina; there were 6 generations of affected members who had multiple flesh colored and hyperpigmented papules over the face, neck, trunk and arms, palmar / solar pits, scalp alopecia, milia and comedones (J Am Acad Dermatol 2000;43:189)
  • The PTCH1 gene on chromosome 9q23 is implicated in both generalized basaloid follicular hamartoma syndrome (GBFH) and Gorlin syndrome; basaloid follicular hamartomas occur in both syndromes and some have suggested that these two diseases are on a spectrum (Anticancer Res 2018;38:471)
  • Basal cell nevus syndrome may also arise as a result of PTCH2 mutation or SUFU mutation, a signaling molecule in the same pathway; as opposed to generalized basaloid follicular hamartoma syndrome, basal cell nevus syndrome is defined by the presence of jaw keratocysts, calcification of the falx cerebri, bifid ribs and macrocephaly
Videos

Clinical phenotype and molecular genetics of Basal cell nevus syndrome

Sample pathology report
  • Skin, cheek, biopsy:
    • Cutaneous adnexal neoplasm, morphologically consistent with basaloid follicular hamartoma (see comment)
    • Comment: Basaloid follicular hamartoma demonstrates significant overlap between basal cell carcinoma and has been shown to progress to malignancy in some cases. Increased surveillance or complete surgical excision may be required depending upon the clinical circumstances.
    • Microscopic description: Anastomosing cords of bland basaloid cells with peripheral palisading and surrounding cellular stroma. Rare papillary mesenchymal bodies are noted. Absent are mitoses, apoptotic bodies and atypical nuclear features.
Differential diagnosis
  • Infundibulocystic basal cell carcinoma:
    • Strikingly similar appearance to basaloid follicular hamartoma but is more likely to have atypia and mitoses
    • Stromal retraction should be more prominent in basal cell carcinoma
    • Some have discussed the presence of a myxoinflammatory stroma as a helpful distinguishing feature
    • Clinical correlation and immunohistochemistry may aid in the diagnosis (Arch Pathol Lab Med 2017;141:1490)
  • Trichoepithelioma / trichoblastoma:
    • Benign tumor composed of separate islands of basaloid cells which may have peripheral palisading
    • Islands may branch and are surrounded by a loose to fibrotic stroma
    • Papillary mesenchymal bodies may be seen
    • The presence of distinct rounded islands as opposed to multiple branching anastomoses signals the diagnosis
  • Tumor of the follicular infundibulum:
    • Helpful distinguishing features are a growth pattern along the dermal epidermal junction resembling superficial basal cell carcinoma with fenestrated areas
    • Another helpful feature is the presence of pale staining glycogen containing cells that are reminiscent of trichilemmoma
  • Pilar sheath acanthoma:
    • Cystically dilated follicle with radiating acanthotic epithelial projections
  • Trichofolliculoma:
    • Central large dilated follicle surround by smaller secondary follicles
>
Board review style question #1

    An elderly man presents to the dermatology clinic with a flesh colored papule over the lateral cheek without arborizing vessels or crust. Biopsy is shown. What is the most likely diagnosis?

  1. Basal cell carcinoma
  2. Basaloid follicular hamartoma
  3. Syringoma
  4. Trichoblastoma
  5. Spiradenoma
Board review style answer #1
B. Basaloid follicular hamartoma

Explanation: A) Basal cell carcinoma should have atypia; C) While, syringoma will have basaloid cells in the dermis, they are separate, small “comma-shaped” islands; D) Trichoblastoma will have rounded basaloid islands in the dermis that may focally connect as opposed to the more predominantly branching stranded pattern of basaloid follicular hamartoma; E) The classic intensely eosinophilic basement membrane material normally found in spiradenoma is lacking in this biopsy as is the “blue balls in the dermis” architecture

Reference: Basaloid follicular hamartoma

Comment Here
Board review style question #2
    Basaloid follicular hamartoma has been reported to occur in both sporadic and syndromic forms. With which of the following syndromes is basaloid follicular hamartoma associated?

  1. Birt-Hogg Dubé syndrome
  2. Gorlin Syndrome
  3. Lynch Syndrome
  4. Rothmund Thomson
  5. Tuberous sclerosus
Board review style answer #2
B. Gorlin syndrome / Basal cell nevus syndrome

Explanation: A) Birt-Hogg Dubé is associated with fibrofolliculoma, trichodiscomas and acrochordons; C) the Muir-Torre variant of Lynch is associated with sebaceous adenoma, sebaceous adenocarcinoma and squamous cell carcinoma; D) Rothmund Thomson is characterized by poikiloderma and increased risk of basal cell carcinoma and squamous cell carcinoma among other findings; E) Tuberous sclerosis is associated with angiofibromas, subungual fibromas, connective tissue hamartomas (Shagreen patch) and hypomelanotic macules

Reference: Basaloid follicular hamartoma

Comment Here

Becker's nevus
Definition / general
  • Sharply demarcated pigmented macule, often with localized hypertrichosis (eMedicine)
  • Dermoscopy: network, focal hypopigmentation, skin furrow hypopigmentation, hair follicles, perifollicular hypopigmentation and vessels (Dermatology 2006;212:354)
Terminology
  • Also called pigmented hairy epidermal nevus, Becker's melanosis
  • Becker's nevus syndrome: Becker's nevus with congenital abnormalities (J Am Acad Dermatol 2004;51:965)
Epidemiology
Sites
  • Usually back, shoulder or chest
Clinical features
  • Sharply demarcated, unilateral, hyperpigmented tan macule on back, shoulder or chest
  • Hypertrichosis in 50%
Clinical images

Images hosted on other servers:
Missing Image

Bilateral Becker's nevus

Case reports
Treatment
Microscopic (histologic) description
  • Increased basal layer pigmentation, mild acanthosis, hyperkeratosis and regular elongation of rete ridges
  • Variable hypertrichosis
  • Areas associated with smooth muscle hamartoma have more pronounced smooth muscle bundles irregularly dispersed within the dermis and unrelated to either hair follicles or vascular channels
  • Does not actually contain nevus cells
Differential diagnosis
  • Congenital smooth muscle hamartoma

Benign (mature) cystic teratoma
Definition / general
  • Congenital tumor (extremely rare in skin) containing tissue components from all 3 germinal layers
Clinical features
  • Very rare with only scattered case reports
  • Lesions present at birth
  • Locations vary including face, back, neck, knee
Case reports
Microscopic (histologic) description
  • Cystic structure with elements from all 3 germinal layers (ectoderm, endoderm and mesoderm)
  • Variable tissue types may be present from the germinal layers:
    • Superficial ectoderm such as epidermis and appendages
    • Neuroectoderm (nervous tissue)
    • Mesoderm such as connective tissue, striated and smooth muscle
    • Endoderm such as respiratory epithelium, thyroid tissue, GI mucosa

Benign lymphangioendothelioma / acquired progressive lymphangioma
Definition / general
Essential features
  • Delicate, thin walled, endothelium lined dilated vascular spaces involving the superficial dermis
  • Intravascular papillary stromal projections resemble papillary endothelial hyperplasia
  • Deeper in dermis, vascular spaces collapse and dissect dermal collagen in angiosarcoma-like pattern
Terminology
  • Also called acquired progressive lymphangioma
ICD coding
  • ICD-10: D18.1 - lymphangioma, any site
Epidemiology
  • Uncommon; around 50 reported cases in English literature
  • Wide age distribution
  • No sex predilection
Sites
  • Trunk / limbs usually; other sites have been described
Pathophysiology
  • Favored to be a benign vascular malformation rather than a true neoplasm
Etiology
  • Some cases possibly related to trauma
Clinical features
Case reports
Treatment
Microscopic (histologic) description
  • Delicate, thin walled, endothelium lined dilated vascular spaces involving the superficial dermis
  • Intravascular papillary stromal projections resemble papillary endothelial hyperplasia
  • Deeper portion of lesions have vascular space collapse and dissect collagen bundles, mimicking patch stage Kaposi sarcoma
  • Preexisting vessels and adnexal structures of the dermis also appear dissected by newly formed vascular channels
  • Smooth muscle often focally present around vascular spaces
  • Endothelial cells may hobnail, may form morula resembling giant cells
  • Crowding of endothelial cells present but no endothelial atypia
  • Vascular spaces lack erythrocytes and hemosiderin deposits
  • No mitotic figures
Microscopic (histologic) images

Contributed by Joel Tjarks, M.D. and Sara C. Shalin, M.D., Ph.D.

6.8x

20x

2x

10x


20x

CD31

D2-40

Positive stains
Negative stains
  • HHV8
  • Most cases are WT1-; however, rare WT1 positivity has been reported
Differential diagnosis
  • Kaposi sarcoma: HHV8+; usually demonstrates slit-like vasculature without marked dilation of vessels and plasma cell infiltrate
  • Atypical vascular lesion: demonstrates more endothelial prominence and atypia; associated with radiation
  • Angiosarcoma: dissects through dermal collagen with significant atypia, atypical intraluminal cells, endothelial stacking (multilayering) and mitoses
  • Superficial lymphangioma (lymphangioma circumscriptum): congenital hamartoma often associated with cytogenetic abnormalities; typically does not dissect dermal collagen but may show dilated superficial dermal vessels
  • Targetoid hemosiderotic hemangioma (hobnail hemangioma): may have similar infiltration pattern to benign lymphangioendothelioma; endothelial cells protruding into vascular lumen (hobnail cells); deep stromal hemosiderin deposition
Board review style question #1
Which of the following immunohistochemical stains is negative in benign lymphangioendothelioma?

  1. CD31
  2. CD34
  3. D2-40
  4. ERG
  5. HHV8
Board review style question #2
Which of the following histologic features argues against the diagnosis of benign lymphangioendothelioma?

  1. Delicate thin walled vessels
  2. Frequent mitotic figures
  3. Hobnail endothelial cells
  4. Infiltration into the deep dermis
  5. Intravascular papillary stromal projections
Board review style answer #2

Blastic plasmacytoid dendritic cell neoplasm

Borst-Jadassohn phenomenon
Table of Contents
Definition / general
Definition / general
  • Previously called intraepidermal epithelioma
  • Not clearly defined
  • Likely heterogeneous group of disorders (Am J Dermatopathol 2011;33:492) including irritated seborrheic keratosis, eccrine poroma and other intraepidermal sweat gland tumors

Bowenoid papulosis

CRTC1:TRIM11 cutaneous tumor
Definition / general
  • Malignant dermal tumor with epithelioid and spindled cells of uncertain origin with partial melanocytic differentiation and CRTC1::TRIM11 translocation
Essential features
  • Histological features include a dermal / subcutaneous circumscribed tumor composed of fascicles and nests of monomorphic epithelioid and spindle cells with prominent nucleoli divided by delicate collagen septa, no / very rare pigment
  • Immunohistochemical features include consistent diffuse SOX10 and MITF staining; S100, MelanA and HMB45 stains are variable
  • Defined by identifying CRTC1::TRIM11 translocation
  • No evidence of EWSR1 rearrangement
  • Appears to have at least some malignant potential, although cases and follow up are limited
Terminology
  • Cutaneous melanocytic tumor with CRTC1::TRIM11 fusion
  • Not recommended: cutaneous melanocytoma with CRTC1::TRIM11 fusion
ICD coding
  • ICD-O: 9044/3 - CRTC1:TRIM11 cutaneous tumor
  • ICD-11: 2C35 - cutaneous sarcoma
Epidemiology
Sites
Pathophysiology
  • Exact mechanism of how the CRTC1::TRIM11 fusion transcript causes neoplasia is not known; hypotheses include
    • CRTC1::TRIM11 fusion deletes a protein - protein interaction site on TRIM11 that is involved in directing misfolded proteins towards proteasomes for degradation (Cureus 2022;14:e33179)
    • CRTC1::TRIM11 fusion retains an interaction site with CREB1, which in turn may drive expression of MITF; MITF acts like an oncogene and can drive MITF synthesis (Am J Surg Pathol 2022;46:1457)
Etiology
  • Unknown
Clinical features
Diagnosis
  • Biopsy
  • Defined by molecular findings
Radiology description
  • Lobulated lesion on imaging
  • May be positron emission tomography (PET) avid
Radiology images

Contributed by Aaron Lloyd Hendler, M.D.
PET avid

PET avid

Prognostic factors
  • Limited case reports and follow up available but appears more indolent than melanoma and clear cell sarcoma at the time of writing
  • 4 of 48 published cases reported have shown regional or distant metastasis (Am J Surg Pathol 2022;46:1457, Histopathology 2023;82:368, Am J Surg Pathol 2019;43:861, J Cutan Pathol 2024;51:181)
  • A recent additional case of CRTC1:TRIM11 cutaneous tumor (CTCT) with regional lymph node metastasis was presented at the American Society of Dermatopathology in 2023 (Tseng, et al.: CRTC1::TRIM11 Cutaneous Tumor With Ulceration and Sentinel Lymph Node Metastasis, oral presentation at American Society of Dermatopathology, Oct 2 - 8, 2023)
Case reports
Treatment
  • Other than complete surgical excision, optimal treatment is unknown
Clinical images

Contributed by Alexandra Easson, M.D.
Ulcerated toe CTCT

Ulcerated toe CTCT



Images hosted on other servers:

Arm CTCT

Finger CTCT

Gross description
  • Circumscribed tumors with pale, firm cut surface without pigmentation
  • Mean size: 1.4 cm (0.4 - 5.1 cm) (Histopathology 2023;82:368)
Gross images

Contributed by Zaid Saeed Kamil, M.B.Ch.B. and Calvin Tseng, M.D.
Amputation specimen

Amputation specimen

Microscopic (histologic) description
  • Morphology is uniform and resembles clear cell sarcoma
  • Circumscribed, unencapsulated dermal nodule, no / very rare pigment, with only rare cases showing epidermal involvement (J Cutan Pathol 2022;49:1025)
  • Some cases show extension into subcutis (Am J Surg Pathol 2019;43:861)
  • Characteristic pattern of nests and intersecting (occasionally herringbone to vaguely palisaded) fascicles of uniform epithelioid to spindled cells with intervening delicate collagen bands (Am J Surg Pathol 2022;46:1457)
  • Abundant pale eosinophilic cytoplasm, monomorphic nuclei with vesicular chromatin and prominent nucleoli
  • Mitotic figures present (Am J Surg Pathol 2022;46:1457)
  • Necrosis reported but uncommon (Am J Surg Pathol 2022;46:1457)
  • Lymphovascular and perineural invasion uncommon (Am J Surg Pathol 2022;46:1457)
  • Pigment and multinucleate giant cells may very rarely be seen
  • A recent case of CTCT with ulceration was reported (Tseng, et al.: CRTC1::TRIM11 Cutaneous Tumor With Ulceration and Sentinel Lymph Node Metastasis, oral presentation at American Society of Dermatopathology, Oct 2 - 8, 2023)
Microscopic (histologic) images

Contributed by Zaid Saeed Kamil, M.B.Ch.B., Stephen M. Smith, M.D. and Calvin Tseng, M.D.
Dermal lesion

Dermal lesion

Fascicular growth pattern

Fascicular growth pattern

Monomorphic cells

Monomorphic cells

Vascular invasion

Vascular invasion

SOX10 diffuse positive

SOX10 diffuse positive


S100 positive

S100 positive

MITF diffuse positive

MITF diffuse positive

MelanA negative

MelanA negative

HMB45 negative

HMB45 negative

PRAME negative

PRAME negative


Pan-TRK positive

Pan-TRK positive

Lymph node metastasis

Lymph node metastasis

Lymph node metastasis SOX10

Lymph node metastasis SOX10

Lymph node weak S100

Lymph node weak S100

Negative MelanA and HMB45

Negative MelanA and HMB45

Positive stains
Molecular / cytogenetics description
Molecular / cytogenetics images

Contributed by Ian King, Ph.D.
RNA <i>CRTC1::TRIM11</i> fusion

RNA CRTC1::TRIM11 fusion



Images hosted on other servers:
CRTC1::TRIM11 FISH

CRTC1::TRIM11 FISH

CRTC1::TRIM11 fusion

CRTC1::TRIM11 fusion

Videos

CRTC::TRIM11 cutaneous tumor

Sample pathology report
  • Skin, amputation, right fifth toe:
    • CRTC1::TRIM11 cutaneous tumor (see comment)
    • Comment: The neoplasm is composed of intersecting fascicles and nests of relatively uniform epithelioid and spindled cells with intervening bands of fibrocollagenous stroma. The neoplastic cells have abundant amounts of amphophilic to eosinophilic cytoplasm and moderately pleomorphic nuclei with prominent nucleoli. Mitotic figures including deep mitoses are identified. There is no evidence of necrosis, lymphovascular invasion or perineural invasion. The neoplastic cells are diffusely positive for SOX10, patchy positive for S100 and negative for MelanA and HMB45. Molecular analysis demonstrated the presence of CRTC1::TRIM11 fusion. Overall, the features are compatible with the recently described CRTC1::TRIM11 cutaneous tumor. Based on the limited reported cases and limited follow up data, the clinical course was indolent in most of the reported cases. However, 4 of the reported cases presented with regional nodal metastasis or distant metastasis as of January 2024. The available data at the time of this report suggest that these tumors have at least some malignant potential.
Differential diagnosis
  • Clear cell sarcoma:
    • Predominantly affects adults ages 30 - 40s
    • Often deeper location (associated with tendons or aponeuroses)
    • Infiltrative growth
    • May have clear cells and melanin pigment
    • Wreath-like multinucleated tumor giant cells
    • More consistently expresses S100, MelanA and HMB45
    • Has ESWR1 translocations, most commonly EWSR1::ATF1 or EWSR1::CREB1
    • Lacks CRTC1::TRIM11 fusion
    • More aggressive behavior
  • Melanoma:
    • In situ component (may not be present for previously excised melanoma, metastatic melanoma or primary dermal melanoma)
    • Infiltrative growth
    • Pleomorphic cytology
    • Pigment production
    • More consistently expresses S100, MelanA and HMB45 (other than in desmoplastic melanoma)
    • PRAME positive
    • Has melanoma driver mutations
    • Lacks CRTC1::TRIM11 fusion
    • More aggressive behavior
  • Spitz tumor:
    • Junctional component of vertically oriented nests and Kamino bodies may be present
    • More consistently expresses S100, MelanA and HMB45
    • Molecular analysis shows HRAS mutation or kinase fusions
    • Lacks CRTC::TRIM11 fusion
  • Cellular blue nevus:
    • Component of dendritic blue nevus is often present
    • Pigmented melanophages present
    • Necrosis is absent
    • Mitotic activity is usually low
    • Consistently positive for MelanA and HMB45
    • Molecular shows GNAQ or GNA11 mutations
    • Lacks CRTC1::TRIM11 fusion
  • Myoepithelial tumors:
    • Myxoid stroma may be present
    • EMA positive
    • Keratin, p63, SMA and calponin may be positive
    • MITF negative
    • EWSR1 rearrangements in syncytial variant
    • Lacks CRTC1::TRIM11 fusion
  • Epithelioid schwannoma:
  • Epithelioid MPNST:
    • Increased pleomorphism
    • MITF negative
    • Subset shows loss of INI1
    • Lacks CRTC1::TRIM11 fusion
  • PEComa:
  • MITF pathway activated melanocytic tumors:
    • Infiltrative growth
    • Prominent clear cytoplasm
    • May have areas of higher grade morphology
    • Can have mitotic activity and perineural invasion
    • Usually expresses either MelanA or HMB45
    • Has ACTIN::MITF or MITF::CREM fusions
    • Lacks CRTC1::TRIM11 fusion
Board review style question #1

A superficial dermal tumor with the above histology is identified. IHC shows diffuse SOX10 positivity, diffuse MITF positivity, partial S100 positivity, partial MelanA positivity and partial HMB45 positivity. PRAME IHC is negative. Molecular investigations identify a CRTC1::TRIM11 fusion. Is it important to differentiate this entity from melanoma and clear cell sarcoma and why?

  1. No, as this entity currently appears to behave similarly to melanoma and clear cell sarcoma
  2. No, as this entity is currently thought to be a subtype of melanoma
  3. Yes, as this entity currently appears to behave less aggressively than melanoma and clear cell sarcoma
  4. Yes, as this entity currently has specific targeted treatments available
Board review style answer #1
C. Yes, as this entity currently appears to behave less aggressively than melanoma and clear cell sarcoma. Based on follow up data available as of October 2023, CRTC1:TRIM11 cutaneous tumor (CTCT) appears to be a low grade malignancy that behaves less aggressively than typical melanomas and clear cell sarcomas. Answer B is incorrect because CTCT has not had melanoma driver mutations identified as of October 2023. Answer A is incorrect because CTCT appears to behave less aggressively than melanoma and clear cell sarcoma based on follow up data available as of October 2023. Answer D is incorrect because no targeted treatment has been identified for CTCT as of October 2023.

Comment Here

Reference: CRTC1:TRIM11 cutaneous tumor
Board review style question #2
Of the following features, which is typical of CRTC1:TRIM11 cutaneous tumor (CTCT)?

  1. Absence of mitotic activity
  2. Presence of an epidermal component
  3. Presence of CRTC1::TRIM11 fusion
  4. Presence of infiltrative growth
Board review style answer #2
C. Presence of CRTC1::TRIM11 fusion. CRTC1:TRIM11 cutaneous tumor (CTCT) is defined by the CRTC1:TRIM11 fusion, which has not yet been identified in other entities. Answer B is incorrect because the majority of CTCT cases do not show an epidermal component. Answer D is incorrect because the majority of CTCT are well circumscribed. Answer A is incorrect because CTCT show mitotic activity.

Comment Here

Reference: CRTC1:TRIM11 cutaneous tumor

Capillary malformations (pending)
[Pending]

Clear cell acanthoma
Definition / general
  • Benign epidermal tumor, typically of the leg, with acanthosis and accumulation of glycogen in keratinocytes leading to pale staining cytoplasm
Essential features
  • Benign intraepithelial tumor composed of pale staining, glycogen rich keratinocytes
  • Pink to tan papule or plaque on the distal lower extremity of older adults
  • Histologic features include bland, pale keratinocytes with abrupt transition to normal epidermis, often associated with psoriasiform hyperplasia with scattered neutrophils
Terminology
ICD coding
  • ICD-10
    • D23 - other benign neoplasms of skin
    • D23.70 - other benign neoplasm of skin of unspecified lower limb, including hip
    • D23.9 - other benign neoplasm of skin, unspecified
  • ICD-11
    • 2F21 - benign keratinocytic acanthomas
    • 2F21.Y - other specified benign keratinocytic acanthomas
Epidemiology
Sites
Pathophysiology
  • Possible upregulation of KGF (keratinocyte growth factor)
  • Defect in phosphorylase enzyme leads to intracellular glycogen accumulation
  • Reference: Exp Dermatol 2006;15:762
Etiology
  • Unknown
  • Hypotheses
    • Inflammatory epithelial hyperplasia (reactive)
      • Produces similar cytokeratin as in psoriasis (Dermatopathol 2020;7:26)
      • Associated with other inflammatory dermatoses, such as stasis dermatitis, bacterial and viral dermatoses, atopic dermatitis and insect bite reactions (Dermatopathol 2020;7:26)
    • Variation of seborrheic keratosis
Clinical features
  • Well demarcated, pink to brown papule or plaque
  • Usually 0.5 - 2.0 cm in diameter
  • Peripheral rim of scale and central erythema with puncta that bleed easily upon trauma
    • Associated with crust from weeping
  • Polypoid, pigmented and giant variants (up to 6 cm) have been described
  • Dermoscopic findings
    • Dotted blood vessels lined up in strings with white surrounding halo
    • Vascular lines may be coiled (glomerular) or more rarely in a hairpin-like structure
  • References: J Am Acad Dermatol 2015;72:S47, Dermatopathol 2020;7:26
Diagnosis
  • Skin biopsy or excision
Case reports
Treatment
  • Preferred management is with complete removal
  • Location, size and number of lesions should be considered
  • Methods
    • Shave removal or curettage followed by electrofulguration
    • Electrofulguration alone
    • Surgical excision
    • Cryotherapy or carbon dioxide laser (when there are multiple lesions)
  • Reference: StatPearls: Clear Cell Acanthoma [Accessed 21 July 2023]
Clinical images

Images hosted on other servers:
Brown papule

Brown papule

Thin, pink papule

Thin, pink papule

Red papule

Red papule

Gross description
  • Well demarcated, pink to brown papule or small plaque
Microscopic (histologic) description
  • Bland, intraepithelial tumor of clear, glycogen rich keratinocytes
  • Abrupt transition to normal epidermis (Dermatopathology (Basel) 2020;7:26)
  • Often in a pattern of psoriasiform hyperplasia
  • Parakeratosis
  • Typically lacks the thinning of the suprapapillary plate seen in psoriasis
  • With or without colonization with melanocytes
  • Often the vessels within the dermal papillae are dilated, tortuous and run vertically up the papillae
  • Often spares hair follicles / adnexal structures
  • Some cases have hyperplasia of underlying sweat ducts
  • Reference: Dermatopathology (Basel) 2020;7:26
Microscopic (histologic) images

Contributed by Carli Cox, M.D.
Pale cytoplasm

Pale cytoplasm

Abrupt clear cell transition

Abrupt clear cell transition

Overlying crust

Overlying crust

Psoriasiform hyperplasia

Psoriasiform hyperplasia

Glycogen rich keratinocytes

Glycogen rich keratinocytes

Glycogen rich keratinocytes

PAS

Positive stains
Negative stains
Videos

Clear cell acanthoma

Sample pathology report
  • Skin, right lower leg, biopsy:
    • Clear cell acanthoma
Differential diagnosis
  • Squamous cell carcinoma in situ:
    • Squamous cell carcinoma in situ may have clear cell variant
    • Cells are pleomorphic with increased N:C ratio
    • Has full thickness atypia
  • Psoriasis vulgaris:
    • Elongation of rete ridges, thinning of suprapapillary plates
    • Lymphocytic infiltrate in upper and middle portions of dermis
    • Lacks the sharply demarcated lateral boundaries and glycogenation
    • Lacks string-like arrangement of vessels on dermoscopy
  • Seborrheic keratosis:
    • Hyperkeratotic with horn pseudocysts
Board review style question #1

A 62 year old man presents with a 1.2 cm sharply demarcated, pink plaque on his left lower leg. Which stain is most likely to be positive?

  1. CEA
  2. GMS
  3. p16
  4. PAS
  5. S100
Board review style answer #1
D. PAS. PAS stains glycogen in the keratinocytes. Answers A, B and C are incorrect because CEA, GMS and p16 are negative in clear cell acanthomas. p16 is often used as a surrogate for HPV, which is not identified in clear cell acanthomas. Answer E is incorrect because clear cell acanthomas are not of neural or melanocytic origin.

Comment Here

Reference: Clear cell acanthoma

Clear cell papulosis
Definition / general
  • Rare condition of unknown histogenesis and pathogenesis, predominantly described in Asian children
  • Originally proposed to be related to Toker's clear cells of the nipple (Am J Surg Pathol 1987;11:827)
  • May be a genetic predisposition based on positive family history in some cases
Clinical features
  • Manifests in early childhood, majority before age 2
  • Rare in adults
  • No gender predilection
  • Predominantly described in Asian and Hispanic populations
  • Presents as multiple white hypopigmented macules or papules most commonly on the pubic area and lower abdomen
  • Less commonly on face, chest, anterior trunk, axilla
  • Can be linear, and may develop along the 'milk line'
  • Range from 2 - 10 mm in size
  • Usually 10 - 20 lesions, with range from 5 to more than 100
  • Hypopigmented to depigmented macules or barely palpable papules
Case reports
Treatment
Clinical images

Images hosted on other servers:
Multiple hypopigmented macules

Multiple hypopigmented macules

Gross description
  • Small hypopigmented macules or papules
Microscopic (histologic) description
  • Intraepidermal oval to round cells with ample pale clear cytoplasm, single or in clusters
  • Clear cells predominantly among basal cells but some may be in the suprabasal epidermal layers
  • Larger than adjacent keratinocytes
  • Nucleoli tiny or indistinct
  • Mitotic figures rare and no pleomorphism
  • Epidermis mildy acanthotic with mild hyperkeratosis
  • Decreased to absent pigmentation of basal epidermis (can be highlighted by Fontana-masson stain)
Microscopic (histologic) images

Images hosted on other servers:
Mild acanthosis

Mild acanthosis

H&E, CK7+

H&E, CK7+

Positive stains
Negative stains
Differential diagnosis
  • Paget disease: histologic mimic - distinction is predominantly based on clinical findings (i.e. age of patient, location, number of lesions and clinical appearance)
  • Pagetoid dyskeratosis: an incidental histologic finding - cells with a pyknotic nucleus, a clear halo, and a rim of pale cytoplasm; clear cells at higher levels in epidermis, negative for PAS / mucicarmine,

Collagenous fibroma
Definition / general
  • Also called desmoplastic fibroblastoma
  • Rare paucicellular tumor consisting of dense collagen fibers with scattered stellate and spindled fibroblasts involving the subcutis or skeletal muscles
  • Benign clinical behavior with no reported tumor recurrence
Essential features
  • Most frequently found in the subcutis, with up to 25% found in the skeletal muscles (Hum Pathol 1998;29:676)
  • Essential to distinguish from other more aggressive fibroblastic and myofibroblastic neoplasms
  • Treatment is simple excision
Terminology
  • Desmoplastic fibroblastoma
ICD coding
  • ICD-O: 8810/0 - desmoplastic fibroblastoma
  • ICD-11: EE6Y & XH2ZF3 - other specified fibromatous disorders of skin and soft tissue & desmoplastic fibroblastoma
Epidemiology
Sites
Pathophysiology
Etiology
  • Unknown at this time
Clinical features
Diagnosis
  • Histopathology is the gold standard for definitive diagnosis
Radiology description
Radiology images

Images hosted on other servers:
Ultrasound showing lobulated, well defined mass

Ultrasound showing lobulated, well defined mass

Ultrasound showing diffuse vascularity

Ultrasound showing diffuse vascularity

MRI showing mass with well defined margin

MRI showing mass with well defined margin

Prognostic factors
Case reports
Treatment
  • Surgical excision
Clinical images

Images hosted on other servers:
Bilateral lesion in hard palate

Bilateral lesion in hard palate

Collagenous fibroma of face

Collagenous fibroma of face

Gross description
  • Firm, well circumscribed, lobulated mass (Hum Pathol 1998;29:676)
  • Uniform white-gray cut surface
  • No necrosis or hemorrhage
Gross images

Images hosted on other servers:
Macroscopic appearance and cut surface of tumor

Macroscopic appearance and cut surface of tumor

Macroscopic appearance of tumor

Macroscopic appearance of tumor

Frozen section description
  • Intraoperative frozen section analysis is usually not performed
Microscopic (histologic) description
  • Well circumscribed but may infiltrate fat or less commonly skeletal muscles (Hum Pathol 1998;29:676)
  • Hypocellular with fibromyxoid or collagenous stroma
  • Bland spindled to stellate shaped fibroblasts
  • Minimal vasculature
  • Minimal to absent mitotic activity
Microscopic (histologic) images

Contributed by Grace Y. Wang, M.D.
Hypocellular lesion

Hypocellular lesion

Collagenous stroma

Collagenous stroma

Bland appearing fibroblast

Bland appearing fibroblasts

Spindled fibroblasts

Spindled fibroblasts

Fascicular arrangement

Fascicular arrangement

Absent mitosis

Absent mitosis

Cytology description
  • Cytology is usually not performed
Positive stains
  • Immunohistochemical (IHC) stains are usually not needed for diagnosis
  • Most IHC stains are nonspecific
  • Diffuse and strong nuclear reactivity with FOSL1 (In Vivo 2021;35:69)
  • Variable SMA positive
  • Rare focal keratin expression
Electron microscopy description
  • Electron microscopy is not usually performed
  • Tumor cells show features of fibroblasts and myofibroblasts
Molecular / cytogenetics description
Molecular / cytogenetics images

Images hosted on other servers:
GTG banded karyotype showing 2;11 translocation

GTG banded karyotype showing 2;11 translocation

Videos

Desmoplastic fibroblastoma (collagenous fibroma) versus sclerotic / plywood fibroma

Desmoplastic fibroblastoma (collagenous fibroma)

Sample pathology report
  • Skin, chest, biopsy:
    • Collagenous fibroma (desmoplastic fibroblastoma) (see comment)
    • Comment: Sections demonstrate a well circumscribed hypocellular proliferation of stellate fibroblasts in a collagenous stroma, consistent with a collagenous fibroma (desmoplastic fibroblastoma).
Differential diagnosis
  • Desmoid type fibromatosis:
    • Predominantly affects young adults (mean age: 41)
    • Deep seated soft tissue tumors, subdivided into extra-abdominal, abdominal and intra-abdominal
    • May be associated with Gardner syndrome
    • Locally aggressive, nonmetastasizing
    • Long fascicles of bland fibroblasts and myofibroblasts
    • Usually more cellular than collagenous fibroma, except for the hypocellular variant
    • Conspicuous thin walled blood vessels with perivascular edema and lymphoid aggregates
    • Beta catenin nuclear staining
    • Mutations CTNNB1 (sporadic) and APC (familial)
  • Low grade fibromyxoid sarcoma:
    • Predominantly affects young adults (mean age: 35 - 45)
    • Deep seated soft tissue tumors of proximal extremities and trunk
    • Protracted clinical course with high risk of local recurrence and late metastasis
    • Short fascicles of bland spindle fibroblasts in alternating myxoid and collagenous stroma
    • Curvilinear blood vessels (Int J Clin Exp Pathol 2018;11:5860)
    • MUC4 positive
    • FUS::CREB3L2, FUS::CREB3L1 and rarely EWSR1::CREB3L1 fusions
  • Fibroma of tendon sheath:
    • Predominantly affects young to middle aged adults (median age: 31)
    • Arises from synovium of tendon sheath in small distal joints
    • 5 - 10% risk of recurrence
    • Well circumscribed paucicellular tumor with bland fibroblasts, collagenous stroma and slit-like vessels
    • SMA positive; may be focal
    • Desmin and nuclear beta catenin negative
    • USP6 gene rearrangement by FISH
Board review style question #1
What molecular findings are associated with collagenous fibroma?

  1. t(2;11)(q31;q12)
  2. t(7;16)(q32-34;p11) FUS::CREB3L2 fusion
  3. t(17;22)
  4. USP6 gene rearrangement
Board review style answer #1
A. t(2;11)(q31;q12). Collagenous fibromas (desmoplastic fibroblastomas) have been reported to have translocations involving 11q12, including t(2;11)(q31;q12) and t(11;17)(q12;p11.2). Answer D is incorrect because USP6 gene rearrangement is associated with fibroma of tendon sheath. Answer C is incorrect because t(17;22) is associated with dermatofibrosarcoma protuberans. Answer B is incorrect because t(7;16)(q32-34;p11) FUS::CREB3L2 fusion is associated with low grade fibromyxoid sarcoma.

Comment Here

Reference: Collagenous fibroma
Board review style question #2

A 50 year old woman presents with a solitary, firm nodule in the left upper arm. It is excised and the histology is shown in the image above. What is the most likely diagnosis?

  1. Collagenous fibroma
  2. Desmoid type fibromatosis
  3. Fibroma of tendon sheath
  4. Low grade fibromyxoid sarcoma
Board review style answer #2
A. Collagenous fibroma. Collagenous fibroma is a hypocellular tumor with bland spindled to stellate shaped fibroblasts and a fibromyxoid to collagenous background. Answer C is incorrect because fibroma of tendon sheath shows bland fibroblasts and collagenous stroma with slit-like vessels. Answer B is incorrect because desmoid type fibromatosis shows long fascicles of bland fibroblasts and myofibroblasts, conspicuous thin walled blood vessels with perivascular edema, lymphoid aggregates and more cellularity. Answer D is incorrect because low grade fibromyxoid sarcoma shows short fascicles of bland spindled fibroblasts in alternating myxoid and collagenous stroma as well as curvilinear blood vessels.

Comment Here

Reference: Collagenous fibroma

Composite hemangioendothelioma

Condyloma

Connective tissue nevus
Definition / general
  • Rare connective tissue hamartoma derived from cells of mesodermal origin
  • May be syndromic (Proteus syndrome, tuberous sclerosis, others) or isolated (collagenoma)
Case reports

Cutaneous adnexal NUT carcinoma (pending)
[Pending]

Cutaneous epithelioid angiomatous nodule
Definition / general
  • Benign vascular tumor comprised of solid sheets of epithelioid endothelial cells in the superficial dermis
  • May be mitotically active
  • Strongly positive for vascular markers
  • Benign clinical behavior with no reported adverse outcomes
Essential features
Terminology
  • CEAN
ICD coding
  • ICD-10: L98.8 - other specified disorders of the skin and subcutaneous tissue
  • ICD-11: XH8SM9 - cutaneous epithelioid angiomatoid nodule
Epidemiology
  • Very rare (< 70 cases reported in the English literature) (J Cutan Pathol 2022;49:765)
  • Wide age range (14 - 84 years old)
  • Slight male predominance (M:F = 1.4:1)
Sites
Pathophysiology
Etiology
  • Unknown at this time
Clinical features
  • Most commonly a solitary lesion but may be multiple
  • Red to violaceous vascular nodule; may be painful
Diagnosis
  • Histopathology is the gold standard for definitive diagnosis
Radiology description
  • Dermal based, palpable; imaging studies are usually not necessary
  • In cases involving nasal cavity, a homogenous mass is seen on noncontrast CT
Prognostic factors
Case reports
Treatment
  • Surgical excision
Clinical images

Images hosted on other servers:

Solitary vascular papule

Arose within a port wine stain

Arose within a port wine stain

Gross description
Frozen section description
  • Frozen sections usually not performed
Microscopic (histologic) description
  • Dermal based (Acta Derm Venereol 2017;97:135, Am J Dermatopathol 2008;30:16)
  • Well circumscribed
  • Nodular proliferation of epithelioid cells with vesicular chromatin, prominent nucleoli and abundant eosinophilic to clear cytoplasm
  • Mitotic figures can be present but not numerous (up to 5/10 high power fields) and without atypical forms
  • In exceptional cases, moderate cytologic has been reported (Diagn Pathol 2018;13:50)
  • Vascular channel formation is focal but intracytoplasmic lumina are common
  • Vascular channels are lined by a single layer of constituent cells
  • Mild background chronic lymphoplasmacytic inflammation with scattered eosinophils
  • Hemosiderin deposition may be seen
Microscopic (histologic) images

Contributed by Grace Y. Wang, M.D.
Dermal nodule

Dermal nodule

Mixed inflammation with hemosiderin deposition

Mixed inflammation with hemosiderin deposition

Epithelioid cells

Epithelioid cells

Intracytoplasmic lumina

Intracytoplasmic lumina


Well circumscribed nodule

Well circumscribed nodule

Vascular channels

Vascular channels

CD31

CD31

CD34

CD34

Sample pathology report
  • Skin, chest, biopsy:
    • Cutaneous epithelioid angiomatous nodule (see comment)
    • Comment: The biopsy demonstrates a well circumscribed nodule, comprised of sheets of epithelioid cells in the superficial dermis. The epithelioid cells are relatively monomorphous and have intracytoplasmic vacuoles. Rare vascular channel formation is noted. A mild stromal lymphocytic and eosinophilic inflammatory infiltrate is present at the periphery. Immunohistochemistry shows that the epithelioid cells are positive for CD34 and ERG. The overall histopathologic and immunohistochemical findings are consistent with cutaneous epithelioid angiomatous nodule, a benign vascular neoplasm with a very low risk of local recurrence and no metastatic potential.
Differential diagnosis
Board review style question #1

A 35 year old man presents with a painful cutaneous upper arm lesion (shown above). What is the diagnosis?

  1. Bacillary angiomatosis
  2. Cutaneous epithelioid angiomatous nodule
  3. Epithelioid angiosarcoma
  4. Epithelioid hemangioma
Board review style answer #1
B. Cutaneous epithelioid angiomatous nodule. Pictured is a dermal based proliferation of epithelioid cells with abundant eosinophilic cytoplasm and chronic inflammation. Answer D is incorrect because there are focal areas of single layered vascular channel formations but they are not prominent. Answer A is incorrect because there are no associated neutrophils or bacteria identified. Answer C is incorrect because there are no readily appreciated mitotic figures and marked cytologic atypia.

Comment Here

Reference: Cutaneous epithelioid angiomatous nodule
Board review style question #2
What is the clinical behavior of cutaneous epithelioid angiomatous nodule?

  1. Indolent
  2. Locally aggressive
  3. Potentially metastatic
  4. Recurs frequently
Board review style answer #2
A. Indolent. Cutaneous epithelioid angiomatous nodule is a benign vascular proliferation that has not been shown to metastasize and does not generally recur following excision.

Comment Here

Reference: Cutaneous epithelioid angiomatous nodule

Cutaneous fibroepithelial polyps
Definition / general
  • Benign nonepithelial tumors arising from mesodermal tissue
Essential features
  • Common benign cutaneous tumor
  • On eyelids, flexural areas
  • F = M
  • Associated with trauma, obesity, diabetes mellitus, pregnancy
Terminology
  • Acrochordon, soft fibroma, fibroma molle, skin tag
ICD coding
  • ICD-10: L91.8 - other hypertrophic disorders of the skin
  • ICD-11: EK71 - skin tags or polyps
  • SNOMED:
    • 201091002 - skin tag (disorder)
    • 31069005 - fibroepithelial polyp (morphologic abnormality)
Epidemiology
Sites
  • Eyelids
  • Flexural areas: axillae, groin
  • Lateral neck
  • Inframammary
Pathophysiology
Etiology
Clinical features
Diagnosis
  • Characteristic clinical features and histopathology
Prognostic factors
  • Benign
Case reports
Treatment
Clinical images

Contributed by Arvind Ranchhod, M.B.Ch.B.
Fibroepithelial polyps Fibroepithelial polyps

Multiple skin tags



Images hosted on other servers:
Penile fibroepithelial polyp

Penile fibroepithelial polyp

Fibroepithelial polyp<br>at posterior wall of<br>external auditory canal

Fibroepithelial
polyp at posterior
wall of external
auditory canal

Gross description
  • Sessile or pedunculated polyp
  • 2 - 5 mm to several centimeters (Australas J Dermatol 2019;60:70)
  • Surface smooth or filiform
  • Smaller ones may be pigmented
  • Larger ones are skin colored
Gross images

Contributed by Alexander Nirenberg, M.B.B.S.
Fibroepithelial polyps

Clinical skin tag

Microscopic (histologic) description
  • Epidermis may be hyperplastic and papillomatous and may have keratotic cysts and pigment in basal epidermal keratinocytes
  • Loose fibrocollagenous stroma, abundant vessels
  • Usually no adnexa
  • Variable adipose tissue in larger ones - lipofibroma
  • Traumatic changes: lichen simplex chronicus, epidermal necrosis, ulceration, pagetoid dyskeratosis, lichen sclerosus-like change (Am J Dermatopathol 2006;28:478, Am J Dermatopathol 2019;41:e64)
  • Pseudosarcomatous change: pleomorphic stellate stromal cells, multinucleated stromal cells, myxoid to collagenous stroma (Ann Diagn Pathol 2008;12:440)
  • On IHC, the pleomorphic cells stain diffusely for vimentin; there is variable staining for CD34 and factor 13a and the cells do not stain for SMA or desmin (Ann Diagn Pathol 2008;12:440)
  • Some of the lesions with pseudosarcomatous change overlap with pleomorphic fibroma
  • A subset of cellular pseudosarcomatous fibroepithelial polyps occurs in the female genital tract including the vulva (Am J Surg Pathol 2000;24:231)
Microscopic (histologic) images

Contributed by Alexander Nirenberg, M.B.B.S.
Classic features

Classic features

Prominent vascular stroma

Prominent vascular stroma

Stromal edema Stromal edema

Stromal edema


Epidermal hyperplasia

Epidermal hyperplasia

Adipose tissue

Adipose tissue

Trauma / inflammation Trauma / inflammation

Trauma / inflammation

Sample pathology report
  • Left neck, shave biopsy:
    • Fibroepithelial polyp / acrochordon (see comment)
    • Comment: A polyp covered by mildly acanthotic epidermis with mild hyperkeratosis. There is a mildly cellular fibrovascular core with scattered dilated small thin walled vessels and a sparse lymphocytic infiltrate. Dysplasia or malignancy is not seen.
Differential diagnosis
Board review style question #1

A 62 year old woman presents with multiple lesions on the neck (shown above) with pathological diagnosis of fibroepithelial polyps, NOS. Which clinical association should you consider?

  1. Anorexia
  2. Birt-Hogg-Dubé syndrome
  3. Diabetes mellitus
  4. Dysplastic nevus syndrome
  5. Neurofibromatosis
Board review style answer #1
C. Diabetes mellitus. Skin tags may be associated with diabetes mellitus / metabolic syndrome as well as obesity. The other listed conditions have not been shown to have increased incidence of fibroepithelial polyps but may have other cutaneous polypoid lesions. Birt-Hogg-Dubé syndrome was initially reported as having increased incidence of skin tags, however, the lesions are polypoid trichodiscomas and fibrofolliculomas.

Comment Here

Reference: Cutaneous fibroepithelial polyps
Board review style question #2

A clinician excises a skin tag from the groin of a 73 year old woman (microscopic image shown above). The best diagnosis is

  1. Cutaneous myxoma
  2. Neurofibroma
  3. Pseudosarcomatous fibroepithelial polyp
  4. Pyogenic granuloma
  5. Traumatized fibroepithelial polyp
Board review style answer #2
E. Traumatized fibroepithelial polyp. This is a fibroepithelial polyp with features of trauma, including an area of epidermal necrosis, stromal edema and stromal inflammatory cells. The stroma is loose and has low cellularity of mesenchymal cells, unlike a neurofibroma. The stroma is edematous rather than myxoid. It lacks the pleomorphic stellate and multinucleated stromal cells of a pseudosarcomatous fibroepithelial polyp. The lesion has dilated vessels, however, it lacks the lobular proliferation of capillaries that characterizes a pyogenic granuloma.

Comment Here

Reference: Cutaneous fibroepithelial polyps

Cutaneous lymphadenoma
Definition / general
  • Rare neoplasm characterized by marked mononuclear infiltrate that appears to be integral component of tumor
  • May be related to eccrine spiradenoma
Case reports
Clinical images

Contributed by Mark R. Wick, M.D.


Images hosted on other servers:

Skin-colored, elastic-hard, well-demarcated nodule

Gross description
  • Firm, well-demarcated nodule
Microscopic (histologic) description
  • Multiple rounded lobules of basaloid cells with some peripheral palisading, focal keratinization, occasional duct formation and mixed with small lymphocytes
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D.

Various images



Images hosted on other servers:

Cords, lobules, and interconnected strands

Immunohistochemical
stainings for
cutaneous
lymphadenoma

Formalin fixed
paraffin embedded
tissue samples

Positive stains
Negative stains
Differential diagnosis

Cutaneous lymphoid hyperplasia
Definition / general
  • Heterogeneous group of disorders with benign T or B cell lymphocytic infiltrates in the skin that mimic cutaneous lymphomas both clinically and histologically (J Am Acad Dermatol 1998;38:877)
Essential features
  • Typically presents as a solitary nodule in the head and neck region
  • Spontaneous regression is common; excellent prognosis
  • Idiopathic; however, some cases are associated with specific antigenic stimuli or triggering factors (e.g., drugs, tattoos, vaccinations, arthropod bites and infections)
  • Classically characterized by a dermal lymphoid infiltrate composed predominantly of reactive polyclonal lymphocytes with a variable admixture of other inflammatory cells
  • Although typically absent, clonal T or B cell populations can occasionally be seen in cutaneous lymphoid hyperplasia
Terminology
  • Pseudolymphoma, lymphocytoma cutis, lymphadenosis benigna cutis
ICD coding
  • ICD-10: L98.8 - other specified disorders of the skin and subcutaneous tissue
  • ICD-11: EE91 - lymphocytoma cutis
Epidemiology
Sites
  • Can involve any area of the skin
  • Most commonly arises in the head and neck regions (particularly the face), followed by the extremities and trunk
Pathophysiology
  • Largely unknown but thought to be a reactive process to certain antigenic stimuli
Etiology
Clinical features
  • Solitary nodule or localized cluster of red to violaceous papulonodules
Diagnosis
  • Requires thorough clinical and histopathologic evaluation and acquisition of appropriate clinical history to identify possible triggering factors
  • In the absence of an identifiable triggering factor, it may pose a diagnostic challenge to pathologists and may potentially be misdiagnosed as a neoplastic process, especially if clonality is found in the cutaneous infiltrate
  • Historically divided into 3 categories based on the predominant cell type in the infiltrate (J Am Acad Dermatol 1998;38:877, Hum Pathol 2003;34:617):
    1. B cell predominant type
    2. T cell predominant type
    3. Mixed B cell / T cell type
  • Newly proposed classification divides cutaneous lymphoid hyperplasia into 4 major groups, based on both histologic and clinical features (J Cutan Pathol 2020;47:76):
    1. Nodular cutaneous lymphoid hyperplasia
    2. Cutaneous lymphoid hyperplasia mimicking mycosis fungoides or other cutaneous T cell lymphomas
    3. Other cutaneous lymphoid hyperplasia with distinct clinical features (e.g., acral pseudolymphomatous angiokeratoma, T cell rich angiomatoid polypoid pseudolymphoma, primary cutaneous angioplasmocellular hyperplasia, lymphoplasmacytic plaque, cutaneous plasmacytosis)
    4. Intravascular cutaneous lymphoid hyperplasia
  • Nodular type is the most common form and encompasses nodular B cell cutaneous lymphoid hyperplasia, Borrelia associated nodular B cell cutaneous lymphoid hyperplasia (also known as lymphocytoma cutis or lymphadenosis benigna cutis), nodular T cell and mixed cutaneous lymphoid hyperplasia and nodular CD30+ cutaneous lymphoid hyperplasia (J Cutan Pathol 2020;47:76)
Prognostic factors
  • Benign clinical course
  • Spontaneous regression is common following excision or topical / intralesional corticosteroids
  • May rarely recur
  • Almost never progresses to an overt lymphoma (although rare anecdotal examples of such progression have been described) (Am J Pathol 1989;135:13, Hum Pathol 2003;34:617)
Case reports
Treatment
Clinical images

Images hosted on other servers:

Erythematous papule

Erythematous nodule

Microscopic (histologic) description
  • Dermal lymphoid infiltrate, forming nodules or diffuse sheets (particularly in nodular cutaneous lymphoid hyperplasia), without significant epidermal involvement
  • Grenz zone, between the dermal infiltrate and the overlying epidermis, is frequently found
  • Perivascular and periadnexal distribution patterns may also be seen
  • Although typically intradermal, the infiltrate may also occasionally extend into the superficial subcutis
  • Prominent lymphocytic exocytosis may occasionally be seen mimicking other cutaneous T cell lymphomas, such as mycosis fungoides
  • Infiltrate predominantly composed of small to medium sized lymphocytes, often indistinguishable from those seen in primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder, with variable admixture of other inflammatory cells (e.g., histiocytes, eosinophils, plasma cells)
  • Well formed reactive germinal centers may or may not be seen
    • If present, reactive germinal centers often show tingible body macrophages and preservation of polarity and mantle zone
Microscopic (histologic) images

Contributed by Woo Cheal Cho, M.D.
Dermal nodular lymphoid infiltrate

Dermal nodular lymphoid infiltrate

Nodules in the superficial and deep dermis

Nodules in the superficial and deep dermis

Lymphocytes, histiocytes and scattered eosinophils

Lymphocytes, histiocytes and scattered eosinophils

Cutaneous lymphoid hyperplasia simulating mycosis fungoides Cutaneous lymphoid hyperplasia simulating mycosis fungoides

Cutaneous lymphoid hyperplasia simulating mycosis fungoides


CD3

CD3

CD4

CD4

CD8

CD8

CD20

CD20

CD30

CD30

PD-1

PD-1


CD3

CD3

CD4

CD4

CD7

CD7

CD8

CD8

Kappa

Kappa

Lambda

Lambda

Positive stains
  • Immunophenotype is variable depending on the predominant component of the infiltrate (Hum Pathol 2003;34:617)
  • In general, the infiltrate is composed of mixed T cells (CD3+) and B cells (CD20+)
  • Although variable depending on the subtypes of cutaneous lymphoid hyperplasia, T cell component may show an increased CD4:CD8 ratio
    • Caution should be taken when evaluating CD4+ cells in a cutaneous lymphoid infiltrate in order to avoid potential overestimation of CD4:CD8 ratio, as Langerhans cells and dermal histiocytes can also express CD4 (typically in weaker intensity than that of CD4+ T cells, though)
  • B cell component expresses CD20, CD79a and BCL6
    • BCL2 is expressed in the mantle zone of B cell pseudolymphomas with well formed reactive germinal centers
    • Ki67 labeling may be increased in reactive germinal centers, albeit nonspecific
  • Subset of T cells may express follicular helper T cell markers (PD-1, BCL6, CXCL13)
  • Plasma cells are usually polytypic (mixed kappa and lambda positive plasma cells)
Negative stains
  • T cell component may show partial loss of CD7 and less commonly, CD5 (loss of pan T cell markers, particularly isolated loss of CD7, however, is not pathognomonic for neoplastic processes)
  • B cell component lacks BCL2, CD5 or cyclin D1 expression; the mantle zone cells will express BCL2 in cases with well formed germinal centers
  • B cell component lacks CD43 coexpression
  • CD30, EBER
    • CD30 can be positive in T cell variants, particularly with persistent arthropod bite reactions, molluscum contagiosum, HSV infection, etc.
    • Scattered cells versus sheets or clusters and weak staining may favor CLH over neoplasia
Molecular / cytogenetics description
Sample pathology report
  • Skin, right arm, punch:
    • Nodular dermal lymphohistiocytic infiltrate admixed with scattered eosinophils, consistent with cutaneous nodular hyperplasia (see comment)
    • Comment: Sections show a nodular dermal lymphoid infiltrate without significant epidermal involvement. There is a grenz zone between the dermal infiltrate and the overlying epidermis. The infiltrate is forming nodules in the superficial and deep dermis, with focal perivascular and periadnexal distribution patterns and is predominantly composed of lymphocytes and histiocytes. Scattered eosinophils are also noted. Definitive germinal center formation is not identified. Immunohistochemical studies highlight the infiltrate to be composed of mixed CD3+ T cells and CD20+ B cells. The CD3+ T cells show a CD4:CD8 ratio of approximately 4:1. Anti-CD4 also highlights numerous background histiocytes. Anti-CD30 highlights scattered cells admixed within the infiltrate. Anti-PD-1 highlights areas with slightly increased PD-1+ T cells. Kappa and lambda by in situ hybridization fail to reveal increased monotypic plasma cells. Special stains (Fite, Gram and PAS) are negative for definitive fungal and bacterial (including mycobacterial) organisms. If available, correlation with the results of microbiology cultures is necessary. The histologic differential diagnoses include primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder and cutaneous lymphoid hyperplasia. Additional molecular studies by polymerase chain reaction for T cell receptor gene rearrangements fail to reveal monoclonal pattern of amplification of both TCRB and TCRG. Taken together, these morphologic, immunophenotypic and molecular features are consistent with cutaneous lymphoid hyperplasia in the appropriate clinical context. Clinical pathologic correlation and a close follow up are necessary.
Differential diagnosis
  • Primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder:
    • Distinction from cutaneous lymphoid hyperplasia (particularly nodular T cell and mixed type) is challenging due to frequently overlapping histologic (i.e., dense lymphoid infiltrate composed of small to medium sized lymphocytes involving the dermis or subcutis), immunophenotypic (i.e., CD3+, CD4+, CD8-, CD30- phenotype) and clinical (i.e., solitary plaque or nodule on the head and neck region) features
    • Loss of 1 or more pan T cell markers (CD2, CD5 and CD7) more frequently seen in primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder than in cutaneous lymphoid hyperplasia
    • Clonal T cell receptor gene rearrangements more commonly seen in primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder than in cutaneous lymphoid hyperplasia
    • Presence of monoclonality, in the absence of identifiable triggering factors, favors primary cutaneous + small / medium T cell lymphoproliferative disorder over cutaneous lymphoid hyperplasia
  • Primary cutaneous marginal zone B cell lymphoma:
    • Simulates Borrelia associated nodular B cell cutaneous lymphoid hyperplasia
    • Sheets of plasma cells and Dutcher bodies more frequently seen in primary cutaneous marginal zone B cell lymphoma than in cutaneous lymphoid hyperplasia
    • Neoplastic B cells express BCL2 while lacking immunoreactivity for CD5, CD10, BCL6 and cyclin D1
    • Neoplastic B cells may coexpress CD43
    • Monotypic plasma cells with light chain restriction (either kappa or lambda) more frequently seen in primary cutaneous marginal zone B cell lymphoma than in cutaneous lymphoid hyperplasia
    • Clonal immunoglobulin heavy chain gene rearrangements are more commonly seen in primary cutaneous marginal zone B cell lymphoma than in cutaneous lymphoid hyperplasia
  • Mycosis fungoides:
    • Cutaneous lymphoid hyperplasia may histologically mimic mycosis fungoides, particularly if the infiltrate is T cell predominant and shows prominent lymphocytic exocytosis
    • Neoplastic cells of mycosis fungoides, however, typically exhibit cytologic atypia with hyperchromatic nuclei with irregular convoluted nuclear contours
    • Both mycosis fungoides and cutaneous lymphoid hyperplasia can show an increased CD4:CD8 ratio in the infiltrate
    • Presence of patches / plaques favors mycosis fungoides over cutaneous lymphoid hyperplasia
  • Primary cutaneous follicular center lymphoma:
    • May simulate nodular B cell cutaneous lymphoid hyperplasia
    • Neoplastic follicles lack tingible body macrophages and have attenuated or absent mantle zones
    • Neoplastic B cells express BCL6, CD10 (variable; positive in follicular pattern but generally negative in diffuse pattern) while typically lacking immunoreactivity for BCL2
Board review style question #1

Which of the following features would most likely favor a diagnosis of cutaneous lymphoid hyperplasia over primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder?

  1. Dermal lymphoid infiltrate predominantly composed of small to medium sized pleomorphic T cells
  2. Presence of B cells coexpressing CD43
  3. Presence of T cell receptor beta (TCRB) and gamma (TCRG) gene rearrangements
  4. Recent history of vaccination to the same site with an erythematous nodule
  5. Presence of PD-1+ and CXCL13+ cells in the infiltrate
Board review style answer #1
D. Recent history of vaccination to the same site with an erythematous nodule. Development of an erythematous nodule at the same site where vaccination was recently administered would favor a reactive process, such as cutaneous lymphoid hyperplasia (CLH). In the absence of identifiable triggering factors, however, distinguishing CLH from primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder (CD4+ small / medium T cell lymphoproliferative disorder) is challenging on histologic grounds alone. Typically, the presence of clonality (choice C) detected by polymerase chain reaction would favor a diagnosis of primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder over CLH, although clonality may also be seen in CLH occasionally. Histologic features of primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder (choice A) are similar to those seen in CLH. Scattered to slightly increased follicular helper T cells (choice E) can be seen in both CLH and primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder. The neoplastic B cells coexpressing CD43 (choice B) may be seen in primary cutaneous marginal zone B cell lymphoma, not in CLH or primary cutaneous CD4+ small / medium T cell lymphoproliferative disorder.

Comment Here

Reference: Cutaneous lymphoid hyperplasia
Board review style question #2
Which of the following statements is true regarding cutaneous lymphoid hyperplasia?

  1. Clonal T cell receptor beta (TCRB) and gamma (TCRG) gene rearrangements are never found in cutaneous lymphoid hyperplasia
  2. Cutaneous lymphoid hyperplasia frequently progresses to a lymphoma
  3. Diagnosis of cutaneous lymphoid hyperplasia requires clinical and pathologic correlation
  4. Presence of monotypic plasma cells with kappa light chain restriction always favors a diagnosis of primary cutaneous marginal zone B cell lymphoma over cutaneous lymphoid hyperplasia
  5. Triggering factors or etiologic agents are always identified in cutaneous lymphoid hyperplasia
Board review style answer #2
C. Diagnosis of cutaneous lymphoid hyperplasia requires clinical and pathologic correlation. Accurate diagnosis of cutaneous lymphoid hyperplasia (CLH) requires clinical and pathologic correlation, including acquisition of appropriate clinical histories to determine the presence of a possible triggering factor. Albeit rare, monotypic plasma cells with a light chain restriction can also be seen in CLH (choice D). CLH is often idiopathic and definitive etiologic agents may not be found (choice E). Progression of CLH to a lymphoma is not a frequent finding (choice B). The presence of clonality in the infiltrate in the skin is not unique to malignancy (choice A) and can also be seen in various nonneoplastic conditions, including lichen sclerosus, lichen planus, pityriasis lichenoides chronica, pityriasis lichenoides et varioliformis acuta and CLH (J Invest Dermatol 2000;115:254, J Cutan Pathol 2002;29:447, Arch Dermatol Res 2000;292:568, Arch Dermatol 2001;137:305, Arch Dermatol 2000;136:1483).

Comment Here

Reference: Cutaneous lymphoid hyperplasia

Cutaneous mastocytosis
Definition / general
  • Most cutaneous mast cell disorders have a good prognosis
  • Mastocytosis: monomorphic population of mast cells with rare eosinophils
  • Telangiectasia macularis eruptive perstans (TMEP): telangiectatic light or dark brown macules
  • Urticaria pigmentosa: common form of mastocytosis, numerous small yellow brown papules, become hives when rubbed
Clinical features
  • At birth or during the first 3 months of life
  • May present with flushing attacks due to high histamine content
  • Spontaneous involution frequently occurs
  • CD117 mutations in patients with mastocytosis
  • Urticaria pigmentosa: most common form of mastocytosis of skin; usually childhood onset of multiple brown macules; systemic variant is malignant and involves liver, spleen, bone marrow, lymph nodes and occasionally peripheral blood (mast cell leukemia)
  • Darier's sign: stroking skin releases histamine, causing hives
  • Dermatographism: dermal edema resembling hives due to stroking with pointed instrument

  • Description:
    • Solitary lesion, or small group as part of urticaria pigmentosa
    • Affect extremities and trunk, not palms and soles
    • May blister
    • Red, brown pink or yellow nodules, or plaques measuring up to 1.0 cm
Case reports
Clinical images

Contributed by Mark R. Wick, M.D.

Breast skin



Images hosted on other servers:

Erythematous macules with telangiectasia

Telangiectatic macules in the scapular area

Microscopic (histologic) description
  • Within the macules and plaque, mast cells are predominantly in papillary dermis
  • Mast cells are round or spindle shaped with abundant eosinophilic cytoplasm, distinct cytoplasmic boundaries, large pale nuclei
  • Eosinophils are often present
  • Also edema of papillary dermis, subepidermal vesiculation
  • Bullous mastocytosis may be diagnosed by Tzank smear; infiltrate may be slight and perivascular
  • In telangiectasia macularis eruptive perstans, features may be subtle, with increased mast cells around dilated superficial capillaries, basal cell hyperpigmentation of overlying epidermis, superficial lymphohistiocytic infiltrate
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D. and Case #344

Mastocytoma

Mastocytosis


Leder

Giemsa

Toluidine blue

Mastocytosis


Breast skin

Urticaria pigmentosa, breast skin

Leder stain, breast


H&E

Toluidine blue

Positive stains
Differential diagnosis
  • Normal skin or dermatoses:
    • Must search to find mast cells even with special stains
  • Chronic dermatitis, nodular prurigo, and venous stasis:
    • Typically associated with granulation tissue and foci of neovascularization

Cutaneous mixed tumor / chondroid syringoma
Definition / general
Essential features
Terminology
  • Other names include:
    • Apocrine mixed tumor
    • Eccrine mixed tumor
    • Benign mixed tumor of skin
    • Chondroid syringoma
ICD coding
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
Epidemiology
Sites
Pathophysiology
  • Benign adnexal proliferation with a wide range of differentiation and metaplastic changes in its epithelial, myoepithelial and stromal components
Etiology
  • Unclear
Clinical features
  • Benign; rarely recurs
  • No distinctive clinical characteristics but may mimic pilomatricoma
  • Most often seen in middle aged men; mean age at presentation is 48 years
  • Most commonly in nose, followed by cheek and upper lip
  • Face / head and neck > extremities, trunk or back
  • Calcification / ossification may occur
  • Typically small (0.5 - 3 cm) and painless
  • No specific findings on dermoscopy
  • Rapid, ulcerative growth can be seen in malignant mixed tumor
  • Reference: Calonje: McKee's Pathology of the Skin, 5th Edition, 2019
Diagnosis
  • Biopsy required for diagnosis
Radiology description
  • Findings are not highly specific for chondroid syringoma over other dermal and soft tissue neoplasms (Australas Radiol 2001;45:240)
  • MRI can be helpful to determine the extent and depth of the lesion and relation to anatomic structures in large tumors
  • In the few cases studied, radiography of malignant chondroid syringoma tends to show higher signal intensity and enhancement (Australas Radiol 2001;45:240)
Prognostic factors
  • Benign, with rare recurrence
  • Malignant counterpart (malignant chondroid syringoma) rare with high rate of metastasis (60%) and death (25%) and should be ruled out on microscopic morphology (Pathology 1994;26:237)
Case reports
Treatment
Clinical images

Images hosted on other servers:
Left supraorbital swelling

Left supraorbital swelling

Single well defined nodule

Single well defined nodule

Atrophy and scarring on surface

Atrophy and scarring on surface

Gross description
  • Nodular, circumscribed, nonulcerated, with marked variation in size
  • Cut surfaces are tan-white, often with grossly apparent chondroid component
  • Slow growing, painless, firm, deep dermal to subcutaneous nodule
Gross images

Images hosted on other servers:
Well circumscribed, lobulated nodule

Well circumscribed, lobulated nodule

Microscopic (histologic) description
  • Well circumscribed but unencapsulated, multilobulated mass (separated by fibrous septae) centered in deep dermis or subcutaneous fat, with a prominent chondroid or myxoid stroma enveloping benign bland appearing epithelial and myoepithelial cells that form secondary structures (glands, ducts, cysts, reticular lace-like networks, nests)
  • Biphasic, with both epithelial and stromal components
    • Epithelial: elongated branching tubular structures with 2 cell layers
      • Outer layer is cuboidal or columnar; inner layer is columnar with eosinophilic cytoplasm
      • Decapitation type secretion, often squamous metaplasia and basal nuclei
      • Often has areas of apocrine, follicular and sebaceous differentiation
      • Presence of any follicular or sebaceous features rules out pure eccrine mixed tumor
      • Wide array of morphologies can be seen with all types of differentiation along the folliculosebaceous unit
      • Occasional cases have neoplastic tubules connecting directly to normal follicular infundibulum
      • Follicular differentiation is the most common type of change in the epithelium and may include:
        • Keratinous cysts with infundibular keratinization
        • Isthmic epithelium
        • Hair bulbs with papillary mesenchyme
        • Matrical differentiation with basaloid, transitional and ghost cells
        • Trichohyaline granules
        • Vellous hair shafts
        • Anagen differentiation
      • Sebaceous differentiation may include single cells or islands of mature sebocytes
      • Metaplastic epithelial changes include clear cell change, columnar change, cytoplasmic vacuolization (may be abortive luminal differentiation), hobnailing, mucinous, oxyphilic, squamous
      • Metaplastic myoepithelial changes include clear cell change, collagenous spherulosis, hyaline cells, plasmacytoid cells, spindled cells
    • Stroma: chondroid, mucoid or focally fibrous / hyaline
      • Metaplastic stromal changes include adipocytic, chondroid and osseous metaplasia
      • Adipocytic metaplasia may be extensive; some adipocytes may have Lochkern nuclei with a single small vacuole indenting the nucleus that resemble lipoblasts
  • In contrast to eccrine mixed tumors, apocrine mixed tumors do not have clear cell change, cribriform areas, osseous metaplasia (prominent), physaliferous-like cells, pseudorosettes
  • No pleomorphism or necrosis, sparse mitoses
  • Benign tumors may have:
    • Asymmetry
    • Mild nuclear pleomorphism within the ductal component
    • Multinucleated, bizarre, hyperchromatic cells
    • Infiltrative or pushing borders
  • Features suggestive of malignant transformation include:
    • Severe atypia
    • Markedly increased cellularity
    • Increased mitotic rate
    • Destruction of surrounding tissue
    • Tumor necrosis
  • Reference: Calonje: McKee's Pathology of the Skin, 5th Edition, 2019
Microscopic (histologic) images

Contributed by Angel Fernandez-Flores, M.D., Ph.D., Aravindhan Sriharan, M.D. and J. Mike Magill, Jr., M.D.
Mixed tumor histology

Mixed tumor histology

Islands of epithelial cells Islands of epithelial cells Islands of epithelial cells

Islands of epithelial cells

Epithelial tubules and ducts Epithelial tubules and ducts

Epithelial tubules and ducts


Epithelial ducts Epithelial ducts

Epithelial ducts

Chondroid stroma

Chondroid stroma

Prominent duct formation Prominent duct formation

Prominent duct formation


Metaplastic change Metaplastic change

Metaplastic change

Cytology description
  • Many epithelial cells with bland nuclei in a chondromyxoid background
  • Marked nuclear pleomorphism and prominent nucleoli may be used to identify malignant features on fine needle aspiration (Acta Cytol 1998;42:1155)
  • Findings for diagnosis of chondroid syringoma on fine needle aspiration (Acta Cytol 2010;54:183)
    • Fibrillary chondromyxoid background
    • Prominent cellular component of epithelial and myoepithelial cells
    • Absence of atypia and tumor diathesis (however, histology still required for definitive exclusion of malignant features)
Cytology images

Images hosted on other servers:
Cellular smear

Cellular smear

Cellular aspirates

Cellular aspirates

Positive stains
Negative stains
Molecular / cytogenetics description
Sample pathology report
  • Skin, nose, biopsy:
    • Chondroid syringoma (benign cutaneous mixed tumor)
    • Microscopic description: Histologic sections demonstrate a well circumscribed nodular dermal (subcutaneous) neoplasm. The tumor consists of cords and islands of banal epithelial cells set in a variably fibrous and chondromyxoid stroma, creating a biphasic appearance. Ductal structures are present, with inner epithelial and outer myoepithelial layers evident. Cystic (or other metaplastic) change is seen. There is minimal nuclear atypia and mitoses are not abundant.
Differential diagnosis
  • Cutaneous chondroma:
    • Lacks the epithelial and myoepithelial elements
  • Malignant mixed tumor / malignant chondroid syringoma:
    • Necrosis, infiltrative growth, marked pleomorphism and greater cellularity
  • Metastatic carcinoma:
    • Can often be distinguished based on immunohistochemistry and cytologic features of malignancy
  • Parachordoma:
    • Rare, no true ducts, actin negative, presence of physaliferous cells, arises adjacent to tendon and synovium in extremities
    • Now often considered a variant of myoepithelioma
  • Collagenous spherulosis:
    • More common in breast, lumina contain dense eosinophilic secretion, no biphasic appearance
  • Hidradenoma papilliferum:
    • No chondroid matrix, not biphasic
  • Myopeithelioma / adenomyoepithelioma of the skin:
    • More common on extremities, myoepithelial areas should predominate, lacks luminal epithelial cells
    • Negative for cytokeratin and CEA
  • Other adnexal tumors with apocrine differentiation
Board review style question #1

Features that are most suggestive of malignant behavior in mixed tumor are

  1. Asymmetry, osseous metaplasia and scattered pleomorphic cells
  2. Cytoplasmic vacuolization and plasmacytoid cells
  3. Increased cellularity and necrosis
  4. Infiltrative borders and asymmetry
  5. Infiltrative borders and bizzare, multinucleated cells
Board review style answer #1
C. Increased cellularity and necrosis. Benign tumors may exhibit asymmetry, infiltrative borders, mild atypia, metaplasia and multinucleated cells. Features that suggest malignant mixed tumor are prominent pleomorphism / atypia, necrosis, increased cellularity and mitoses with tissue destruction.

Comment Here

Reference: Chondroid syringoma
Board review style question #2
Most common molecular abnormality seen in chondroid syringoma is

  1. EWSR1 amplification
  2. EWSR1 rearrangement
  3. HMGA2 fusion
  4. PLAG1 deletion
  5. PLAG1 rearrangement
Board review style answer #2
E. PLAG1 rearrangement. Although the characteristic PLAG1 or HMGA2 fusions seen in pleomorphic adenoma of salivary glands are not seen in chondroid syringoma, PLAG1 rearrangement has been detected in a subset of cutaneous mixed tumors. Nuclear immunohistochemical expression of PLAG1 is also seen. A subset of tumors show EWSR1 rearrangement.

Comment Here

Reference: Chondroid syringoma

Cylindroma
Definition / general
  • Benign tumor with debated origin from eccrine sweat glands or hair follicles
  • Also called turban tumor, particularly when a large, multicentric scalp tumor
Familial form (turban tumor syndrome, Brooke-Spiegler syndrome)
  • Autosomal dominant, multiple tumors of children / teenagers that may also involve trunk and extremities
  • In association with spiradenoma, trichoepithelioma, milia or membranous variant of basal cell adenoma of salivary glands
  • Due to mutations in CYLD gene at chromosome 16q12-13
  • Rarely undergoes malignant transformation
Spiradenocylindromas
  • Features of cylindroma and eccrine spiradenoma
  • Benign
  • Solitary or multiple
  • Familial inheritance possible
Essential features
Terminology
  • Cutaneous or dermal cylindromas
  • Turban tumor
  • Salivary cylindroma or cystic adenoid carcinoma
ICD coding
  • ICD-O: 8200/0 - eccrine dermal cylindroma
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
  • ICD-11: 2F22 - benign neoplasms of epidermal appendages
Epidemiology
Sites
Pathophysiology
  • Unclear pathophysiology, currently debated to be from eccrine, apocrine or follicular origin (Int J Dermatol 2015;54:275)
  • MYB gene fuses with NFIB gene forming single sporadic tumors (J Pathol 2016;239:391)
  • Germ line mutation on CYLD tumor suppressor gene on chromosome 16 leads to Brooke-Spiegler syndrome; autosomal dominant inheritance pattern (Head Neck Pathol 2016;10:125)
Etiology
  • Currently unknown
Clinical features
  • Solitary or multiple
  • Primarily on head, neck and scalp
  • Usually ages 40+ years
  • F > M
  • Rarely associated with similar tumors in major salivary glands
  • Rare malignant transformation, local infiltration common (Lancet Oncol 2015;16:e460)
  • Brooke-Spiegler syndrome associated with spiradenoma and trichoepithelioma formation (Dermatol Clin 2017;35:61)
Diagnosis
  • Diagnosis based on biopsy histology (Dermatol Clin 2017;35:61)
  • Consider genetic testing in patients with multiple cylindromas, spiradenomas or trichoepitheliomas and in patients with one of these solitary tumors and a first degree relative with any of these tumors (Dermatol Clin 2017;35:61)
Radiology description
  • MRI imaging utilized with suspected malignant transformation of scalp tumors to detect intracranial invasion (Lancet Oncol 2015;16:e460)
Prognostic factors
Case reports
Treatment
  • Treatment for cylindromas with CYLD mutations or tumors that are disfiguring or uncomfortable is with surgical excision (Dermatol Clin 2017;35:61)
  • Mohs micrographic surgery may be necessary with recurrent tumors to confirm histologic clearance (Lancet Oncol 2015;16:e460)
  • Radiotherapy is contraindicated as it can further induce DNA damage (Dermatol Clin 2017;35:61)
Clinical images

Images hosted on other servers:
Patient with Brooke-Spiegler syndrome

Brooke-Spiegler syndrome

Multiple scalp tumors

Facial tumor

Scalp tumor

Gross description
  • Pink-red dome shaped nodule with smooth surface
Gross images

Images hosted on other servers:

White lobulated cut surface

Microscopic (histologic) description
  • Compact nests of basaloid cells that fit together like a jigsaw puzzle, surrounded by thick basement membrane (Dermatol Clin 2017;35:61)
  • Cylindrical shape appreciated in cross section (Dermatol Clin 2017;35:61)
  • 2 groups of cells:
    • Undifferentiated peripheral palisading epithelial cells with small dark nuclei
    • Centrally located increasingly differentiated ductal cells with large pale nuclei (Dermatol Clin 2017;35:61)
Microscopic (histologic) images

Contributed by Mary Dick, M.D.
Unencapsulated tumor

Unencapsulated tumor

Tumor lobule nests

Tumor lobule nests

Surrounding hyaline

Surrounding hyaline

Hyaline droplets within tumor

Hyaline droplets within tumor

Two cell populations

2 cell populations

Virtual slides

Images hosted on other servers:
Dermal tumor in jigsaw pattern

Dermal tumor in jigsaw pattern

Positive stains
Electron microscopy description
  • Differentiation towards intradermal coiled duct region of eccrine sweat glands
  • Compact small cells with dark nuclei on the periphery with larger cells containing light colored nuclei more centrally
  • Ductal structures and thick hyaline bands present
Molecular / cytogenetics description
  • Mutation in CYLD on chromosome 16 associated with multiple familial cylindromas
  • MYB gene mutations associated with sporadic cylindromas
  • Chromosome 16 CYLD mutation common
    • CYLD codes for recessive cylindromatosis familial tumor suppressor gene a ubiquitin specific protease (Lancet Oncol 2015;16:e460)
    • Majority of mutations are frameshift and result in truncated proteins (Hum Mutat 2009;30:1025)
    • Mutations detected in 84% of reported Brooke-Spiegler patients and 88% in familial cylindromatosis (Hum Mutat 2009;30:1025)
  • MYB activation (~12%) and MYB-NFIB fusion oncoproteins (~55%) present in sporadic tumors (J Pathol 2016;239:197)
Videos

Spiradenoma and cylindroma sweat gland tumor

Sample pathology report
  • Forehead, excision:
    • Benign adnexal tumor (see comment)
    • Comment: There are cylindrically shaped nests of epithelial cells arranged in a jigsaw pattern surrounded by thick hyaline basement membrane. Immunohistochemically the tumor nests are express CK7 and 8 positivity, with EMA and CEA positivity in the accompanying duct structures. Tumor is CK20, GCDFP-15, ER and PR negative. These features support the diagnosis of cylindroma.
    • Malignant transformation is unlikely. If multiple tumors are present, consider genetic testing for CYLD mutations as this is associated with Brooke-Spiegler.
Differential diagnosis
  • Trichoepithelioma:
    • Hair follicle tumor with basaloid cells in nests surrounded by dense fibrous stroma and spindle cells
    • Mucin pools may be present within basaloid nests
    • Lack of clefting distinguishes from basal cell carcinoma
  • Spiradenoma:
    • Can occur both separately and in combination with cylindromas
    • Similar formation with peripheral small basaloid cells and larger paler cells centrally
    • Lobulated tumor islands separated by thin fibrous tissue containing blood vessels
  • Basal cell carcinoma:
    • Lacks tight hyaline membrane separating epithelial islands
    • Mucinous stroma with clefting artifact present
Board review style question #1
Cylindroma Cylindroma


    A 31 year old woman who was previously diagnosed with Brooke-Spiegler syndrome has developed three painless cutaneous tumors on her scalp. Tumor biopsy shows a jigsaw pattern of basaloid ovoid nests of tumor lobules located within the dermis, ductal structures and thick hyaline bands. Which of the following is true concerning this adnexal tumor?

  1. It can appear both separately and in conjunction with spiradenomas
  2. It most commonly appears on elderly males
  3. Malignant transformation occurs frequently
  4. Tumors are normally found on the lower extremities
Board review style answer #1
A. It can appear both separately and in conjunction with spiradenomas

Comment Here

Reference: Cylindroma
Board review style question #2
    A 26 year old woman presents with multiple tumors on her scalp, neck and face. She otherwise feels well. Her mother has similar appearing tumors that have previously been diagnosed as cylindromas and spiradenomas. This is her first time seeking diagnosis as her tumors are disfiguring but generally painless. Testing for mutation in which gene should be recommended for this patient?

  1. CYLD
  2. MYB
  3. MYB-NFIB
  4. No genetic testing is recommended
Board review style answer #2
A. CYLD

Comment Here

Reference: Cylindroma

Cystic / cavernous lymphangioma

Dermatofibroma (cutaneous fibrous histiocytoma)
Definition / general
  • Dermatofibromas (also known as fibrous histiocytomas) are a spectrum of benign dermal based lesions with fibroblastic and histiocytic differentiation
  • There is debate as to whether dermatofibroma is a reactive or neoplastic process
Essential features
  • Benign dermal based, nodular proliferation of fibroblasts and histiocytes
  • Collagen trapping at periphery and follicular induction commonly seen
  • IHC is nonspecific for dermatofibroma
Terminology
  • Benign fibrous histiocytoma and fibrous histiocytoma
ICD coding
  • ICD-10: D23 - benign neoplasm of skin
  • ICD-11: 2F23.0 - dermatofibroma
Epidemiology
Sites
  • Typically occurs on distal extremities (legs > arms > trunk)
  • May present on any part of the skin surface
Pathophysiology
  • Cause unknown
  • No evidence associates it with insect bites or trauma
Etiology
Diagrams / tables

Images hosted on other servers:

Various presentations under dermoscopy

Clinical features
  • Painless
  • 5 mm - 2 cm
  • Variable color: skin colored to brown to purple
  • Variable shape: plaques, nodules or polyps
  • Covered by intact skin
  • Suspected by pinching the nodule between the fingers and observing that the tumor is fixed within the dermis
  • Pinch sign: overlying skin dimples on pinching the lesion (An Bras Dermatol 2014;89:472)
Diagnosis
  • Combination of clinical and histologic findings
Prognostic factors
Case reports
Treatment
  • Complete excision is curative
  • Wide local excision is adequate to prevent recurrence
  • Spontaneous regression has been reported
Clinical images

Images hosted on other servers:

Dermatofibroma of the skin

Dermatofibroma of the skin

Pinch sign of dermatofibroma


Hyperpigmented dermatofibroma

Numerous eruptive dermatofibromas

Dermatofibroma of the lower lip

Dermatofibroma presenting as eruptive papules

Dermatofibroma of the shoulder


Patterns of dermatofibroma

Vascular structures in dermatofibromas

Other dermoscopic structures

Gross description
  • Most classic pattern: pigment network and central white patch
  • Wide range of presentations
Gross images

Images hosted on other servers:
Ulcerated cutaneous mass

Polypoid, ulcerated mass

Microscopic (histologic) description
  • Symmetric and predominantly dermal based
  • Relatively circumscribed but have irregular and unencapsulated borders
  • Patterns range from diffuse to reticular to hemangioma-like to keloid-like
  • Classic pattern: storiform, pinwheel or curlicue pattern
    • Made up of spindled fibroblasts or histiocytes
  • Some areas densely cellular, while others are sclerotic and hypocellular
    • Early lesions: more cellular
    • Later lesions: more sclerotic
  • Spindled cells: thin, elongated nuclei with pointed ends and eosinophilic cytoplasm
  • Histiocytic cells: epithelioid shaped cells with abundant pale cytoplasm
  • May see varying amounts of inflammatory cells
  • Cytologic atypia and pleomorphism are variable
  • Mitotic activity is variable
  • Touton giant cells and ringed lipidized siderophages may be present
  • Collagen trapping at periphery
    • Spheres of eosinophilic collagen (collagen balls) surrounded by the fibroblast proliferation
  • Grenz zone (sparing of the superficial papillary dermis) commonly seen
  • Follicular or sebaceous induction and basilar hyperpigmentation
  • Tumor may extend into superficial fat
  • Numerous histological subtypes (An Bras Dermatol 2014;89:472):
Microscopic (histologic) images

Contributed by Brandon Zelman, D.O. and Jodi Speiser, M.D.
Spindled fibroblasts and epithelioid histiocytes

Spindled fibroblasts and epithelioid histiocytes

Dermal based proliferation Dermal based proliferation

Dermal based proliferation

Dermal proliferation

Dermal proliferation

Whirled appearance

Whirled appearance

Collagen trapping

Collagen trapping


Increased cellularity and collagen trapping

Increased cellularity and collagen trapping

Dermal based proliferation that appears “busy”

Dermal based proliferation that appears busy

 Increased dermal cellularity

Increased dermal cellularity

Spindled fibroblasts and epithelioid histiocytes

Spindled fibroblasts and epithelioid histiocytes

CD34-

CD34-

CD10+

CD10+


Keratin negative

Keratin-

S100-

S100-

SMA weakly expressed

SMA weakly expressed

CD34- CD34-

CD34-

CD10+

CD10+



AFIP images

Aneurysmal
Nonendothelial lined clefts or lakes containing blood Nonendothelial lined clefts or lakes containing blood

Nonendothelial lined clefts or lakes containing blood

Large amounts of hemosiderin

Large amounts of hemosiderin

Hemosiderin with marked sclerosis

Hemosiderin with marked sclerosis



Cellular

More cellular than classic fibrous histiocytoma

Elongated cells are arranged in fascicles or a storiform pattern

Tumor cells are more histiocyte like and foam cells are present


Epithelioid
Histiocyte-like cells with abundant cytoplasm, no / rare spindle cells Histiocyte-like cells with abundant cytoplasm, no / rare spindle cells

Histiocyte-like cells with abundant cytoplasm, no / rare spindle cells

Epithelioid cells in hyalinized stroma

Epithelioid cells in hyalinized stroma

Virtual slides

Images hosted on other servers:

Dermatofibroma

Positive stains
Negative stains
Videos

Dermatofibroma

Anerysmal fibrous histiocytoma


Cellular dermatofibroma

Lipidized dermatofibroma

Sample pathology report
  • Skin, right arm, excision:
    • Dermatofibroma

  • Skin, left shin, excision:
    • Dermatofibroma (see comment)
    • Comment: Immunohistochemical stains were performed on the above specimen. The cells are positive for factor XIIIa and negative for CD34. This supports the above diagnosis.
Differential diagnosis
Board review style question #1
Which of the following is true of dermatofibromas?

  1. Cellular proliferation involves both the epidermis and dermis
  2. CD34 is positive at the center of the lesion
  3. Collagen trapping is seen at the periphery of the lesion
  4. Deep margins of the lesion typically show extensive infiltration
Board review style answer #1
C. Collagen trapping is seen at the periphery of the lesion

Comment Here

Reference: Dermatofibroma (cutaneous fibrous histiocytoma)
Board review style question #2
Which immunohistochemical profile would be seen in a dermatofibroma?

  1. S100- / CD34- / CD163+ / MART1-
  2. S100- / CD34+ / CD163+ / MART1-
  3. S100+ / CD34- / CD163- / MART1+
  4. S100+ / CD34- / CD163+ / MART1-
  5. S100+ / CD34+ / CD163+ / MART1-
Board review style answer #2
A. S100- / CD34- / CD163+ / MART1-

Comment Here

Reference: Dermatofibroma (cutaneous fibrous histiocytoma)

Dermatofibrosarcoma protuberans (DFSP)
Definition / general
  • Locally aggressive, superficial mesenchymal neoplasm with fibroblastic differentiation
Essential features
  • Locally aggressive, superficial mesenchymal neoplasm with fibroblastic differentiation
  • Virtually all cases contain fusion genes; COL1A1::PDGFB is the most common fusion product, although others have recently been reported
  • Fibrosarcomatous transformation imparts an increased risk of recurrence and metastasis
ICD coding
  • ICD-O:
    • 8832/3 - dermatofibrosarcoma protuberans, NOS
    • 8833/3 - pigmented dermatofibrosarcoma protuberans
    • 8834/1 - giant cell fibroblastoma
  • Fibrosarcomatous dermatofibrosarcoma protuberans: no distinct coding identified
Epidemiology
Sites
  • Can ostensibly involve any area of skin but the trunk and extremities are the most common locations
Pathophysiology
  • Tumors are generally presumed to occur sporadically
  • Virtually all cases contain fusion genes; COL1A1::PDGFB is the most common fusion product, although others have been reported
  • Possible association with adenosine deaminase deficient severe combined immunodeficiency (J Allergy Clin Immunol 2012;129:762)
Clinical features
  • Classically an exophytic, nodular cutaneous mass; however, often initially presents as a flat plaque (JAMA Netw Open 2019;2:e1910413)
  • Initially may show persistent slow growth, often for many years, then sudden progression (Cancer 1962;15:717)
  • Fibrosarcomatous transformation is associated with metastatic potential (Cancer 2000;88:2711)
Diagnosis
  • Tumors are morphologically distinctive and frequently amenable to classification based on H&E
  • Immunohistochemistry for CD34 is a useful adjunct since most cases are diffusely positive
  • Molecular testing is helpful, particularly in the context of limited sampling or unusual morphology
Radiology description
Radiology images

Images hosted on other servers:
Multiple examples from radiopaedia.org

Nodular soft tissue mass

Prognostic factors
  • Incomplete resection is a risk factor for recurrence
  • Fibrosarcomatous transformation (fibrosarcoma ex DFSP) imparts an increased risk of recurrence and metastasis (Am J Surg Pathol 2006;30:436)
  • Metastases typically occur following multiple local recurrences
  • Increased age, male sex and tumor size are associated with worse overall survival (JAMA Dermatol 2016;152:1365)
Case reports
Treatment
Clinical images

Images hosted on other servers:
Papulonodular thigh lesions

Papulonodular thigh lesions

Before and after imatinib treatment

Before and after imatinib treatment

Gross description
Gross images

Images hosted on other servers:
Scalp tumor

Scalp tumor

Microscopic (histologic) description
Microscopic (histologic) images

Contributed by Brendan C. Dickson, M.D., M.Sc.
Infiltration along fibrous septa

Infiltration along fibrous septa

Honeycomb pattern

Honeycomb pattern

Nuclear monomorphism

Nuclear monomorphism

Storiform pattern

Storiform pattern

Herringbone pattern in fibrosarcoma ex DFSP

Herringbone pattern in fibrosarcoma ex DFSP

Brisk mitotic activity in fibrosarcoma ex DFSP

Brisk mitotic activity in fibrosarcoma ex DFSP


Pigmented DFSP

Pigmented DFSP

Myxoid DFSP

Myxoid DFSP

DFSP post imatinib therapy

DFSP post imatinib therapy

Diffuse CD34 staining

Diffuse CD34 staining

Diminished CD34 in fibrosarcoma ex DFSP

Diminished CD34 in fibrosarcoma ex DFSP

Virtual slides

Images hosted on other servers:
35 year old man with lesion on right upper arm

35 year old man with lesion on right upper arm

81 year old woman with breast lesion

81 year old woman with breast lesion

40 year old man with lesion on abdomen

40 year old man with lesion on abdomen

27 year old man with lesion on abdomen

27 year old man with lesion on abdomen

Positive stains
Negative stains
Electron microscopy description
Molecular / cytogenetics description
Molecular / cytogenetics images

Images hosted on other servers:
Fusion product of <i>COL1A1</i> to <i>PDGFB</i> Fusion product of <i>COL1A1</i> to <i>PDGFB</i>

Fusion product of COL1A1 to PDGFB

<i>PDGFB</i> break apart FISH

PDGFB break apart FISH

Sample pathology report
  • Skin, back, biopsy:
    • Dermatofibrosarcoma protuberans (see comment)
    • Comment: Within the dermis and subcutis there is a spindle cell neoplasm with a storiform pattern. The cytoplasm is eosinophilic. The nuclei are ovoid and monomorphic with rare mitotic activity. There is sparing of adnexal structures and infiltration of the subcutaneous fat with a honeycomb pattern. The cells are diffusely positive for CD34; they are negative for desmin, smooth muscle actin, S100 and keratin (AE1 / AE3).
Differential diagnosis
  • Cellular fibrous histiocytoma:
    • Plump spindle cells with peripheral collagen entrapment
    • Inflammation is often present, including foamy macrophages, lymphocytes and plasma cells; occasionally multinucleated giant cells (J Cutan Pathol 2012;39:747)
    • Immunohistochemistry for CD34 may highlight peripheral dermal fibroblasts; rarely, tumors may be positive (J Cutan Pathol 2012;39:747)
  • Cutaneous leiomyosarcoma:
    • Plump spindle cells with a fascicular architecture
    • Ovoid / cigar shaped nuclei, atypical mitoses may be identified
    • Immunohistochemistry typically positive for desmin and h-caldesmon and negative for CD34
  • Solitary fibrous tumor:
    • Spindle cells with a patternless distribution; prominent branching vasculature
    • Keloid-like collagen bundles
    • Immunohistochemistry will also be positive for STAT6
Board review style question #1

Which immunohistochemical stain would be diffusely positive in typical dermatofibrosarcoma protuberans tumor (such as the one shown in the image above)?

  1. CD34
  2. Desmin
  3. HMB45
  4. Pankeratin
  5. S100
Board review style answer #1
A. CD34 is typically diffusely positive in cases of dermatofibrosarcoma protuberans while desmin, HMB45, pankeratin and S100 are not expressed.

Comment Here

Reference:Dermatofibrosarcoma protuberans (DFSP)
Board review style question #2
The presence of which of the following fusion genes can be used to support a diagnosis of dermatofibrosarcoma protuberans in a primary dermal spindle cell neoplasm?

  1. COL1A1::PDGFB
  2. EWSR1::FLI1
  3. FUS::DDIT3
  4. JAZF1::SUZ12
  5. MYB::NFIB
Board review style answer #2
A. COL1A1::PDGFB fusion product is present in most cases of dermatofibrosarcoma protuberans. It is important to note that additional fusion genes are possible but are missed by FISH or reverse transcription PCR assays that are restricted to PDGFB. It is also important to note that other tumors may harbor this fusion product (e.g., uterus, cervix). The other fusion gene options do not occur in dermatofibrosarcoma protuberans and are characteristic of other neoplasms:
  • EWSR1::FLI1: Ewing sarcoma
  • FUS::DDIT3: myxoid liposarcoma
  • JAZF1::SUZ12: low grade endometrial stromal sarcoma
  • MYB::NFIB: adenoid cystic carcinoma

Comment Here

Reference: Dermatofibrosarcoma protuberans (DFSP)
Board review style question #3

Which of the following is true regarding the entity shown above?

  1. Positive for Fontana-Masson and CD34
  2. Positive for Fontana-Masson and factor XIIIa
  3. Positive for Prussian blue and CD34
  4. Positive for Prussian blue and factor XIIIa
Board review style answer #3
A. Positive for Fontana-Masson and CD34. This is an example of pigmented dermatofibrosarcoma protuberans (Bednar tumor). The pigment is melanin, which is positive with the Fontana-Masson stain but not Prussian blue. The tumor cells are typically diffusely positive for CD34 but negative for factor XIIIa.

Comment Here

Reference: Dermatofibrosarcoma protuberans (DFSP)
Board review style question #4
The molecular pathogenesis of most cases of dermatofibrosarcoma protuberans is characterized by which of the following fusion gene products?

  1. COL6A3::CSF1
  2. COL1A1::PDGFB
  3. COL6A3::PDGFD
  4. COL1A1::USP6
Board review style answer #4
B. COL1A1::PDGFB. COL6A3::CSF1 is found in a subset of tenosynovial giant cell tumors. COL6A3::PDGFD is only rarely encountered in dermatofibrosarcoma protuberans. COL1A1::USP6 may be present in myositis ossificans, a fibro-osseous pseudotumor of the digits and aneurysmal bone cyst.

Comment Here

Reference: Dermatofibrosarcoma protuberans (DFSP)

Dermatomyofibroma
Definition / general
Case reports
Clinical images

Contributed by Mark R. Wick, M.D.


Images hosted on other servers:
Ill defined bluish firm plaque on the left flank

Ill defined bluish firm plaque on the left flank

Gross description
  • Reddish brown plaque or nodule
Microscopic (histologic) description
  • Fascicles of monomorphic spindle cells parallel to epidermis
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D.
Breast skin Breast skin

Breast skin

Soft tissue Soft tissue

Soft tissue

MSA stain

MSA stain



Images hosted on other servers:
H&E and smooth muscle actin H&E and smooth muscle actin

H&E and smooth muscle actin

Negative stains
Differential diagnosis

Dermatosis papulosa nigra
Definition / general
  • Benign epidermal growth presented as hyperpigmented or skin colored papules on face and neck, most commonly found on patients with darker phototypes (Fitzpatrick type IV, V or VI)
  • Likely a variant of seborrheic keratosis
Essential features
  • Small (several millimeters) hyperpigmented or flesh colored papules seen most commonly in patients with skin of color, particularly darker phototypes (Fitzpatrick type IV, V or VI)
  • Increase in size and number of papules with aging
  • Familial
  • Clinical: filiform, verrucous or pedunculated pigmented papule
  • Histology: acanthosis, papillomatosis, hyperkeratosis and hyperpigmentation
Terminology
  • First described in 1925 by Dr. Aldo Castellani
  • Seborrheic keratosis
ICD coding
  • ICD-10: L82.1 - other seborrheic keratosis
Epidemiology
  • Common in patients with skin of color, particularly darker phototypes (Fitzpatrick type IV, V or VI), affecting 10 - 75% of the population (Clin Dermatol 2017;35:491)
  • Onset often late in adolescence, increasing numbers with age, common in adulthood and age > 60 years
  • Familial in majority of cases (Int J Dermatol 2017;56:957)
  • F:M = 2:1
Sites
  • Mainly in photoexposed areas: head, neck and upper trunk
Pathophysiology
  • Not entirely known
  • Frequent positive family history and possible genetic predisposition
  • Somatic activation mutation in FGFR3 has been rarely reported (Br J Dermatol 2010;162:508)
  • Cumulative ultraviolet (UV) exposure may play a role
Etiology
  • Not entirely known
  • Aging
  • UV exposure
Clinical features
  • Round, 1 - 5 mm, skin colored to hyperpigmented fleshy papules mimicking skin tags, warts or seborrheic keratoses
  • Filiform to sessile, smooth surface to wrinkled papules in a symmetric distribution
  • Distribution is typically head and neck, upper chest
  • Generally asymptomatic papules without crusting, scaling or ulceration
  • No spontaneous resolution (Int J Dermatol 2017;56:957)
Diagnosis
  • Diagnosis is usually clinical
    • Dermatoscopy: fissures and ridges in a cerebriform pattern in 59% of cases, comedo-like openings in 27% and milia-like cysts (Int J Dermatol 2017;56:957)
  • Biopsy often performed if concerned for malignancy
  • Histologic findings are confirmatory
Prognostic factors
  • No risk for transforming to malignancy
  • Never regresses spontaneously
Case reports
Treatment
  • Not necessary; for cosmetic purposes
  • Caution in choosing treatment option due to tendency of darker skin to dyspigmentation
  • Snip excision with scissors
  • Light curettage with or without anesthesia
  • Electrodesiccation (Dermatol Surg 2009;35:1079)
  • Cryotherapy
  • Potassium titanyl phosphate (KTP), pulse dye laser (PDL), Nd:YAG and carbon dioxide lasers (Indian J Dermatol 2015;60:321, Dermatol Surg 2010;36:1968)
Clinical images

Contributed by Sara C. Shalin, M.D., Ph.D.

Well circumscribed filiform lesions

Flat topped papules

Gross description
  • Round to filiform, skin colored or hyperpigmented polyp / papule ranging from 1 - 5 mm
Microscopic (histologic) description
  • Irregular acanthosis of epidermis, hyperkeratosis and papillomatosis
  • Elongated and interconnected rete ridges with hyperpigmentation of basal layer
  • No increase in melanocyte number
  • Keratin filled invagination of epidermis
  • Prominent and well developed fibrous stroma within papillomatous acanthotic structures
  • Similar features to reticulate and acanthotic types of seborrheic keratosis (Int J Dermatol 2017;56:957)
Microscopic (histologic) images

Contributed by Sara C. Shalin, M.D., Ph.D.

Hyperkeratotic papule with papillomatosis

Pedunculated papule

Basal hyperpigmentation

Positive stains
Negative stains
  • No stains are required to make the diagnosis
  • Melanocytic markers (SOX10, MART1, HMB45) will show well spaced melanocytes at the dermal epidermal junction (no increase)
Sample pathology report
  • Skin, right shoulder, shave biopsy:
    • Dermatosis papulosa nigra
Differential diagnosis
  • Seborrheic keratosis:
    • Essentially identical histopathologic features
    • Distinguishing features are predominantly clinical
    • Not all clinicians agree with dermatosis papulosa nigra being a variant of seborrheic keratosis
    • Affects all skin types, including lighter skin phototypes (Fitzpatrick skin type I to II) (Int J Dermatol 2007:46:45)
    • Not limited to sun exposed areas
    • Can be larger in size (up to several cm)
  • Verruca vulgaris:
    • Papillomatosis, tiers of parakeratosis, valleys of hypergranulosis and dilated papillary dermal blood vessels
  • Acrochordon / fibroepithelial polyp / skin tag:
    • Polypoid fragment of skin lined by unremarkable (not significantly acanthotic) epidermis with abundant loose fibrocollagenous stroma, abundant vessels and no adnexal structures
  • Melanocytic nevi:
    • Nest of melanocytes in dermoepidermal junction or within dermis
  • Actinic keratosis:
    • Proliferation of atypical keratinocytes at basal layer, loss of granular layer, parakeratosis, solar elastosis in dermis
Board review style question #1

(Disclaimer: this question has not been reviewed or approved by Dr. Shalin)

65 year old woman with darkly pigmented skin (Fitzpatrick type V) presented with multiple pigmented lesions on her cheek with the microscopic features shown above. What is the best diagnosis?

  1. Actinic keratosis
  2. Cutaneous fibroepithelial polyp
  3. Dermatosis papulosa nigra
  4. Verruca vulgaris
Board review style answer #1
C. Dermatosis papulosa nigra. This is a dermatosis papulosa nigra showing irregular acanthosis, hyperpigmentation of basal layer and papillomatosis.

Comment Here

Reference: Dermatosis papulosa nigra
Board review style question #2
(Disclaimer: this question has not been reviewed or approved by Dr. Shalin)

What are the most important histology findings of dermatosis papulosa nigra?

  1. Increased melanocyte proliferation in epidermis
  2. Papillomatosis, acanthosis and hyperkeratosis
  3. Papillomatosis, parakeratosis, valleys of hypergranulosis and dilated papillary dermal blood vessels
  4. Parakeratosis, proliferation of atypical keratinocytes at basal layer and loss of granular layer
Board review style answer #2
B. Papillomatosis, acanthosis and hyperkeratosis. Microscopic findings of dermatosis papulosa nigra are hyperkeratosis, papillomatosis, elongated and interconnected rete ridges with hyperpigmentation of basal layer and irregular acanthosis.

Comment Here

Reference: Dermatosis papulosa nigra

Dermoid cyst
Definition / general
  • Benign cutaneous developmental anomaly
  • Arises from the entrapment of ectodermal elements along the lines of embryonic closure
Essential features
  • Derived from both ectoderm (stratified squamous epithelium) and mesoderm (cutaneous adnexal structures)
  • Most commonly on the lateral aspect of the upper eyelid
  • Can be complicated by intracranial involvement
  • Total resection of the cyst, along with the adhesions and sinuses, is crucial
Terminology
  • Cystic teratoma (not true teratoma), nasal dermoid sinus cyst (when located in sinus)
ICD coding
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
Epidemiology
Sites
Pathophysiology
Etiology
  • Mostly unknown
Clinical features
Diagnosis
  • Based on clinical features, imaging modalities and histopathological findings
Radiology description
  • CT imaging is better at delineating the changes in bony erosions
  • Magnetic resonance imaging (MRI) is the preferred means of revealing evidence of intracranial extension
  • Ultrasound (USG) demonstrates a well defined homogenous and hypoechoic cystic lesion
  • Radiological features include characteristic signal intensities, absence of perilesional edema and the presence of well defined margins to the cyst (Case Rep Neurol Med 2021;2021:9917673)
Radiology images

Contributed by Aayushma Regmi, M.B.B.S. and Jodi Speiser, M.D.
Ultrasonography of a palpable lump over the left eyebrow Ultrasonography of a palpable lump over the left eyebrow

Ultrasonography of a palpable lump over the left eyebrow



Images hosted on other servers:
Lateral dermoid cyst with characteristic hyperdense cyst wall and hypodense cyst cavity.

CT image without contrast

Prognostic factors
Case reports
Treatment
  • Complete surgical resection is the treatment of choice
  • Imaging done prior to determine the extent of underlying tissue involvement
  • Resected carefully to avoid spilling of contents intracranially
  • Recurrences have been reported in case of incomplete excision (Pediatr Dermatol 2013;30:706)
Clinical images

Contributed by Aayushma Regmi, M.B.B.S. and Jodi Speiser, M.D.
Left lateral eyebrow mass

Left lateral eyebrow mass

Left limbal mass

Left limbal mass



Images hosted on other servers:
Lower lid

Lower lid

Lateral eyebrow

Lateral eyebrow

Microscopic (histologic) description
  • Well defined wall lined by stratified squamous epithelium with mature skin appendages (hair follicles and sebaceous glands) and a lumen filled with keratin and hair shafts (Yale J Biol Med 2014;87:349, Pediatr Dermatol 2013;30:706)
  • Occasionally, smooth muscle may be present but in contrast to benign cystic teratoma, cartilage and bone has not been described (Yale J Biol Med 2014;87:349)
  • Eccrine sweat glands present in 35% of cases and apocrine glands in 15%
  • Ruptured cyst with evidence of granulomatous inflammation with giant cells, most concentrated around exposed hair follicles
  • Implantation dermoid cysts have no adnexal structures and therefore, they are easily distinguished from congenital forms on histology (BMJ Case Rep 2019;12:e228831)
Microscopic (histologic) images

Contributed by Aayushma Regmi, M.B.B.S. and Jodi Speiser, M.D.
Well defined cyst Well defined cyst

Well defined cyst

Dermoid cyst wall and contents Dermoid cyst wall and contents

Dermoid cyst wall and contents

Dermoid cyst wall and contents Dermoid cyst wall and contents

Dermoid cyst wall and contents


Lining epithelium in dermoid cyst Lining epithelium in dermoid cyst

Lining epithelium in dermoid cyst

Ruptured dermoid cyst Ruptured dermoid cyst Ruptured dermoid cyst

Ruptured dermoid cyst

Virtual slides

Images hosted on other servers:

Well defined dermoid cyst and ruptured cyst

Positive stains
Negative stains
Videos

Cutaneous dermoid cyst

Sample pathology report
  • Skin, right lower eyelid, excision:
    • Dermoid cyst
  • Skin, left shin, excision:
    • Dermoid cyst (see comment)
    • Comment: The sections show a dermal cyst lined by stratified squamous epithelium with attached sebaceous gland and hair follicle. The lumen contains loose keratin flakes and scattered hair shafts.
Differential diagnosis
  • Epidermal inclusion cyst / epidermoid cyst:
    • Smooth, dome shaped firm, skin colored nodule that is freely movable on palpation and sometimes has a small, dilated punctum
    • Lined only by stratified squamous epithelium; no adnexal structures identified
    • Lumen contains abundant keratin flakes
  • Vellus hair cyst:
    • Multiple, small, smooth, skin colored, reddish, bluish, yellowish, brown, violaceous or grayish papules
    • Mid dermal cyst containing laminated keratin and many vellus hairs
    • Epithelial lining consists of several layers of squamous epithelium, often with a granular cell layer
  • Steatocystoma:
    • Skin colored to yellowish dermal cystic papules or nodules
    • Cyst lined by stratified squamous epithelium without a granular layer with attached sebaceous gland
    • Diagnostic, corrugated eosinophilic layer lining luminal surface
  • Congenital dermoid fistula:
    • Presents at birth as a superficial fistula tract
  • Pilomatrixoma:
    • Benign tumor arising from hair matrix
    • Solid nests of basaloid cells undergoing abrupt trichilemmal type keratinization
    • Ghost cells, calcification or ossification with extramedullary hematopoiesis and frequent rupture leading to giant cell reaction
  • Encephalocele:
    • Can also be found in glabella area
    • Compressible / soft and bluish in color
    • Typically transilluminate
  • Nasal glioma:
    • Not a true neoplasm
    • Typically a firm nodule that is incompressible and red
    • Can be found in glabella area
    • Mature astrocytes, gloss and variable stromal fibrosis
    • S100 and GFAP positive
  • Pilar / trichilemmal cyst:
    • Firm and well circumscribed nodule on palpation
    • Lined by stratified squamous epithelium, which lacks granular layer
    • Contains dense eosinophilic keratin
  • Teratoma:
    • Represents true neoplasm comprised of tissues from all 3 germ layers: ectoderm, mesoderm and endoderm
Board review style question #1
A 5 year old girl with no significant past medical history presents with a slowly growing, nontender mass on the lateral third of her eyebrow. The mass is a firm, nonpulsatile, pale to flesh colored, subcutaneous nodule. Which of the following is the next best step in the management of this condition?

  1. Biopsy
  2. Excision
  3. MRI
  4. Radiograph (Xray)
Board review style answer #1
C. MRI. Before doing therapeutic excisional biopsy, proper imaging should be done due to risk of possible bony deformities, intracranial extension or intraspinal extension. Biopsy may lead to further complications worsening the infection due to rupture / spill of the content of the cyst. MRI is better than radiograph (Xray) as it evaluates the extent of intracranial and intraspinal extension, whereas Xray may detect the bone deformities.

Comment Here

Reference: Dermoid cyst
Board review style question #2

What findings in the above picture are specific to dermoid cyst?

  1. Cyst lined by stratified squamous epithelium and abundant keratin flakes
  2. Cyst lined by stratified squamous epithelium and sebaceous glands
  3. Cyst lined by stratified squamous epithelium with multiple hair shafts
  4. Cyst lined by stratified squamous epithelium with retained the granular layer
  5. Cyst lined by stratified squamous epithelium, sebaceous glands and hair follicle
Board review style answer #2
E. Dermoid cyst is lined by stratified squamous epithelium, sebaceous glands and hair follicles and is composed of abundant keratin flakes. Pilar cyst is lined by stratified squamous epithelium lacking the granular layer and contains dense laminated eosinophilic keratin. Epidermoid cyst is lined by stratified squamous epithelium with retained the granular layer and keratin flakes. Vellus hair cyst is lined by stratified squamous epithelium with multiple hair shafts. Steatocystoma is a cyst lined by stratified squamous epithelium and sebaceous glands.

Comment Here

Reference: Dermoid cyst

Diffuse dermal angiomatosis (pending)
[Pending]

Digital myxoid cyst (pending)

Digital papillary adenocarcinoma
Definition / general
  • Malignant adnexal tumor with a marked predilection of acral sites
  • Helwig first described as eccrine acrospiroma in 1979 (J Cutan Pathol 1987;14:129)
Essential features
  • Malignant adnexal tumor with a marked predilection of acral sites
  • Usually fingers and toes, rarely palm and sole
  • Clinically confused with a cystic lesion
  • Tumor cells are basaloid / cuboidal to low columnar epithelial cells and myoepithelial cells with mild to moderate cytologic atypia
  • Rarely, metastasis to lung and lymph node
Terminology
  • Also called digital papillary eccrine adenoma, aggressive digital papillary adenoma, digital adenocarcinoma
ICD coding
  • ICD-O: 8408/3 - Digital papillary adenocarcinoma
Epidemiology
Sites
Pathophysiology
  • Ultrastructural finding (see below) and eccrine glands are abundant in acral sites
    • This is because digital papillary adenocarcinoma is an eccrine tumor
Etiology
  • Some patients have trauma
Clinical features
  • Slow growing, asymptomatic nodule (average 1.7 cm, range: 0.4 - 4.3 cm)
  • Clinically confused with a cystic lesion, ulceration is rare
  • Pain may occur if the tumor involves the underlying bone, joint, nerves
  • Rarely, metastatic disease (lung, lymph node) (Am J Surg Pathol 2012;36:1883, J Am Acad Dermatol 2017;77:549)
Diagnosis
  • Clinically, may be suspected from the site of origin and clinical course
  • Histologic examination of tissue provides a definitive diagnosis
Prognostic factors
  • Local recurrence and metastasis are 5 - 21% and 26 - 50%
  • Histopathological features appear not to be predictive
  • Delayed occurrence of metastases and a protracted course, long term follow up is necessary (Am J Surg Pathol 2012;36:1883)
Case reports
Treatment
Clinical images

Images hosted on other servers:

Finger tumor with ulcer

Finger tumor

Gross images

Images hosted on other servers:

Fingertip tumor

Microscopic (histologic) description
  • Well circumscribed multinodular tumors within dermis and superficial subcutis
  • Solid and cystic portions, papillary projections, tubular or ductal structures (Am J Surg Pathol 2012;36:1883)
  • Occasionally show infiltrative margin
  • Uncommonly epidermal hyperplasia (3/31), ulceration (2/31), focal epidermal connection (1/31) (Am J Surg Pathol 2012;36:1883)
  • Tumor cells are basaloid, cuboidal to low columnar epithelial cells and myoepithelial cells with mild to moderate cytologic atypia
  • Severe cytologic atypia (3/31) and tumor necrosis (6/31) is noted in a small subset of tumors (Am J Surg Pathol 2012;36:1883)
  • Mitoses may be inconspicuous or frequent (1 - 15 mm2) (Am J Surg Pathol 2012;36:1883)
  • Spindle cell, squamoid differentiation, clear cell changes may be encountered (Am J Surg Pathol 2012;36:1883)
Microscopic (histologic) images

Contributed by Haruto Nishida, M.D., Ph.D.

Fused glands

Solid area with focal necrosis

Sheet like growth pattern

Focal squamous differentiation

Small nest and lymphatic invasion

Virtual slides

Images hosted on other servers:

Digital papillary adenocarcinoma

Positive stains
Electron microscopy description
  • Ultrastructural studies demonstrated three types of neoplastic cells: clear cells, dark cells and myoepithelial cells
  • Some dark cells have dense granules (J Cutan Pathol 1987;14:129)
Molecular / cytogenetics description
Sample pathology report
  • Skin, left ring finger, punch biopsy:
    • Digital papillary adenocarcinoma (see comment)
    • Comment: The histopathological examination shows a well circumscribed multinodular tumor within the dermis to superficial subcutis. It shows solid and cystic portions with focal papillary projections and tubular structures. Two cell layers are present; inner cells are cuboidal epithelial cells with mild to moderate cytologic atypia and outer cells are myoepithelial cells. Some mitoses are seen (1 - 3 mm2). The inner layer is immunoreactive for EMA and CEA which highlighted the luminal border of tubules. The myoepithelial cell (outer cell) layer is positive for SMA, calponin and p63. These histologic features are consistent with digital papillary adenocarcinoma.
Differential diagnosis
Board review style question #1

    A 56 year old man presented with a tumor on his middle finger. The tumor shows solid and cystic areas with focal papillary projections and tubular structures. Which of the following is most likely the correct diagnosis?

  1. Apocrine hidrocystoma
  2. Bowen disease
  3. Digital papillary adenocarcinoma
  4. Hidradenoma
  5. Papillary eccrine adenoma
Board review style answer #1
C. Digital papillary adenocarcinoma

Reference: Digital papillary adenocarcinoma

Comment Here
Board review style question #2
    Which of the following is true about digital papillary adenocarcinoma?

  1. Dual layered cystic structures are characteristic
  2. It arises only in digits
  3. It does not occur in young people
  4. Tumor does not show metastasis
Board review style answer #2
A. Dual layered cystic structures are characteristic

Reference: Digital papillary adenocarcinoma

Comment Here

Dilated pore of Winer (pending)

Eccrine angiomatous hamartoma
Definition / general
  • Rare benign hamartoma of eccrine glands and blood vessels that often occurs in the extremities (Ann Dermatol 2013;25:208)
Clinical features
  • May be present at birth or develop during childhood (Dermatology Online Journal 2009;15:10)
  • May be painful and be associated with hyperhidrosis, which can be helpful in diagnosis
  • Typically slow growing with benign behavior
Case reports
Treatment
Gross description
  • 2.0 x 1.5 cm skin covered nodule; cut section showed a homogenous dark brown appearance (Case of the Week #324)
Gross images

Case #324


Contributed by Koteeswaran Govindaswamy, M.D. and Rajasekaran Koteeswaran, M.D.

56 year old man with 3 cm nodule on dorsum of foot for 40 years

Microscopic (histologic) description
  • Increased number of eccrine glands in the superficial dermis, with blood vessels between and around the glands and ducts
  • Vessels varied in size, but were predominantly small and lined by endothelial cells
  • Other areas of dermis showed smooth muscle bundles, fat cells and focal areas of myxoid change (Case of the Week #324)
Microscopic (histologic) images

Case #324




Contributed by Koteeswaran Govindaswamy, M.D. and Rajasekaran Koteeswaran, M.D.

56 year old man with 3 cm nodule on dorsum of foot for 40 years

Differential diagnosis

Eccrine spiradenoma
Definition / general
  • Benign (multi) nodular solid tumor showing eccrine differentiation
Essential features
  • Benign neoplasm, excision is curative
  • Predilection for head and neck
  • Usually asymptomatic but can be painful, belonging to the painful skin tumors in the acronym BLEND TAN EGG (blue rubber bleb nevus, leiomyoma, eccrine spiradenoma, neuroma, dermatofibroma, tufted angioma, angiolipoma, neurilemmoma, endometrioma, granular cell tumor, glomus tumor) (Indian J Dermatol Venereol Leprol 2019;85:231)
  • Association with the Brooke-Spiegler syndrome
  • Multiple dark nests of cells confined to dermis (blue balls in the dermis) composed of 2 epithelial cell types: small dark peripheral cells and larger pale central cells, with intratumoral lymphocytes
Terminology
  • Spiradenoma
  • Eccrine spiradenoma
ICD coding
  • ICD-0: 8403/0 - eccrine spiradenoma
  • ICD-10: D23 - other benign neoplasms of skin
Epidemiology
Sites
  • Head / face and neck
  • Other sites (extremities and trunk) less common
Clinical features
  • Commonly presenting as solitary nodule
  • Rarely as multiple lesions in combination with other types of adnexal tumors as part of the Brooke-Spiegler syndrome
  • Predilection site: head and neck area, less commonly trunk and extremities
  • Usually asymptomatic, although often cited as a painful skin tumors in the acronym BLEND TAN EGG (blue rubber bleb nevus, leiomyoma, eccrine spiradenoma, neuroma, dermatofibroma, tufted angioma, angiolipoma, neurilemmoma [schwannoma], endometrioma, granular cell tumor, glomus tumor) (Indian J Dermatol Venereol Leprol 2019;85:231)
  • Firm, round and smooth surface nodule
  • Skin colored, gray-pink, reddish or blue
  • Small, usually 1 - 3 cm in size, may be larger, rare giant variants
Diagnosis
Prognostic factors
  • Benign neoplasm
  • Malignant transformation is a very rare phenomenon with the malignant component classified into basal cell adenocarcinoma-like, low grade pattern, basal cell adenocarcinoma-like, high grade pattern, invasive adenocarcinoma, NOS, sarcomatoid carcinoma / carcinosarcoma and malignant epithelial myoepithelial carcinoma (Am J Surg Pathol 2009;33:705, Int J Surg Pathol 2020;28:427)
Case reports
Treatment
  • Complete excision is curative
Clinical images

Contributed by Nicole Riddle, M.D. and Mark R. Wick, M.D.

Scalp

Missing Image

Breast skin



Images hosted on other servers:

Erythematous scalp nodule

Brooke-Spiegler
syndrome, multiple
face and
scalp nodules

Supraorbital spiradenoma with malignant transformation

Auricular nodule

Gross description
  • Usually less than 1 cm, rarely can be larger
  • Gray-pink or blue nodule
  • Either uninodular or multinodular tumor centered within the dermis
  • Extension into the subcutaneous tissue is common
  • Usually not connected to epidermis (Arch Craniofac Surg 2017;18:211)
Frozen section description
  • Not typically submitted for frozen section
  • Histologic features described below
  • Might be mistaken for basal cell carcinoma at the time of Mohs surgery (Wien Med Wochenschr 2018;168:218)
Microscopic (histologic) description
  • Circumscribed uninodular or multinodular basophilic tumor (see Figure 1)
    • Mostly centered in the dermis (blue balls)
    • Some tumors are encapsulated (see Figure 2)
  • Extension into the subcutis is common
  • 2 cell populations in the neoplastic nodules: centrally located large pale cells and small dark basaloid cells at the periphery (see Figure 3)
  • Neoplastic cells are arranged in trabecular, reticular or solid fashion
  • Intratumoral lymphocytes as an integral component of the tumor (see Figure 3)
  • Tubular / ductal structures (see Figure 4) and PAS positive basement membrane material (see Figure 5)
  • Stroma between the nests of epithelial cells is often edematous with prominent vascularity (see Figure 6); may be hyalinized (see Figure 7)
  • Vessels may be widely dilated (see Figure 7) / hemangiopericytoma-like
  • Many T lymphocytes and Langerhans cells often scattered throughout lobules
  • Mitoses extremely rare
  • Morphologic variations:
    • Architectural variations:
    • Cytologic variations:
      • Squamous metaplasia (see Figure 12)
      • Clear cell changes / metaplasia (see Figure 13)
      • Mucinous metaplasia and sebaceous differentiation
    • Stromal variation: edematous, myxoid, lipomatous metaplasia, osseous metaplasia, myoid metaplasia
Microscopic (histologic) images

Contributed by Tien-Anh Tran, M.D.

Multinodular

Encapsulated

2 cell populations and lymphocytes

Dilated ducts

Basement membrane


Fibrovascular stroma

Hyalizined stroma

Cystic change

Adenomatous changes

Adenomyoepithelioma


Cylindroma-like morphology

Squamous metaplasia

Clear cells

S100

SMA

Virtual slides

Images hosted on other servers:

Spiradenoma

Cytology description
  • Rare case reports describing the fine needle aspiration (FNA) cytologic findings of spiradenoma
  • Tight multilayered clusters of uniform benign cuboidal epithelial cells along with spindle shaped myoepithelial cells and occasional lymphocytes (J Lab Physicians 2014;6:130)
  • Prominent basement membrane deposition with an irregular tubular and nesting growth pattern and bland basaloid cells (Acta Cytol 1990;34:275)
Cytology images

Images hosted on other servers:

Cells with hyaline

Tight clusters of cells

Positive stains
Electron microscopy description
  • Lobules separated by amorphous / fibrillar material
  • Large pale cell has mitochondria, vesicles, glycogen in cytoplasm (J Invest Dermatol 1962;38:289)
Molecular / cytogenetics description
Videos

Spiradenoma: 5 minute pathology pearls

Spiradenoma and cylindroma: sweat gland tumor pathology

Spiradenoma for beginners

Sample pathology report
  • Skin, left cheek, excision:
    • Spiradenoma, completely excised with negative surgical margins
    • Negative for malignancy
Differential diagnosis
  • Cylindroma:
    • Closely related tumor with morphologic overlap but generally more abundant eosinophilic material around clusters of dark cells with jigsaw pattern and usually lack of lymphocytes
  • Poroma:
    • Only basaloid cells, absence of the second pale cell population and intratumoral lymphocytes
  • Spiradenocarcinoma / malignant spiradenoma / carcinoma ex spiradenoma:
    • Infiltrative growth and nuclear atypia, arising from spiradenoma (both components identified)
Board review style question #1

Which of the following is true about this cutaneous lesion?

  1. Mitoses are very rare
  2. Most commonly involves the extremities
  3. Typically presents as multiple nodules
  4. Usually not often painful
Board review style answer #1
A. Mitoses are very rare in spiradenoma

Comment Here

Reference: Eccrine spiradenoma
Board review style question #2
Which of the following is true of spiradenoma?

  1. Composed of nests of a single cell type, forming dark nests in the dermis
  2. Even with complete excision, local recurrence is typical
  3. Intratumoral lymphocytes are a common component
  4. Usually centered in the epidermis
Board review style answer #2
C. Intratumoral lymphocytes are a common component

Comment Here

Reference: Eccrine spiradenoma
Board review style question #3
Spiradenoma is an adnexal tumor which commonly occurs in what hereditary syndrome?

  1. Birt-Hogg-Dube syndrome
  2. Brooke-Spiegler syndrome
  3. Cowden syndrome
  4. Tuberous sclerosis
Board review style answer #3
B. Brooke-Spiegler syndrome

Comment Here

Reference: Eccrine spiradenoma

Eccrine syringofibroadenoma
Definition / general
  • Rare, benign eccrine proliferation of anastomosing cords with ductal structures set in a fibrovascular stroma, often located on the extremities
Essential features
  • Erythematous, solitary or multiple papules and nodules often on acral sites
  • Association with Schöpf-Schulz-Passarge syndrome and Clouston syndrome
  • Thin anastomosing cords and strands of basaloid monomorphous cuboidal cells extending from the basal layer of epidermis into dermis with ductal structures and set in a fibrovascular stroma
Terminology
  • Eccrine syringofibroadenoma (of Mascaro)
  • Other names include
    • Eccrine syringofibroadenomatous hyperplasia
    • Acrosyringeal adenomatosis
    • Eccrine poromatosis
    • Linear eccrine poroma
    • Acrosyringeal nevus or acrosyringeal nevus of Weedon and Lewis
    • Nevus syringoadenomatosus papilliferum
ICD coding
  • ICD-O: 8392/0 - syringofibroadenoma
  • ICD-10: D23 - other benign neoplasms of skin
  • ICD-11
    • 2F22 - benign neoplasms of epidermal appendages
    • XH06Y5 - syringofibroadenoma
Epidemiology
  • Rare
  • Wide age range, most cases presenting in the seventh to eighth decade (Am J Dermatopathol 1992;14:328)
    • Schöpf-Schulz-Passarge syndrome associated cases usually present in adolescence; slightly later presentation in Clouston syndrome
  • No clear racial or gender predilection
Sites
Pathophysiology
  • Morphologic, immunohistochemical and electron microscopic evidence supports origin from eccrine duct cells
  • Reactive subtype postulated to be induced by repair / remodeling process (An Bras Dermatol 2021;96:255)
Etiology
  • Unclear; neoplastic / tumoral, hamartomatous and reactive origins postulated
Clinical features
  • Presents as slow growing, solitary or multiple, flesh colored nodule(s)
  • 5 major clinical types (J Am Acad Dermatol 1997;36:569)
    • Solitary: most common form, often on lower extremities
    • Multiple with hidrotic ectodermal dysplasia (Schöpf-Schulz-Passarge syndrome and Clouston syndrome): palms and soles, younger patients (15 - 25 years old)
    • Multiple without associated cutaneous findings: palms and soles
    • Nevoid (i.e., nonfamilial unilateral linear eccrine syringofibroadenoma [ESFA]): rare, unilateral linear papules and plaques
    • Reactive, as can be seen in association with the following
      • Inflammatory dermatoses / processes (i.e., erosive lichen planus, uncontrolled psoriasis, bullous pemphigoid, epidermolysis bullosa, primary cutaneous amyloidosis, thermal scar, venous stasis, chronic diabetic foot ulcer, neuropathy, peristomal skin)
      • Infectious etiologies (i.e., leprosy, HPV)
      • Neoplasms / tumors (i.e., nevus sebaceus, clear cell acanthoma, acantholytic dyskeratotic acanthoma, squamous cell carcinoma, basal cell carcinoma and Merkel cell carcinoma)
  • Associated syndromes
Diagnosis
  • Biopsy / histologic examination and clinicopathologic correlation necessary for diagnosis
Prognostic factors
Case reports
Treatment
Clinical images

Images hosted on other servers:
Vegetating erythematous plaque

Vegetating erythematous plaque

Skin colored fibrotic nodule

Skin colored fibrotic nodule

Pink nodules and plaques

Pink nodules and plaques

Coalescing palmar papules

Coalescing palmar papules

Multiple irregular nodules

Multiple irregular nodules

Fleshy nodules of ESFA

Fleshy nodules of ESFA

Gross description
  • Verrucous erythematous papules, nodules and ulcerative plaques
  • Multiple nodules may be arranged in a symmetrical or unilateral nevoid fashion
Microscopic (histologic) description
  • Proliferation of thin anastomosing reticulated epithelial cords and strands often forming a lattice and extending from the epidermis into the dermis (J Cutan Aesthet Surg 2022;15:335)
  • Lesional cells are monomorphous, basaloid, cuboidal and slightly smaller than adjacent keratinocytes
  • Epithelial cords contain structures resembling eccrine ducts (Ann Dermatol 2020;32:57)
  • Background fibrovascular stroma
Microscopic (histologic) images

Contributed by Candice E. Brem, M.D.
Circumscribed papulonodular lesion

Circumscribed papulonodular lesion

Background fibrous stroma

Background fibrous stroma

Background fibrous stroma

CCA with ESFA

ductal structures

Ductal structures


epidermal based interconnected strands

Epidermal based interconnected strands

lattice-like architecture

Lattice-like architecture

basaloid cuboidal lesional cells

Basaloid cuboidal lesional cells

Positive stains
Negative stains
Electron microscopy description
  • Tumor cells with tonofilaments / desmosomes, numerous glycogen granules
  • Basal lamina
  • Globular keratohyalin granules around ducts / cysts
  • No lamellar granules
  • Poorly developed cornified cell envelopes (Am J Dermatopathol 1996;18:207)
Videos

Eccrine syringofibroadenoma: rare skin adnexal tumor / pattern

Sample pathology report
  • Leg, shave biopsy:
    • Benign appendageal neoplasm with eccrine differentiation consistent with an eccrine syringofibroadenoma (see comment)
    • Comment: The lesion extends to the deep margin. The specimen exhibits an epidermal based proliferation of monomorphous basaloid cells arranged as anastomosing cords forming a lattice with intermixed ductal structures and set in a fibrovascular stroma. These findings support the histologic diagnosis.
Differential diagnosis
Board review style question #1

A 74 year old woman presents with a 1 year history of asymptomatic, erythematous papules on the left heel. A subsequent shave biopsy is performed (see image above). Few intermixed ducts in the epithelial cords are also noted on examination of multiple tissue levels. What is the best histologic diagnosis for this lesion?

  1. Basal cell carcinoma, infiltrative type
  2. Eccrine syringofibroadenoma
  3. Porocarcinoma
  4. Poroma
  5. Squamous cell carcinoma
Board review style answer #1
B. Eccrine syringofibroadenoma (ESFA). The clinical presentation paired with the histologic features of anastomosing reticulated cords and strands of basaloid monomorphous cuboidal cells extending from the epidermis into dermis with few ductal structures set in a fibrovascular stroma are consistent with eccrine syringofibroadenoma. Answer A is incorrect because basal cell carcinoma demonstrates peripheral palisading, clefting and a fibromucinous stroma, while generally lacking intermixed ductal structures. Answer D is incorrect because poroma lacks the reticular and corded architecture of ESFA. Answer C is incorrect because porocarcinoma lacks the corded architecture and has malignant cytologic changes. Answer E is incorrect because squamous cell carcinomas often demonstrate eosinophilic to glassy keratinocytes, cytologic atypia and an infiltrative growth pattern.

Comment Here

Reference: Eccrine syringofibroadenoma
Board review style question #2
In the pediatric population, eccrine syringofibroadenoma (ESFA) has been associated with which genetic disorder?

  1. Birt-Hogg-Dubé
  2. Brooke-Spiegler
  3. Rubinstein-Taybi
  4. Schöpf-Schulz-Passarge
Board review style answer #2
D. Schöpf-Schulz-Passarge. ESFA has been recognized as a cutaneous marker of Schöpf-Schulz-Passarge syndrome. Answer C is incorrect because to date, no cases of ESFA have been seen in Rubinstein-Taybi, a syndrome usually associated with pilomatricomas, hemangiomas and keloids. Answer A is incorrect because Birt-Hogg-Dubé patients have a high frequency of developing fibrofolliculomas, trichodiscomas and lipomas. Answer B is incorrect because Brooke-Spiegler, due to a CYLD mutation, has an increased risk of developing spiradenomas, cylindromas, trichoepitheliomas and basal cell carcinomas.

Comment Here

Reference: Eccrine syringofibroadenoma

Embryonal rhabdomyosarcoma

Endocrine mucin producing sweat gland carcinoma
Definition / general
  • Low grade adenocarcinoma of cutaneous sweat gland origin
  • Predilection for the eyelids of older women
Essential features
  • Rare, low grade adenocarcinoma of sweat gland origin that often arises on or near the eyelids of older women
  • Can recur if incompletely excised and may be a precursor to mucinous carcinoma; however, there are no reported cases of metastases
  • Must be distinguished from metastatic adenocarcinoma; resembles mammary solid papillary carcinoma / endocrine ductal carcinoma with cystic and solid architecture (solid components show cribriform, papillary and pseudopapillary patterns punctuated by mucin filled spaces)
  • Key feature is neuroendocrine expression; elaboration of a myoepithelial layer by immunohistochemistry is helpful to confirm primary cutaneous origin and exclude metastatic adenocarcinoma
ICD coding
  • C44.191 (eyelid): other specified malignant neoplasm of skin of unspecified eyelid, including canthus
Epidemiology
Sites
  • Head and neck, eyelid most common
Etiology
  • Given the low incidence of this disease, its etiology is incompletely understood
  • Tumor is thought to be analogous to mammary solid papillary carcinoma / endocrine ductal carcinoma in situ of and exhibits a similar immunophenotype, including evidence of a surrounding myoepithelial cell layer
Clinical features
  • Lesions can be solitary or less often multiple (regional)
  • Translucent cyst (sometimes multiloculated) may resemble hidradenoma
  • Cases with lesser developed cystic components appear as skin colored nodules
Diagnosis
  • Diagnosis requires correlations with immunohistochemistry, clinical history and clinical examination findings to exclude metastatic adenocarcinomas from breast, salivary gland or viscera
Prognostic factors
  • No reported cases of metastasis
  • Some cases have contiguous primary cutaneous mucinous carcinoma
  • Recurrence is common when the tumor is incompletely excised
Case reports
Treatment
  • Excision or Moh's micrographic surgery
Clinical images

Images hosted on other servers:

EMPSGC on the eyelid

Microscopic (histologic) description
  • Nodular, often multilobulated dermal tumor with solid areas and cystic spaces
  • Solid areas may show cribriform architecture or pseudorosettes, papillae with fibrovascular cores or smaller pseudopapillae
  • Solid areas are often punctuated by mucin filled cystic spaces that range in appearance from small holes and clefts to large, hidrocystoma-like components
  • Some lesions closely resemble hidrocystoma on low power but show multilayering of neoplastic cells and Roman bridges analogous to breast ductal carcinoma in situ
  • Lesions frequently appear contiguous with benign ducts
  • Most lesions have an inconspicuous and discontinuous myoepithelial cell layer surrounding them; however, there is disagreement as to whether to classify endocrine mucin producing sweat gland carcinoma as purely in situ carcinoma or invasive adenocarcinoma (Am J Surg Pathol 2005;29:1330, Am J Dermatopathol. 2013;35:117, Requena: Cutaneous Adnexal Neoplasms, 1st Edition, 2018)
  • Has 1 to 3 layers of bland columnar secretory cells with occasional apical snouts, intracellular and extracellular mucin, moderate amounts of cytoplasm with a basophilic hue, ovoid nuclei (slightly larger than those of surrounding eccrine ductal epithelia), bland or sometimes stippled nuclear chromatin and minimal to no nuclear pleomorphism or mitotic activity (Am J Surg Pathol 2005;29:1330)
  • Lesions are sometimes contiguous with primary cutaneous mucinous carcinoma (or adjacent acellular pools of mucin suspicious for mucinous carcinoma) or have a component of infiltrative nests of cells with mild nucleomegaly and hyperchromasia
Microscopic (histologic) images

Contributed by Robert E. LeBlanc, M.D.

Anodular, solid and cystic appearance

Solid component with cribriform architecture

Small and monotonous columnar epithelial cells

Synaptophysin

Calponin

Comparison with
primary cutaneous
mucinous carcinoma

Positive stains
Negative stains
  • CK20
  • p63 does not stain tumor cells
Molecular / cytogenetics description
Sample pathology report
  • Skin, left upper eyelid, shave:
    • Endocrine mucin producing sweat gland carcinoma, present at the peripheral edge and base of specimen (see comment)
    • Comment: The bland cytomorphology, evidence of neuroendocrine expression (INSM1 stains tumoral nuclei and synaptophysin stains tumoral cytoplasm) and variable presence of a p63 positive myoepithelial layer surrounding the lesion together support this diagnosis.
Differential diagnosis
  • Apocrine adenoma:
    • Extensive decapitation secretion, absence of neuroendocrine differentiation, absence of mucin and enlarged epithelioid nuclei with conspicuous nucleoli
  • Apocrine carcinoma:
    • High grade cytologic atypia exceeding that of endocrine mucin producing sweat gland carcinoma (EMPSGC), usually no mucin production
  • Basal cell carcinoma:
    • Rare cases exhibit neuroendocrine differentiation; however, palisading of peripheral cells with retraction of the adjacent stroma and increased apoptoses and mitoses favor the latter
  • Dermal duct tumor:
    • Absence of neuroendocrine differentiation, no cribriform architecture or mucin filled clefts, more prominent ductular differentiation with secretory spaces lined by cuticle, foci of necrosis
  • Hidrocystoma:
    • Bland lining lacks architectural complexity, cytologic atypia or neuroendocrine marker expression
  • Nodular hidradenoma:
    • Also has variable cystic and solid conformation but an absence of neuroendocrine differentiation, attachment to the epidermis and vascular spaces favor hidradenoma and a greater diversity of cell types including eosinophilic polygonal cells, squamous cells, clear cells and goblet cells
  • Primary cutaneous mucinous adenocarcinoma:
    • May be contiguous with EMPSGC suggesting that EMPSGC could be a precursor lesion; small cribriform or tubular islands of epithelioid cells in pools of mucin partitioned by strands of dermal collagen
Additional references
Board review style question #1
Staining with which of the following markers would favor the diagnosis of metastatic adenocarcinoma over endocrine mucin producing sweat gland carcinoma?

  1. CK7
  2. CK20
  3. GATA3
  4. p63
Board review style answer #1
B. CK20 expression would raise consideration for metastatic carcinoma of colorectum. Endocrine mucin producing sweat gland carcinoma otherwise shares an immunophenotype with metastatic carcinomas of breast and salivary gland and expresses CK7 and GATA3. p63 is not expressed by the epithelial cells of EMPSGC but highlights a discontinuous myoepithelial layer surrounding the neoplasm.

Comment Here

Reference: Endocrine mucin producing sweat gland carcinoma
Board review style question #2
Which of the following is the most common site of origin for endocrine mucin producing sweat gland carcinoma?

  1. Eyelid
  2. Inferior helix
  3. Nasal ala
  4. Scalp
Board review style answer #2
A. Endocrine mucin producing sweat gland carcinoma has a predilection for the head and neck and is most often located on or around the eyelids of older adults.

Comment Here

Reference: Endocrine mucin producing sweat gland carcinoma

Endometriosis
Definition / general
  • This topic discusses features of cutaneous endometriosis only - a general discussion of endometriosis is found in the Ovary topic
  • Ectopic endometrial tissue, usually described in pelvic organs, but 0.5% - 1.0% of cases are cutaneous
  • Women of reproductive age, often due to iatrogenic seeding of umbilicus during C-section (Villar’s nodule) but may occur in almost any site
  • Carcinoma rarely arises in preexisting endometriosis
  • Women with endometriosis may have mildly increased risk of melanoma (Int J Epidemiol 2009;38:1143)
Case reports
Treatment
  • Gonadotropin releasing hormone analogues
  • Excision
Gross description
  • Dark red to bluish nodule, may form cystic structure
Gross images

Images hosted on other servers:

Villar nodule

Microscopic (histologic) description
  • Endometrial glands, endometrial stroma and hemorrhage
  • May have marked decidual changes
Microscopic (histologic) images

Images hosted on other servers:

Endometriosis in periumbilical dermis

Various images

Positive stains
Negative stains
Differential diagnosis
  • Sweat gland tumor
  • Urachal duct cyst
  • Pyogenic granuloma
  • Hernia
  • Residual embryonic tissue
  • Primary or metastatic adenocarcinoma (Sister Joseph's nodule)
  • Cutaneous endosalpingiosis

Epidermal (epidermoid) type
Definition / general
  • Benign skin tumor
  • Cystic mass containing keratin
Essential features
  • Cystic mass with soft white keratin contents
  • Histologically cystic mass with squamous epithelium and keratin flakes
Terminology
  • Epidermoid cyst, epidermal cyst, epidermal inclusion cyst, infundibular cyst
ICD coding
  • ICD-10: L72.0 - epidermal cyst
Epidemiology
Sites
  • Face, neck, trunk, perineal area, cerebellopontine angle
  • Less commonly spine, intrapancreatic accessory spleen
Pathophysiology
  • Follicular orifice becomes plugged with bacteria and keratin, leading to cystic dilation and entrapment of keratin debris 
  • Presence of multiple epidermal inclusion cysts has been documented in Gardner syndrome, a variant of familial adenomatous polyposis with benign osteomas and intestinal fibromatoses
  • Less frequently, patients may have lipomas, pilomatrixomas (including epidermoid cysts with pilomatrical lining) or leiomyomas
  • Multiple and large epidermoid cysts may occur with the use immunosuppressants in the posttransplantation setting, for example, with cyclosporine or tacrolimus (Cutis 1992;50:36Ann Dermatol 2011;23:S182)
  • May complicate penetrating trauma to skin, such as a sewing needle, with resultant implantation of squamous epithelium into the dermis (Turk J Pediatr 2011;53:108)
Diagrams / tables

Images hosted on other servers:

Age distribution

Site distribution

Clinical features
  • Present as smooth dome shaped swellings varying in size from a few millimeters to a few centimeters (Ann Dermatol 2017;29:33)
  • Usually occur on the face, neck or trunk but can occur anywhere
  • Overlying skin may be taut with the pressure of the cyst and have a central punctum
  • Generally occurs in postpubertal individuals
  • Freely mobile unless ruptured, in which case a foreign body giant cell reaction may make them more adherent to surrounding connective tissue
  • May become painful and inflamed with external manipulation
  • Cyst contents white, cheesy macerated keratin that may have an odor
Diagnosis
  • Often based on clinical examination
  • Pathological diagnosis relies on recognizing the cyst wall and contents
  • In cases of extensive foreign body giant cell reaction or fibrosis, the classic features may not be visualized as readily and a diligent search for keratin flakes may lead to diagnosis
Laboratory
  • No specific laboratory findings
Radiology description
  • Ultrasound:
    • Subcutaneous rounded structure that is mostly anechoic or hypoechoic with focal present inner echoes (pseudotestis appearance) representing cystic debris
  • MRI:
    • Fluid-like enhancement
    • High intensity of cyst contents on T2 weighted images and peripheral cyst wall enhancement with T1 gadolinium enhancement
    • Cyst rupture results in irregular enhancement (AJR Am J Roentgenol 2006;186:961)
Radiology images

Images hosted on other servers:

Lumbar spine epidermoid cyst

Abdominal epidermal inclusion cyst

Case reports
Treatment
  • Complete excision of the cyst is curative
Clinical images

Contributed by Jeremy Hugh, M.D.

White subcutaneous nodule

White nodule in hair bearing area



Images hosted on other servers:

Cheesy contents of
cystectomy in
mastoid region after
penetrating trauma

Gross description
  • Pearly glistening cyst with creamy contents
Gross images

Contributed by the University of Colorado Department of Pathology

Disrupted cyst wall and adjacent soft tissue

Microscopic (histologic) description
Microscopic (histologic) images

Contributed by the University of Colorado Department of Pathology, Jijgee Munkhdelger, M.D., Ph.D. and Andrey Bychkov, M.D., Ph.D.

Loose keratin flakes

Stratified squamous epithelium lining

Ruptured epidermoid cyst

Giant cells, cholesterol clefts, mixed inflammation


Foreign body giant cell reaction

Epidermal inclusion cyst in cerebellopontine angle

Lamellated
keratin flakes in
cranial epidermal
inclusion cyst

Unilocular dermal cyst

Cytology description
  • Anucleate keratinizing squamous cells with some nucleated squamous cells
  • Frequent erythrocytes, leukocytes, multinucleated giant cells and cholesterol crystals (J Nat Sci Biol Med 2014;5:460)
Cytology images

Contributed by the University of Colorado Department of Pathology

Anucleate squamous cells

Keratinizing squamous cells

Anucleate squames and inflammatory cells

Sample pathology report
  • Skin, neck, excision:
    • Epidermal inclusion cyst
    • Microscopic description: Cyst lined by squamous epithelium with granular layer containing lamellated keratin.
Differential diagnosis
  • Proliferating epidermoid cyst:
    • May show carcinomatous changes, invasion and be locally aggressive
  • Trichilemmal (pilar) cyst:
    • Lack a granular layer in the cyst lining
    • Dense lamellated keratin cyst contents
    • More likely to occur on the scalp
  • Pilomatricoma:
    • Eosinophilic cellular outlines of squamous cells ("ghost cells") in addition to the more basophilic matrical cells
    • More likely to occur in a pediatric population
  • Hybrid cyst:
  • Dermoid cyst:
    • Similar in appearance
    • Has adnexal structures (ie. sebaceous glands) in cyst wall
    • Arises at sites of embryonic closure such as the lateral eyebrow
  • Pilonidal sinus:
    • Sinus tract surrounded by epithelium and mixed inflammation
    • Characteristically contains broken hair shafts
  • Steatocystoma:
    • Compressed sebaceous gland within the cyst wall
    • Characteristic wavy eosinophilic crenulated cuticle of the lining
  • Odontogenic keratocyst:
    • Attenuated squamous epithelium with parakeratosis
    • Retraction of epithelium with basal palisade can be a helpful finding
    • Often filled with keratin debris like epidermoid cyst
Board review style question #1
An elderly man has a soft, round subcutaneous mass on the back of the neck with a central punctum. Which of the following is the most likely diagnosis?

  1. Angiosarcoma
  2. Epidermoid cyst
  3. Lymphadenitis
  4. Pilar cyst
  5. Spindle cell lipoma
Board review style answer #1
B. Epidermoid cyst

Comment Here

Reference: Epidermal (epidermoid) cyst
Board review style question #2

The above image is a section of a subcutaneous mass removed from the axilla of a 40 year old woman. What is the most likely diagnosis?

  1. Dermoid cyst
  2. Dilated pore of Weiner
  3. Epidermoid cyst
  4. Hailey-Hailey
  5. Trichilemmal cyst
Board review style answer #2
C. Epidermoid cyst

Comment Here

Reference: Epidermal (epidermoid) cyst

Epidermal nevus
Definition / general
  • Epidermal nevus is a developmental malformation that involves the epidermis and clinically manifests as a discrete congenital linear patch or plaque along the Blaschko lines
  • Histologically characterized by a hamartomatous keratinocyte proliferation that results in epidermal papillomatosis and acanthosis with no associated adnexal structures
  • Here, the term epidermal nevus is discussed within a narrow definition and does not include organoid, sebaceous, eccrine and pilar nevi
  • Reference: Calonje: McKee's Pathology of the Skin, 5th Edition, 2019
Essential features
  • Hamartomatous proliferation of the epidermis with no associated adnexal structures, commonly presenting at birth
  • Clinically presents as single to multiple coalescing small pale to dark brown plaques, occasionally markedly papillomatous or hyperkeratotic in a Blaschko linear distribution
  • It is estimated that 33% of patients with epidermal nevi have other organ involvement and occurrence is associated with multiple syndromes
  • Commonly caused by mosaic postzygotic activating mutations in FGFR2 / FGFR3, PIK3CA or RAS genes in the epidermis, occurring in early embryonic development
  • Development of malignant neoplasms in epidermal nevi is a rare phenomenon and almost always occurs during adulthood
Terminology
  • Epithelial nevus
    • Essentially a synonym for epidermal nevus; though both terms are used frequently to cover hamartomas of the epidermis along with those with adnexal malformations, this page uses the term epidermal nevus to discuss nevi arising from the epidermal keratinocytes only
  • Common keratinocyte nevus
  • Linear epidermal nevus (Am J Dermatopathol 2014;36:430)
  • Linear verrucous epidermal nevus
  • Nonorganoid epidermal nevus (Dermatol Online J 2010;16:12)
ICD coding
  • ICD-11
    • LC00.0 - epidermal nevus
    • LC02 - complex epidermal hamartoma
Epidemiology
  • Typically present at birth or develops in early childhood, with a prevalence of ~1 in 1,000 live newborns (Pediatr Dermatol 2004;21:432)
  • F = M
  • Secondary tumors are infrequent compared to organoid epidermal nevi (Dermatol Pract Concept 2022;12:e2022087)
  • Extremely low rate of malignant transformation (Clin Exp Dermatol 2019;44:238)
  • Can be associated with the following syndromes
    • Schimmelpenning-Feuerstein-Mims syndrome (organoid nevi, abnormalities of the central nervous system and eyes, oral lesions and skeletal defects)
    • Phakomatosis pigmentokeratotica type I (capillary malformations and epidermal nevus)
    • Nevus comedonicus syndrome (ocular, skeletal and neural abnormalities, ipsilateral congenital cataract and malformations of fingers and toes) (Pediatr Dermatol 2021;38:359)
    • Angora hair nevus syndrome (Angora hair nevus ocular, skeletal and neural abnormalities)
    • Becker nevus syndrome (Becker nevus, poor unilateral breast or nipple development, supernumerary nipples, loss of subcutaneous fat, loss of axillary hair, musculoskeletal abnormalities)
    • Proteus syndrome (overgrowth of skin and connective tissue)
    • Type II segmental Cowden disease (neural defects, overgrowth of limbs and toes, polyps and nevi)
    • Fibroblast growth factor receptor 3 epidermal nevus syndrome (keratinocytic epidermal nevus, acanthosis nigricans and neurological abnormalities) (J Pediatr Neurosci 2014;9:66)
    • CHILD syndrome (congenital hypoplasia with ichthyosiform nevus and limb defects)
  • Associated with several other less well defined epidermal nevus syndromes
  • Reference: DermNet: Epidermal Naevus [Accessed 28 July 2023]
Sites
  • Nearly all lesions occur on the neck, trunk and extremities
  • Rare sites of presentation include intraoral and the face
Pathophysiology
  • Epidermal nevi typically result from a postzygotic somatic mutation in an embryotic cell
  • This leads to genetic mosaicism and the phenotypic consequence of the somatic mutation depends on how early the mutation arises, with very early mutations leading to extensive epidermal nevi and organ involvement (Dermatol Online J 2010;16:12)
Clinical features
  • Epidermal nevi exhibit diverse clinical patterns, which can be attributed to a variety of histological patterns
  • Epidermal nevi typically present as multiple warty brown patches or plaques along the lines of Blaschko
  • Epidermal nevi can form a linear or zosteriform lesion
  • At puberty, lesions become thicker, more verrucous and hyperpigmented (Curr Derm Rep 2012;1:186)
  • Terms for different clinical patterns
    • Nevus verrucosus (verrucous epidermal nevus): localized wart-like lesions
    • Nevus unius lateris: unilateral, long, linear lesions on the extremities
    • Ichthyosis hystrix: large, bilateral nevi on the trunk
  • En bloc movement on the wobble test (characteristic of an epidermal growth)
  • Patches of discoloration with minimal scale as a presentation have been described
  • Epidermal nevi measure a few millimeters to several centimeters in length
  • Reference: Calonje: McKee's Pathology of the Skin, 5th Edition, 2019
Diagnosis
  • Usually clinically suspected and diagnosed
  • Skin biopsy may be required for confirmation or assessment of neoplasms developing in the lesion
Prognostic factors
Case reports
Treatment
  • Topical treatments (i.e., retinoic acid or 5-fluorouracil) are used to improve the cosmetic appearance of epidermal nevi by removing the keratotic surface
  • Smaller lesions can be surgically excised
  • Larger lesions can be treated by laser or cryotherapy
  • Pulsed erbium: yttrium aluminium garnet (YAG) laser is effective and has a low incidence of scarring (Dermatol Surg 2004;30:378)
  • Reference: Calonje: McKee's Pathology of the Skin, 5th Edition, 2019
Clinical images

Images hosted on other servers
Verrucous epidermal nevus Verrucous epidermal nevus

Verrucous epidermal nevus

Nevus unius lateris

Nevus unius lateris

Keratinocytic epidermal nevus

Keratinocytic epidermal nevus

Oral linear epidermal nevus

Oral linear epidermal nevus

Epidermal nevus

Epidermal nevus

Gross description
  • Well defined, elevated, tan verrucous linear plaque
Microscopic (histologic) description
  • Epidermal nevus can show a spectrum of histological patterns with occasionally more than 1 pattern existing in a single lesion; the patterns are described below
  • Common keratinocyte pattern (> 60% of cases)
    • Hyperkeratosis (occasionally columns of parakeratosis)
    • Irregular acanthosis
    • Broad papillomatosis with flat vertical peaks
    • Sharply demarcated from the adjacent normal epidermis
    • Occasional focal thickening of the granular layer
    • Hyperpigmentation of the basal keratinocytes
    • Uncommonly has associated mild inflammation or perivascular lymphocytes
  • Seborrheic keratosis-like pattern
    • Hyperkeratosis
    • Acanthosis and papillomatosis (with flattening of the lower border, Mesa sign)
    • Horn pseudocysts
  • Verrucoid pattern
    • Hyperkeratosis (orthokeratosis and parakeratosis)
    • Vacuolated cells in the granular and upper spinous layer
    • Increased keratohyaline granules
    • No prominent vascularity in the papillary dermis (typical of verruca vulgaris)
  • Acrokeratosis verruciformis pattern
    • Marked hyperkeratosis
    • Acanthosis and papillomatosis with peaks resembling church spires
    • Thickened granular layer
  • Porokeratotic pattern
    • Orthokeratosis and acanthosis are less prominent that other variants of epidermal nevus
    • Parakeratotic columns (with focal occasional vacuolated keratinocytes below)
    • Cornoid lamellae (with a focal absent granular layer below)
  • Focal acantholytic dyskeratotic Darier disease-like pattern
    • Hyperkeratosis
    • Acanthosis and papillomatosis
    • Acantholytic dyskeratotic cells (corps ronds) and acantholytic basophilic keratinocytes (grains) in the granular layer and spinous layers
    • Scattered acantholytic spinous cells above suprabasal clefts
  • Acanthosis nigricans-like pattern (Australas J Dermatol 1983;24:130)
    • Hyperkeratosis
    • Papillomatosis
    • Mild irregular acanthosis with poorly developed rete ridges
  • Hailey-Hailey disease-like pattern (Br J Dermatol 1985;112:349)
    • Mild hyperkeratosis
    • Papillomatosis
    • Suprabasal acantholysis, similar to a dilapidated brick wall
    • Rare dyskeratotic cells
  • Incontinentia pigmenti-like (verrucous phase) pattern (Pediatr Dermatol 1985;3:69)
    • Hyperkeratosis (vertical parakeratosis)
    • Acanthosis and papillomatosis
    • Thickened granular layer
    • Many individual dyskeratotic cells within the epidermis
  • Epidermolytic hyperkeratosis pattern (Australas J Dermatol 1983;24:130)
    • Hyperkeratosis
    • Acanthosis and papillomatosis
    • Perinuclear vacuolization
    • Increased irregularly shaped keratohyaline granules
    • Indistinct cell borders
  • Inflammatory linear verrucous epidermal nevus (ILVEN), epidermal nevus with skyline basal cell layer (PENS) and nevus comedonicus are regarded as distinct entities, although they are sometimes included as histological patterns of epidermal nevus
  • Reference: Am J Dermatopathol 1982;4:161
Microscopic (histologic) images

Contributed by Ronan Knittel, M.D.
Basal hyperpigmentation Basal hyperpigmentation

Basal hyperpigmentation

Acanthosis Acanthosis

Acanthosis

Hyperkeratosis Hyperkeratosis

Hyperkeratosis


Horn pseudocysts Horn pseudocysts

Horn pseudocysts

Papillomatosis Papillomatosis

Papillomatosis

Adjacent normal epidermis Adjacent normal epidermis

Adjacent normal epidermis


Flattened epidermal base

Flattened epidermal base

Virtual slides

Images hosted on other servers
Epidermal nevus.

Epidermal nevus

Molecular / cytogenetics description
  • Typically not required for diagnosis
  • 40% of epidermal nevi harbor a postzygotic activating mutation in FGFR3 and PIK3CA
  • Another 40% harbor a RAS (i.e., KRAS, NRAS, HRAS) postzygotic activating mutation (typically HRAS G13R)
  • Another 5 - 10% harbor an embryonic postzygotic FGFR2 activating mutation (J Invest Dermatol 2016;136:1718)
  • Missense mutation c.109G>T in PTCH1 gene was detected in a patient with epidermal nevi and cerebral infarction (J Med Case Rep 2022;16:343)
Sample pathology report
  • Skin, neck, excision:
    • Epidermal nevus (see comment)
    • Comment: Histological examination demonstrates skin to the subcutis with a lesion comprising an acanthotic and papillomatous epidermis with overlying hyperkeratosis. The underlying dermis appears unremarkable with no abnormal pilosebaceous units identified. There is no evidence of an associated neoplastic growth or malignancy in the material examined.
Differential diagnosis
  • Nevus sebaceus (of Jadassohn):
    • Clinically often presents as a single yellow patch of alopecia on the scalp
    • Histologically similar epidermal changes, though with additional sebaceous gland alterations and ectopic dilated apocrine glands
  • Seborrheic keratosis:
    • Unusual diagnosis in children / young adults
    • Histologically similar, though more likely to have pseudohorn cysts, squamous eddies and compact basaloid cells
  • Verruca vulgaris:
    • Papillomatosis, hypergranulosis and hyperparakeratosis
    • Elongated rete ridges with inward bending at the edge of the lesion
    • Intracorneal hemorrhage and ectatic capillaries
    • Koilocytes may be seen
  • Acanthosis nigricans:
    • Usually found in flexural areas of the body, particularly the axilla
    • Associated with an underlying metabolic disorder or malignancy
    • Usually, lesser degrees of hyperkeratosis and papillomatosis
  • Inflammatory linear verrucous epidermal nevus:
    • Alternating orthokeratosis and parakeratosis
    • Psoriasiform epidermal hyperplasia
    • Prominent granular layer underlies the orthokeratosis while the epidermis under the parakeratosis lacks a granular layer
    • Mild perivascular lymphocytic infiltrate in the upper dermis
  • Nevus with skyline basal cell layer (PENS):
    • Mild orthohyperkeratosis, slight papillomatosis and broad acanthosis
    • Distinctly palisaded basilar layer with a widened space separating this layer from the overlying spinous layer keratinocytes
  • Confluent and reticulated papillomatosis (CARP) (of Gougerot and Carteaud):
    • Clinically confluent patches or plaques centrally with a reticular pattern peripherally around the upper trunk, neck and axillae
    • Usually, lesser degrees of hyperkeratosis and papillomatosis
    • Acanthosis is present in the base of the papillae
Board review style question #1

A 3 month old boy presents with a linear plaque on the neck along the lines of Blaschko. The image above is seen on histological examination. What is the most likely diagnosis?

  1. Acanthosis nigricans
  2. Epidermal nevus
  3. Nevus sebaceus (of Jadassohn)
  4. Seborrheic keratosis
  5. Verruca vulgaris
Board review style answer #1
B. Epidermal nevus. This clinical history and histology is classic for an epidermal nevus. Answer D is incorrect because though the histology raises the possibility of seborrheic keratosis, the clinical history rules it out. Answer A is incorrect because the clinical history rules out acanthosis nigricans. Answer C is incorrect because the histology shows there are no abnormal adnexal structures. Answer E is incorrect because there is no hemorrhage, parakeratosis or prominent vascularity in the papillary dermis.

Comment Here

Reference: Epidermal nevus
Board review style question #2
What is the most likely molecular alteration in an epidermal nevus?

  1. Germline activating mutation in FGFR3
  2. Germline inactivating mutation in FGFR3
  3. Postzygotic activating mutation in FGFR3
  4. Postzygotic inactivating mutation in FGFR3
  5. Somatic activating mutation in FGFR3 after birth
Board review style answer #2
C. Postzygotic activating mutation in FGFR3 is one of the most common molecular alterations observed in epidermal nevus. Answer D is incorrect because an inactivating mutation of FGFR3 would potentially have a different consequence. Answer A is incorrect because epidermal nevi result from postzygotic and not germline mutations in FGFR3 gene. Answer B is incorrect because epidermal nevi result from postzygotic (and not germline) and activating mutations in FGFR3 gene. Answer E is incorrect because an epidermal nevus essentially arises from a postzygotic activating mutation in FGFR3 gene.

Comment Here

Reference: Epidermal nevus

Epidermolytic acanthoma
Definition / general
  • Cutaneous acanthotic benign lesion with epidermolytic hyperkeratosis spanning more than 50% of its surface
Essential features
  • Solitary or multiple lesions with acanthosis and epidermolytic hyperkeratosis
  • Epidermolytic hyperkeratosis spanning > 50% of the lesion surface
ICD coding
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
  • ICD-11: 2F21.Y - other specified benign keratinocytic acanthomas
Epidemiology
Sites
  • Extragenital (67% of cases): head and neck, upper limbs, trunk or back, lower limbs (uncommon), oral (uncommon)
  • Genital (33% of cases): vulvar, penile, perianal, perineal, scrotal
Pathophysiology
Clinical features
  • Most frequently a single lesion but may be multiple (5% of instances) (J Cutan Pathol 2019;46:305)
  • Erythematous, skin colored or brown papules or nodules with scale
  • Multiple epidermolytic acanthomas are more likely to be found on genital sites
  • May be pruritic
  • Dermoscopy may show pearly white areas, irregular pigmented grooves and peripheral pigmented radial streak-like areas (J Am Acad Dermatol 2017;77:e37)
Diagnosis
  • H&E stain with light microscopy
Prognostic factors
  • Benign
Case reports
Treatment
Clinical images

Contributed by Simon F. Roy, M.D. and Jennifer M. McNiff, M.D.
Solitary epidermolytic acanthoma

Solitary epidermolytic acanthoma



Images hosted on other servers:
Multiple epidermolytic acanthomas on scrotum

Multiple epidermolytic acanthomas on scrotum

Gross description
Microscopic (histologic) description
Microscopic (histologic) images

Contributed by Simon F. Roy, M.D. and Jennifer M. McNiff, M.D.
Papillomatous pattern of epidermolytic acanthoma Papillomatous pattern of epidermolytic acanthoma

Papillomatous pattern

Cup-like / endophytic pattern Cup-like / endophytic pattern

Cup-like / endophytic pattern

Positive stains
  • Not usually performed
Videos

Epidermolytic hyperkeratosis

Sample pathology report
  • Skin, left forearm, punch biopsy:
    • Epidermolytic acanthoma
Differential diagnosis
  • Seborrheic keratosis:
    • Acanthosis and papillomatosis but no epidermolytic hyperkeratosis
    • Focal incidental epidermolytic hyperkeratosis may be present
      • Still classified as seborrheic keratosis if covering < 50% of the surface
  • Verruca or condyloma:
    • Hypergranulosis and finely speckled keratohyaline granules
    • No lace-like reticular degeneration
    • No dense cytoplasmic eosinophilia
    • HPV induced
  • Incidental epidermolytic hyperkeratosis in another acanthotic lesion:
    • Epidermolytic hyperkeratosis is found spanning < 50% of the lesion surface
  • Epidermolytic ichthyosis (formerly bullous congenital icthyosiform erythroderma):
    • Generalized desquamating plaques rather than a solitary lesion
    • Hereditary ichthyosiform condition due to alterations in keratin genes
    • Overlapping histology with solitary epidermolytic acanthoma, clinicopathologic correlation is crucial
  • Clear cell acanthoma:
    • Pale epithelial cells with acanthosis but no epidermolytic hyperkeratosis
  • Solitary keratosis with hypergranulotic dyscornification:
    • Dense keratohyaline granules rather than finely speckled
    • Both may display eosinophilic cytoplasm but perinuclear vacuolization is more characteristic of epidermolytic acanthoma
    • Hypergranulotic dyscornification may be a focal incidental finding in other lesions
Board review style question #1


What is the best diagnosis based on the images above, considering this is a solitary lesion?

  1. Clear cell acanthoma
  2. Epidermolytic acanthoma
  3. Epidermolytic ichthyosis
  4. Seborrheic keratosis
Board review style answer #1
B. Epidermolytic acanthoma. Epidermolytic acanthoma is an acanthotic benign lesion that displays epidermolytic hyperkeratosis on more than 50% of its surface. Clear cell acanthoma and seborrheic keratosis do not show epidermolytic hyperkeratosis on their surface. Epidermolytic ichthyosis does not present as a solitary lesion but rather as a hereditary generalized desquamative eruption.

Comment Here

Reference: Epidermolytic acanthoma
Board review style question #2

What is the histopathological epidermal reaction pattern seen in the image above?

  1. Acantholysis
  2. Dyskeratotic acantholysis
  3. Epidermal necrosis
  4. Epidermolytic hyperkeratosis
Board review style answer #2
D. Epidermolytic hyperkeratosis. Epidermolytic hyperkeratosis is the epidermal reaction pattern displayed here, with reticular (lace-like) clearing of the epidermis, dense eosinophilia of the keratinocytes and coarse keratohyalin granules. Acantholysis is rather a rounding of keratinocytes with detached intercellular attachments (desmosomes). Dyskeratotic acantholysis shows acantholysis in addition to dense eosinophilia of keratinocytes but no reticular degeneration of finely speckled keratohyalin granules.

Comment Here

Reference: Epidermolytic acanthoma

Epithelial sheath neuroma (pending)
[Pending]

Epithelioid hemangioendothelioma

Extramammary Paget disease
Definition / general
  • May originate from intraepidermal portion of sweat glands or primitive basal cells with ability to differentiation towards glandular elements
  • Labia majora, scrotum and perineum are most common sites
  • Due to underlying carcinoma in 10 - 20% of cases of vulvar disease
Case reports
Treatment
  • Complete surgical excision
Clinical images

Contributed by Mark R. Wick, M.D.


Images hosted on other servers:
Red firm mass

Red firm mass

Gross description
  • Erythematous, eczematous or ring shaped
  • Often multicentric, extensive, pigmented
Microscopic (histologic) description
  • Malignant cells in epidermis with differentiation towards local glandular structures
  • Almost always simultaneous involvement of eccrine glands or hair follicles
  • Dermal invasion is rare in vulva, more common in perianal region)
  • Single, clusters or glandular formations of large cells with pale, vacuolated cytoplasm, usually just above basal layer of epidermis
  • May have cleft-like spaces between Paget cells and neighboring keratinocytes
  • No intercellular bridges, no dyskeratosis
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D.
Breast

Breast

Vulva

Vulva

Extramammary Extramammary Extramammary Extramammary

Extramammary

Extramammary Extramammary Extramammary

Extramammary

Dermal invasion Dermal invasion

Dermal invasion

GCDFP15

GCDFP15


PAS PAS

PAS



Images hosted on other servers:
Site unspecified

Site unspecified

Paget cells

Paget cells

GCDFP-15, CK7, CK20

GCDFP-15, CK7, CK20

Positive stains
Electron microscopy description
  • Cells with glandular differentiation, not melanocytes or keratinocytes
Differential diagnosis

Extramammary Paget disease

Extranodal NK / T cell lymphoma

Fibrofolliculoma / trichodiscoma
Definition / general
  • Considered a hamartoma of hair follicle

Birt-Hogg-Dubé syndrome
  • Folliculofibroma associated with Birt-Hogg-Dubé (BHD) syndrome
  • Autosomal dominant, multiple folliculomas on head and neck, acrocordons and trichodiscomas; also renal cell carcinoma [various types], renal oncocytoma and oncocytic hybrid tumors, lung cysts and spontaneous pneumothorax)
  • Human gene at 17p11.2 encodes folliculin, normally expressed in skin and adnexae (Mod Pathol 2004;17:998)
  • Frameshift mutations occur in BHD causing premature protein termination
Case reports
Clinical images

Images hosted on other servers:

Various images

Gross description
  • Multiple skin-colored papules
Microscopic (histologic) description
  • Thin epidermal strands originating from a central hair follicle with prominent connective tissue
Microscopic (histologic) images

Images hosted on other servers:

Various images

Differential diagnosis

Glomus tumor

Granular cell tumor
Definition / general
  • Dermal or subcutaneous tumor with abundant granular cytoplasm and Schwannian differentiation
  • Nonneural variant (primitive polypoid granular cell tumor) shows similar granular cytomorphology but does not have immunohistochemical evidence of Schwannian differentiation
Essential features
  • A predominantly dermal based tumor composed of cells with abundant eosinophilic to basophilic granular cytoplasm
  • Often have an infiltrative configuration
  • Tumors demonstrate Schwannian differentiation (S100+, SOX10+)
  • Epidermis can exhibit pseudocarcinomatous hyperplasia in a subset of cases
Terminology
ICD coding
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
Epidemiology
Sites
  • Common sites are trunk, upper limbs and head / neck
  • May also arise on feet, hands, anogenital region and breast
Pathophysiology
  • Dysregulation of endosomal pH due to inactivation of accessory proteins of the vacuolar H+ ATPase can give rise to conventional granular cell tumors (Nat Commun 2018;9:3533)
  • A subset of nonneural granular cell tumors (S100-) harbor ALK gene fusions (Am J Surg Pathol 2018;42:1133)
Etiology
Clinical features
Diagnosis
  • Fanberg-Smith et al. proposed diagnostic criteria for atypical and malignant tumors (Am J Surg Pathol 1998;22:779):
    • Increased N:C ratio
    • Nuclear pleomorphism
    • Necrosis, tumor type
    • Spindling of tumor cells
    • Vesicular nuclei with prominent nucleoli
    • > 2 mitoses per 10 high power fields
    • Atypical: 1 - 2 features
    • Malignant: > 2 features
  • Nasser et al. proposed diagnostic criteria for granular cell tumor of uncertain malignant potential and malignancy (Pathol Res Pract 2011;207:164):
    • Necrosis, single cell or en masse
    • > 2 mitoses per 10 high power fields
    • Uncertain malignant potential: 1 - 2 features
    • Malignant: evidence of metastatic disease
Case reports
Treatment
Clinical images

Images hosted on other servers:

Pedunculated buttock lesion

Subcutaneous waist lesion

Soft tissue swelling on the back

Hyperchromic nodule on right cubital fossa

Gross description
  • Grayish white to pale yellow
  • Oftentimes, not well circumscribed
Gross images

Images hosted on other servers:

Rough surface, ulceration and necrosis

Lobulated, solid leg mass

Infiltrative paraspinal tumor

Microscopic (histologic) description
  • Infiltrative or circumscribed architecture
  • Can involve the subcutis
  • Large polygonal cells with abundant eosinophilic granular cytoplasm and small, central nuclei
  • Epidermis can show pseudocarcinomatous hyperplasia
  • Lysosomal macroinclusions (pustulo-ovoid bodies of Milian) are usually present (J Cutan Pathol 2007;34:405)
  • Can exhibit accentuation around arrector pili muscles or peripheral nerves (J Clin Pathol 2014;67:19)
  • Nonneural granular cell tumors (S100-) can exhibit nucleomegaly, pleomorphism and variable mitotic activity (Am J Surg Pathol 1991;15:48, Histopathology 2005;47:179)
Microscopic (histologic) images

Contributed by Jarish Cohen, M.D., Ph.D.

Dermal wedge shaped tumor

Nests and cords

Pustulo-ovoid bodies

Infiltrative array

Accentuation around arrector pili


Abundant granular cytoplasm

SOX10

Deep dermal tumor

Tumor surrounds nerve

Granular pink cytoplasm


Nonneural granular cell tumor

Nonneural granular cell tumor


Atypical granular cell tumor

Positive stains
Negative stains
Electron microscopy description
Molecular / cytogenetics description
Videos

Granular cell tumor

Sample pathology report
  • Skin, left antecubital fossa, shave biopsy:
    • Granular cell tumor (granular cell schwannoma) (see comment)
    • Comment: The lesional cells are positive with an immunostain for SOX10 protein, a finding that is typical of this entity.

  • Skin, left thigh, shave biopsy:
    • Nonneural granular cell tumor (see comment)
    • Comment: The biopsy shows a polypoid mass of large, oval cells with ample pallid and both finely and coarsely granular cytoplasm, with some pustulo-ovoid bodies. The lesional cells are S100 negative, MITF negative and NKI-C3 strongly positive. An ALK immunostain shows moderate patchy positivity. A phosphohistone H3 shows that cells in mitosis are exceedingly rare, under 1/mm2. The histopathologic and immunohistochemical findings are consistent with nonneural granular cell tumor (also known as primitive polypoid granular cell tumor).
Differential diagnosis
Board review style question #1
Missing Image Missing Image


    A 40 year old woman presents with a nodule on her trunk. The lesion is biopsied and shows a dermal based proliferation of cells with abundant eosinophilic granular cytoplasm. Which of the following statements is true?

  1. The abundant granular cytoplasm is indicative of numerous membrane bound cytoplasmic granules containing aggregates of mitochondria
  2. An S100 or SOX10 stain is negative in most tumors
  3. Perineural accentuation of cellularity is a feature of malignancy
  4. These tumors can display infiltrative architecture and can sometimes involve the subcutaneous fat
Board review style answer #1
D. These tumors can display infiltrative architecture and can sometimes involve the subcutaneous fat. This statement is true since granular cell tumors often show infiltrative borders and can occasionally involve the subcutis. No studies have demonstrated an increased aggressive behavior of granular cell tumors that exhibit these features.

Comment Here

Reference: Granular cell tumor
Board review style question #2
    Which of the following statements about granular cell tumors is true?

  1. A subset are negative for S100 and other neural markers and can have a polypoid appearance
  2. Atypical features include association with pseudocarcinomatous hyperplasia and accentuation around peripheral nerves
  3. NF1 is commonly mutated in conventional granular cell tumors
  4. Tumors never demonstrate metastatic potential
Board review style answer #2
A. A subset are negative for S100 and other neural markers and can have a polypoid appearance. These so called nonneural granular cells tumors are negative for neural markers (e.g., S100 and SOX10). In addition, a subset has been shown to harbor ALK gene fusions and are positive by ALK immunohistochemistry.

Comment Here

Reference: Granular cell tumor

HPV associated SIL

HPV independent SIL

Hair follicle nevus
Definition / general
  • Rare, congenital, hamartomatous proliferation of mature vellus hairs
  • Also known as congenital vellus hamartoma
Clinical features
  • Usually present at birth or early childhood
  • Solitary skin colored papule, less than 1 cm in greatest dimension
  • Sometimes with hypertrichosis
  • Most commonly on head and neck
  • Rarely multiple or arranged in a linear distribution, following the lines of Blaschko (J Am Acad Dermatol 2002;46:S125)
Case reports
Microscopic (histologic) description
  • Increased numbers of closely situated mature vellus hair follicles
  • Some are associated with small sebaceous glands or have thickened perifollicular fibrous sheaths
  • Follicles may be located abnormally high within the dermis
  • No cartilage (seen in accessory tragus), central cysts or a central canal (seen in trichofolliculoma)
Differential diagnosis
  • Accessory tragus: also has increased vellus hairs, but usually has other types of tissue (i.e. mature adipose tissue or cartilage); location around the ear is also helpful
  • Trichofolliculoma: has central cyst or canal; deeper levels may be necessary to find trichofolliculoma in a lesion that resembles hair follicle nevus

Hemangioma & variants
Definition / general
Essential features
  • Most cases arise in children, with equal gender distribution
  • Composed of small capillary sized blood vessels with a larger feeding vessel commonly present
  • Most cases are treated with topical or systemic beta blockers in isolation or in combination with other modalities like laser therapy, excision, etc.
  • Recurrence is uncommon and only exceptional examples show malignant transformation
Terminology
  • Hemangioma is commonly used with a qualifier (e.g. congenital hemangioma)
ICD coding
  • ICD-O: 9120/0 - hemangioma, NOS
  • ICD-11: 2E81.0Y - other specified neoplastic hemangioma
Epidemiology
Sites
Pathophysiology
  • Specific pathogenesis has not been fully understood
  • Currently regarded as a multifactorial condition, resulting in endothelial proliferation, with uncontrolled angiogenesis and abnormal function of downstream pathways (notably HIF1α, VEGF and PI3K / Akt) (Biomed Res Int 2021;2021:5695378)
Etiology
  • Not known
Clinical features
Diagnosis
  • Requires correlation of detailed history and physical examination with microscopic features, as the latter overlaps for various clinical presentations
Laboratory
  • Transient mild to moderate thrombocytopenia due to consumption may be seen in rapid growth phase (F1000Res 2019;8:F1000)
Radiology description
  • Mostly required for extracutaneous cases
  • Skeletal (bone) hemangiomas: radiology is highly nonspecific (BMC Musculoskelet Disord 2021;22:27)
  • Ultrasound: highly effective; shows well defined mass with high vascularity
  • Contrast enhanced ultrasonography (CEUS): gold standard for hepatic hemangiomas (Korean J Radiol 2000;1:191)
  • High flow in proliferative phase of infantile hemangioma; slow flow in involuted phase
  • Central high flow for NICH; slow flow for RICH
  • Fluttering sign: recently described feature for grayscale ultrasound in hepatic hemangiomas (Ultrasound Med Biol 2021;47:941)
  • MRI: useful in deep locations to estimate extent of tissue involvement
  • Infantile hemangioma: hyperintense on T2; isointense on T1 with postcontrast enhancement
  • Congenital hemangioma: heterogeneous appearance on MRI
  • CT scan: hypodense, with postcontrast enhancement of periphery (World J Gastroenterol 2020;26:11)
Radiology images

Images hosted on other servers:
Hepatic hemangioma Hepatic hemangioma

Hepatic hemangioma

Intrathoracic hemangioma

Intrathoracic hemangioma

Infantile thyroid hemangioma

Infantile thyroid hemangioma

Infantile hepatic hemangioma

Infantile hepatic hemangioma

Choroidal hemangioma (fundoscopy)

Choroidal hemangioma (fundoscopy)


Hemangioma in long bones Hemangioma in long bones

Hemangioma in long bones

Hemangioma in long bones Hemangioma in long bones Hemangioma in long bones

Hemangioma in long bones

Prognostic factors
Case reports
Treatment
Clinical images

Contributed by Nasir Ud Din, M.B.B.S.
Noninvoluting congenital hemangioma

Noninvoluting congenital hemangioma



Images hosted on other servers:
Infantile hemangioma

Infantile hemangioma

Caption

Infantile hemangioma with ulceration

Infantile hemangioma in beard distribution

Infantile hemangioma in beard distribution

Rapidly involuting congenital hemangioma

Rapidly involuting congenital hemangioma

Partially involuting congenital hemangioma

Partially involuting congenital hemangioma

Laryngeal hemangioma (laryngoscopic view)

Laryngeal hemangioma (laryngoscopic view)

Gross description
Gross images

Images hosted on other servers:
Atrial hemangioma

Atrial hemangioma

Hepatic hemangioma

Hepatic hemangioma

Pulmonary hemangioma

Pulmonary hemangioma

Microscopic (histologic) description
  • Lobules of capillary sized vascular channels, lined by single layer of flattened endothelial cells
  • Large feeding vessel is usually seen at the deeper aspect
  • Associated lymphocyte infiltrate may be seen (Cardiovasc Pathol 2017;28:59)
  • Anastomosing hemangioma: anastomosing vascular channels lined by flattened endothelium; deep occurrence
  • Angiomatosis: involvement of multiple tissue planes with irregular and poorly circumscribed edges
  • Cavernous hemangioma: shows predominantly ectatic channels
  • Congenital hemangioma:
    • Solid appearance in rapid growth phase with poorly canalized vessels and mitotically active endothelium
    • Surrounding pericyte layer is present
    • With maturation, the lumina become prominent and blood flow ensues
    • Combination of solid and vascular areas in varying proportions may be seen
    • Noninvoluting congenital hemangioma (NICH) shows well formed capillaries and vascular channels
    • Involuting examples show thickening of basement membranes and fibrosis in the background
  • Epithelioid hemangioma:
    • Well formed small vessels are lined by plump endothelial cells with abundant eosinophilic cytoplasm and round enlarged nuclei, accompanied by abundant eosinophils in the background
    • Lobulated, well demarcated with maturation of vascular lumina at the periphery
  • Glomeruloid hemangioma: resembles glomerular capillaries
  • Hobnail hemangioma: hobnail nuclei protruding into vascular lumina; circumscribed
  • Infantile hemangioma:
    • Proliferation of capillary lobules; has a distinct natural history involving three stages (i.e., proliferation, partial regression and complete regression):
      • Early proliferative stage: lobules of immature dendritic type cells with intervening stroma, large feeding vessels and occasional presence of perineural involvement
      • Early regression: capillaries dilate and eventually start disappearing; apoptotic debris in basement membrane with increased numbers of pericapillary mast cells.
      • Late regression / end stage: ghosts of capillaries, rings of basement membrane with rare endothelial cells having immunophenotype of placental capillaries (GLUT1, LeY, CD15, CCR6, IDO and IGF2 positive)
  • Intramuscular angioma:
    • Arises within skeletal muscle in association with variable amount of mature adipose tissue, phleboliths and metaplastic bone formation
    • Shows a combination of lymphatics, variable sized veins and arteriovenous component
  • Lobular capillary hemangioma / pyogenic granuloma: ulceration with dense mixed inflammation
  • Microvenular hemangioma:
    • Poorly defined, in superficial and deep dermis
    • Small venule-like channels lined by single layer of endothelial cells which lack mitotic activity, surrounded by single layer of pericytes; these venules appear to dissect hyalinized collagen bundles of the dermis but lack multilayering and HHV8 positivity
  • Sinusoidal hemangioma:
    • Well demarcated proliferation of dilated, congested and thin vascular channels anastomosing in a sinusoidal manner
    • Intervening stroma is scant and scant smooth muscle may be present in the wall
    • Mitotic activity is not seen
  • Spindle cell hemangioma: proliferating spindle cells with intraluminal phleboliths
  • Verrucous hemangioma: hyperkeratosis and involvement of several tissue planes
Microscopic (histologic) images

Contributed by Nasir Ud Din, M.B.B.S.
Anastomosing hemangioma Anastomosing hemangioma

Anastomosing hemangioma

Skeletal hemangioma Skeletal hemangioma Skeletal hemangioma

Skeletal hemangioma

Cavernous hemangioma

Cavernous hemangioma


Hobnail hemangioma Hobnail hemangioma

Hobnail hemangioma

Hepatic hemangioma Hepatic hemangioma

Hepatic hemangioma

Infantile hemangioma Infantile hemangioma

Infantile hemangioma


Infantile hemangioma

Infantile hemangioma

Lobular capillary hemangioma Lobular capillary hemangioma Lobular capillary hemangioma

Lobular capillary hemangioma

Spindle cell hemangioma Spindle cell hemangioma

Spindle cell hemangioma


Spindle cell hemangioma

Spindle cell hemangioma

Phleboliths in spindle cell hemangioma

Phleboliths in spindle cell hemangioma

Synovial hemangioma

Synovial hemangioma

Verrucous hemangioma

Verrucous hemangioma

Virtual slides

Images hosted on other servers:
Extensive evolution

Extensive evolution

Involuting infantile hemangioma

Involuting infantile hemangioma

Congenital hemangioma

Congenital hemangioma

Cytology description
  • Aspiration is not advised, as there is high risk of uncontrolled bleeding
Positive stains
Negative stains
Electron microscopy description
  • Not required for diagnosis in routine cases
  • Endothelial cells show cytoplasmic folds on luminal surface, junctional complexes and cytoplasmic pinocytic vesicles (Cancer Res 1990;50:4787)
Electron microscopy images

Images hosted on other servers:
Endothelial differentiation

Endothelial differentiation

Cutaneous hemangioma

Cutaneous hemangioma

Molecular / cytogenetics description
Molecular / cytogenetics images

Images hosted on other servers:
Sanger sequencing for <i>RASA1</i>

Sanger sequencing for RASA1

Videos

Spindle cell hemangioma

Cutaneous vascular tumors

Sample pathology report
  • Liver, lobectomy:
    • Benign vascular lesion, consistent with hemangioma (see comment)
    • Comment: The tumor is completely excised with free margins. Evidence of embolization is seen, with presence of acellular material within the lumina of large caliber vessels, accompanied by areas of infarction within the lesion.
Differential diagnosis
  • Granulation tissue:
    • Sites of trauma, commonly cutaneous or mucosal
    • Shows associated inflammatory infiltrate and edema
    • Feeding vessels are not seen commonly
    • Lobular capillary hemangioma is common mimicker
    • Correlation with clinical history is important
  • Hemangioendothelioma:
    • Mimics proliferating phase of congenital hemangioma
    • Retiform type mimics hobnailing seen in proliferating phase of congenital hemangioma
    • Not well circumscribed
    • Clinical presentation with consumptive coagulopathy is common in both
    • May be associated with other anomalies
    • Requires more aggressive treatment
  • Kaposi sarcoma:
    • Mimics proliferating phase of congenital hemangioma due to compact cellularity and slit-like spaces
    • Eosinophilic hyaline globules are not seen in hemangioma
    • HHV8 association is not seen in hemangioma
    • Requires aggressive treatment
  • Angiosarcoma:
    • Malignant vascular tumor with infiltrative growth
    • Marked nuclear pleomorphism and brisk mitotic activity
    • Visceral location is more common compared to hemangioma
Board review style question #1

A newborn shows a nodular growth on his scalp. Over the next couple of weeks, the lesion starts shrinking. Despite reassurances from the pediatrician, the parents insist on excision. Upon pathological examination, it shows a network of capillary sized vessels with large feeding vessels near the base, shown in the above photomicrograph. The diagnosis in this case is

  1. Infantile hemangioma
  2. Noninvoluting hemangioma
  3. Partially involuting hemangioma
  4. Rapidly involuting hemangioma
Board review style answer #1
A. Infantile hemangioma. This case is an example of infantile hemangioma, which typically shows regression after birth, unlike congenital hemangioma, which grows with the patient, at least to some extent. Within the umbrella of congenital hemangioma, there is partial involution over time with shrinkage and secondary changes. Noninvoluting examples grow with the baby and rapidly involuting ones regress at a much faster rate, ultimately disappearing completely.

Comment Here

Reference: Hemangioma
Board review style question #2

A 9 year old boy presents with a purple nodular / bosselated lesion on the left leg, with involvement of the plantar and dorsal aspects of foot (as shown in the clinical photograph). He has had this lesion since birth and it has grown with him since then. The microscopic examination shows well formed vascular channels of variable size lined by a single layer of endothelial cells. The most likely diagnosis in this case is

  1. Birth mark
  2. Infantile hemangioma
  3. Kaposiform hemangioendothelioma
  4. Lobular capillary hemangioma
  5. Noninvoluting congenital hemangioma
Board review style answer #2
E. Noninvoluting congenital hemangioma. Noninvoluting congenital hemangioma is present at birth and grows with the child. As the name implies, there is no microscopic evidence of involution if a biopsy is performed. Birth marks commonly present with a flat, pigmented / reddish appearance of the skin rather than the appearance given in the clinical photograph, which shows prominent vascular marking, like the appearance of the lesion with some areas showing bosselation. Infantile hemangioma rapidly regresses after birth. Lobular capillary hemangioma presents as a nodular growth, commonly associated with ulceration of the skin or mucosa. Kaposiform hemangioendothelioma clinically presents commonly on the head and neck and trunk region, with a strong association with Kasabach-Merritt syndrome (consumptive coagulopathy). Microscopically, it shows glomeruloid proliferation and spindle cell morphology with slit-like spaces on microscopic examination.

Comment Here

Reference: Hemangioma

Hidradenoma
Definition / general
Essential features
  • Presents as a slow growing, nodular, solid or cystic cutaneous mass usually measuring up to 3 cm in diameter (Dermatology 2016;232:78)
  • Characterized by variably sized nests and nodules of epithelial cells within the upper or mid dermis, typically with no overlying connection to the epidermis (J Clin Pathol 2007;60:145)
Terminology
  • Also known as nodular hidradenoma, eccrine acrospiroma, clear cell hidradenoma, eccrine sweat gland adenoma and solid cystic hidradenoma
Epidemiology
Sites
Clinical features
  • Presents as a slow growing, nodular, solid or cystic cutaneous mass usually measuring up to 3 cm in diameter (Dermatology 2016;232:78)
  • Overlying skin can be skin colored, erythematous or blue and may exhibit superficial ulceration or serous discharge (Indian Dermatol Online J 2016;7:410)
Case reports
Treatment
Gross description
Microscopic (histologic) description
  • Well circumscribed but unencapsulated, lobulated / cystic mass with variably sized nests and nodules of epithelial cells within the upper or mid dermis, typically with no overlying connection to the epidermis (J Clin Pathol 2007;60:145)
  • Shows both solid and cystic components (Arch Pathol Lab Med 2005;129:e113)
    • Solid portion composed of 2 types of cells: polyhedral cells with basophilic cytoplasm and glycogen containing clear cells with eccentric round nucleus
    • Cystic areas secondary to degeneration of tumor cells
  • Tubular lumina are lined by cuboidal or columnar secretory cells (J Oral Maxillofac Pathol 2013;17:136)
  • Fibrovascular, collagenous or hyalinized stroma (J Cutan Pathol 2007;34:497, Arch Pathol Lab Med 2005;129:e113)
  • Focal apocrine decapitation secretion, squamous differentiation, squamous eddy formation, keratinization, mucinous change or sebaceous differentiation can be seen (Am J Dermatopathol 2012;34:461, J Cutan Pathol 2007;34:801, J Cutan Pathol 2007;34:497)
  • Malignant hidradenocarcinoma presents with infiltrative growth pattern, nuclear atypia and pleomorphism, predominantly solid cell islands, angiolymphatic invasion, necrosis and ≥ 4 mitoses per 10 high power fields (Middle East Afr J Ophthalmol 2010;17:374, Mod Pathol 2009;22:600)
  • Variants:
    • Clear cell hidradenoma (Cutis 2014;94:268)
      • Has a biphasic cellular population: (1) round, fusiform, or polygonal cells with vesicular nuclei and eosinophilic cytoplasm and (2) cells with clear cytoplasm and often eccentrically located nuclei
    • Solid cystic hidradenoma (Arch Pathol Lab Med 2005;129:e113)
      • Shows both solid and cystic components
      • Cysts probably represent cystic degeneration
    • Mucinous hidradenoma (J Cutan Pathol 2007;34:497)
      • Demonstrates diffuse and prominent mucinous cell proliferation
    • Poroid hidradenoma (Ann Dermatol 2021;33:289)
      • Has architectural features of the apocrine hidradenoma and cytological features of poroid and cuticular cells
      • Poroid cells are uniform, small cuboidal cells with an oval to round nuclei
      • Cuticular cells have an abundant eosinophilic cytoplasm with a larger nucleus that shows occasional multinucleation
    • Pigmented nodular hidradenoma (Arch Dermatol 1971;104:117)
      • Shows melanin pigmentation
Microscopic (histologic) images

Contributed by Jasmine Saleh, M.D., M.P.H. and Jodi Speiser, M.D.

Intradermal nodule

Polyhedral basophilic cells

Clear cells

CK8/18

CEA

Positive stains
Negative stains
Molecular / cytogenetics description
Sample pathology report
  • Skin, neck, excision:
    • Hidradenoma, extending to the deep margin
Differential diagnosis
  • Metastatic renal cell carcinoma
    • Absence of ductal differentiation/sweat production
    • Positive for PAX8 and CD10
    • Negative for p63
  • Basal cell carcinoma with eccrine differentiation:
    • Peripheral palisading and no cystic areas
    • Stromal epithelial retraction
    • Epidermal connection
  • Hidradenocarcinoma:
    • Greater cytologic atypia, mitoses and infiltrative
  • Squamous cell carcinoma:
    • Epidermal connection, keratin pearls and infiltrative
    • Absence of secretory differentiation
  • Trichilemmoma:
    • Broad connection with the epidermis
    • CD34, thickened hyalinized basement membrane and peripheral cell palisading
  • Poroma:
    • Epidermal connection and broad, anastomosing cords
    • Cells are more basaloid while cells of hidradenoma are more eosinophilic
    • Recurrent YAP1-MAML2 and YAP1-NUTM1 fusions identified in a subset
  • Cylindroma:
    • Jigsaw puzzle pattern
    • Hyaline globules and thickened basement membranes
Board review style question #1

    Which of the following is true about this identity?

  1. Always expresses CD10
  2. Has no connection to the epidermis
  3. Is typically encapsulated
  4. Presents as a fast growing mass
Board review style answer #1
B. Has no connection to the epidermis

Reference: Hidradenoma

Comment Here

Hidradenoma papilliferum
Definition / general
  • Hidradenoma papilliferum is a benign dermal papulonodular tumor almost always found in the vulva / perianal region of women
Essential features
  • Benign neoplasm of modified anogenital mammary-like glands, predominantly found in the vulva / perianal region of women
  • Histopathology features include characteristic dermal maze-like growth with papillary / glandular / cyst-like architecture, often with apocrine differentiation
Terminology
  • Also referred to as papillary hidradenoma
ICD coding
  • ICD-O: 8405/0 - papillary hidradenoma / hidradenoma papilliferum
  • ICD-10: D23.5 - other benign neoplasm of skin of trunk
  • ICD-10: D28.0 - benign neoplasm of vulva
Epidemiology
Sites
  • Vulva or perianal region
  • Rarely involves ectopic sites (head, neck, chest, abdomen) (Indian J Pathol Microbiol 2018;61:287)
    • 60% of ectopic HP is present in head and neck region, including eyelid, forehead, face, external auditory canal, post auricular region
    • Thought to arise from modified apocrine glands in eyelids / external auditory canal or heterotopic apocrine glands
    • Rare case reports of sebaceous differentiation in ectopic HP (Pathology 2019;51:362)
Pathophysiology
Etiology
  • Unknown
Clinical features
Diagnosis
Prognostic factors
  • Generally excellent prognosis and curative with complete excision
  • Can very rarely transform into malignant hidradenocarcinoma papilliferum or aggressive adenosquamous carcinoma (Gynecol Oncol 1989;35:395)
Case reports
Treatment
Clinical images

Images hosted on other servers:
Hidradenoma papilliferum of anus

Hidradenoma papilliferum of anus

Eyelid papule

Eyelid papule

Left areola erythematous papule

Left areola erythematous papule

External auditory canal

External auditory canal

Gross description
  • Solid, skin colored papule, usually < 1 - 2 cm
Microscopic (histologic) description
  • Well circumscribed dermal maze-like or arborizing proliferation (rarely connected to epidermis), with a combination of papillary, cystic or glandular architecture
  • 2 types of epithelia:
  • Entrapment of epithelial cells in connective tissue may mimic infiltrative pattern (Arch Gynecol Obstet 2011;284:1015)
  • May have connection to the epidermis with associated dermal plasma cell infiltrate (mimics syringocystadenoma papilliferum) (Am J Dermatopathol 2016;38:598)
  • Stromal changes: dense stromal plasma cell infiltrate, sclerosis, hyperplasia of myofibroblast-like cells, foamy macrophages
  • Metaplasia occasionally present
    • Most commonly oxyphilic (eosinophilic, granular cytoplasm, conspicuous nucleoli, occasional pleomorphism), oftentimes leading to misdiagnosis of malignancy (Am J Dermatopathol 2005;27:102)
    • Rarely squamous or mucinous metaplasia
  • Remnants of anogenital mammary-like glands (AGMLG) often present
  • Mitotic rate can be variable and sometimes high but high mitotic rate does not correlate with an aggressive outcome (Am J Dermatopathol 2006;28:322)
  • Rare case reports with sebaceous differentiation (in association with ectopic HP) and extramammary Paget disease (J Dermatol 2006;33:256, Am J Dermatopathol 2016;38:598)
Microscopic (histologic) images

Contributed by Bitania Wondimu, M.D.
Well circumscribed dermal lesion

Well circumscribed dermal lesion

Cystic lesion

Cystic lesion

Columnar and myoepithelial layers

Columnar and myoepithelial layers

Apocrine differentiation

Apocrine differentiation


Tubulopapillary architecture

Tubulopapillary architecture

CK7 highlights columnar cells

CK7 highlights columnar cells

SMA highlights myoepithelial layer

SMA highlights myoepithelial layer

p63 highlights myoepithelial layer

p63 highlights myoepithelial layer

Virtual slides

Images hosted on other servers:

Vulvar lesion

Vulvar biopsy

Negative stains
  • S100 (will highlight myoepithelial cells), HMWK
Molecular / cytogenetics description
  • Described to harbor mutations in genes involved in PI3K/ATK/MAPK signaling pathways (Pathology 2019;51:362)
  • Non-PI3K/AKT mutated cases display mutations in various other genes (PIK3CA PIK3R1, SYNE1, AR, IL6ST, PDGFRB, KMT2C, AR, BTK, DST, KAT6A, BRD3, RNF213, USP9X, ADGRB3, MAGI1, and IL7R) involved in PI3K/AKT signaling
  • Rare cases described mutations in BRAF, APC, ERBB4 (Genes Chromosomes Cancer 2016;55:113)
Videos

Diagnostic pearls of hidradenoma papilliferum

Sample pathology report
  • Skin, vulva, biopsy:
    • Diagnosis: hidradenoma papilliferum (see comment)
    • Comment: The biopsy contains a sharply circumscribed, maze-like glandular and papillary architecture without connection to overlying epithelium. The glands are lined by an inner layer of cuboidal cells and an outer layer of myoepithelial cells. Mitotic figures are rare. The morphologic features are most consistent with hidradenoma papilliferum.
Differential diagnosis
Board review style question #1

The lesion shown above is found in the vulva. What is the diagnosis?

  1. Hidradenocarcinoma
  2. Hidradenoma papilliferum
  3. Syringocystadenoma papilliferum
  4. Tubular apocrine adenoma
Board review style answer #1
B. Hidradenoma papilliferum

Comment Here

Reference: Hidradenoma papilliferum
Board review style question #2

A 57 year old woman develops a lesion on her vulva. Excision of the lesion shows a glandular and papillary proliferation (shown above). Which of the following is true concerning this lesion?

  1. Higher mitotic activity indicates worse prognosis
  2. It is often associated with a plasma cell infiltrate
  3. It is positive for EMA and GCDFP-15
  4. Malignant transformation occurs frequently
Board review style answer #2
C. It is positive for EMA and GCDFP-15

Comment Here

Reference: Hidradenoma papilliferum

Hidrocystoma
Definition / general
  • Uncommon benign adenomatous cystic proliferation derived from apocrine glands
  • Also called apocrine hidrocystoma, although apocrine cystadenoma may be preferable for lesions with true papillary projections and active epithelial secretions; these lesions are usually small (< 2 cm), on face of older adults (see also J Cutan Pathol 1997;24:249)
Sites
  • Despite its apocrine derivation, rare at sites rich in normal apocrine glands (groin, axilla, anogenital region, eyelids [Moll's glands], ears [ceruminous glands])
  • Usually solitary, but multiple lesions have been documented
  • Often occurs as cystic lesion of head and neck (commonly check), rarely in axilla
  • Similar lesion on eyelid known as Moll's gland cyst
Clinical features
  • Presents as intradermal, moderately firm, dome-shaped, translucent, blue, bluish-black or purple cystic nodule up to 1 cm
  • No gender preference
  • Lesions on face, also called Robinson variant, usually in middle aged women
  • Solitary lesions are not familial, but multiple apocrine hidrocystomas are a feature of ectodermal dysplasia (Acta Derm Venereol 2008;88:607) and focal dermal hypoplasia (Goltz syndrome)
  • Although solitary apocrine hidrocystomas lack seasonal variation, multiple lesions in some patients worsen in summer or with excessive heat and improve during winter
  • May be precursor to apocrine carcinoma
Case reports
Treatment
  • Complete excision recommended
Clinical images

Images hosted on other servers:
Eyelid, malar area, ear lobe

Eyelid, malar area, ear lobe

Dome shaped cystic mass

Dome shaped cystic mass

Lesion below lower lip

Lesion below lower lip

Capsule in subepidermis

Capsule in subepidermis

Skin colored nodules

Skin colored nodules

Gross description
  • Unilocular cystic lesion with clear to brown fluid
Microscopic (histologic) description
  • Large unilocular or multilocular cystic space within dermis
  • Fibrous pseudocapsule is often present
  • Typically, cystic spaces are lined by double layer of epithelial cells: an outer layer of flattened vacuolated myoepithelial cells and an inner layer of tall columnar cells with eosinophilic cytoplasm and basally located, round or oval vesicular nuclei
  • Decapitation secretion is usually present
  • Often adjacent to hyperplastic apocrine glands
Microscopic (histologic) images

Contributed by Angel Fernandez-Flores, M.D., Ph.D. and Dr. V. Zaitsev



Images hosted on other servers:
Decapitation secretion

Decapitation secretion

Cyst wall

Cyst wall

SMA, MIB1, GCDFP-15, CK7, CK18, CK19

SMA, MIB1, GCDFP-15, CK7, CK18, CK19

Positive stains
Electron microscopy description
  • Two types of cells: secretory cells with granules resembling lipid droplets, and basal (myoepithelial) cells
  • Lumen filled with cytoplasmic fragments detached from apical portions of secretory cells (Arch Dermatol 1979;115:194)
Differential diagnosis
  • Eccrine cystadenoma
  • Eccrine hidrocystoma

Histiocytoses, overview (pending)
[Pending]

ILVEN (pending)
[Pending]

Intravascular

Intravascular papillary endothelial hyperplasia
Definition / general
  • Also called Masson hemangioma (Am J Dermatopathol 2003;25:71)
  • Often on fingers and in hemorrhoids
  • In skin, associated with preexisting pyogenic granuloma or hemangioma, vascular malformations or hemangiomas of blue rubber bleb nevus syndrome
  • Due to exuberant recanalization of a thrombus
  • May be secondary to trauma or superimposed on pyogenic granuloma or cavernous hemangioma
  • Slow growing, tender, blue red, deep dermal to subcutaneous mass
  • Rarely recurs after excision
Case reports
Microscopic (histologic) description
  • Dilated vessel contains papillary proliferation of plump endothelial cells without atypia overlying fibrous tissue
  • Fibrin deposition and thrombi present
  • Variable pleomorphism and stratification, no/rare mitotic figures, no necrosis, no solid cellular areas
Microscopic (histologic) images

Images hosted on other servers:

Histological features of IPEH

Differential diagnosis
  • Angiosarcoma: infiltrating, freely anastomosing channels lined by spindled to epithelioid endothelial cells with marked atypia, surrounding adnexae and dissecting dermal collagen

Inverted follicular keratosis
Definition / general
  • Benign tumor of the follicular infundibulum
Essential features
  • Benign, endophytic follicular tumor with characteristic presence of squamous eddies
Terminology
  • Inverted follicular keratosis (IFK)
ICD coding
  • ICD-9: 216 - benign neoplasm of skin
  • ICD-10: L82.1 - other seborrheic keratosis
Epidemiology
Sites
Pathophysiology
  • Unknown at this time
Etiology
Clinical features
Diagnosis
  • Can be made on biopsy
Case reports
Treatment
  • Biopsy is usually curative
  • Larger lesions treated by complete excision
Clinical images

Images hosted on other servers:
Pedunculated nodule in the upper cutaneous lip

Pedunculated nodule in the upper cutaneous lip

Microscopic (histologic) description
  • Well circumscribed, endophytic tumor with large lobules or finger-like extensions that resemble expanded follicles
  • Variable number of squamous eddies
  • Occasional mitoses within peripheral basaloid cells
  • Histologic variants have been described (J Cutan Pathol 1984;11:387):
    • Papillomatous wart-like: exophytic with overlying hyperkeratosis and parakeratosis
    • Keratoacanthoma-like: central exoendophytic mass
    • Cystic type: irregular clefts within tumor and formation of small cysts
Microscopic (histologic) images

Contributed by Gregory A. Hosler, M.D., Ph.D.

Silhouette

Squamous eddies

Desmoplastic features

Mucin deposition

Pseudoviral features

Positive stains
  • BCL2: positive dendritic cells are present in increased numbers in the suprabasal areas, compared to seborrheic keratosis (J Cutan Pathol 2006;33:498)
Videos

Inverted follicular keratosis:
5 minute pathology pearls

Inverted follicular keratosis with great squamous eddies

Sample pathology report
  • Skin, upper cutaneous lip, shave biopsy:
    • Inverted follicular keratosis
Differential diagnosis
  • Irritated seborrheic keratosis:
    • Presence of squamous eddies; however, lacks endophytic growth
  • Tricholemmoma:
    • Similar lobulated, endophytic architecture
    • Clear glycogenated cells, palisaded basilar layer and thickened basement membrane
  • Verruca vulgaris:
    • More exophytic than endophytic with inward curving papillomatosis, coarse hypergranulosis and koilocytes
    • Associated with human papillomavirus
Board review style question #1

Which of the following is true about inverted follicular keratosis (IFK)?

  1. BCL2 is downregulated in dendritic cells
  2. Squamous eddies are an atypical finding in IFK
  3. These lesions are more common in young women
  4. These lesions are more commonly found on the head and neck
Board review style answer #1
D. These lesions are more commonly found on the head and neck. BCL2 has been found to be upregulated in the dendritic cells of IFK but not seborrheic keratosis. IFK is typically found on the head and neck of older men. Squamous eddies are a usual finding in IFK.

Comment Here

Reference: Inverted follicular keratosis
Board review style question #2

This image comes from the head of an elderly patient. What is the most likely diagnosis?

  1. Inverted follicular keratosis
  2. Poroma
  3. Squamous cell carcinoma
  4. Trichilemmoma
  5. Verruca vulgaris
Board review style answer #2
A. Inverted follicular keratosis

Comment Here

Reference: Inverted follicular keratosis

Juvenile xanthogranuloma
Definition / general
  • Rare, sporadic, benign disorder which results from the proliferation of factor XIIIa positive dendritic cells
  • Most common form of non-Langerhans cell histiocytosis
  • Typically a disorder of early childhood
Essential features
  • Juvenile xanthogranuloma (JXG) is a rare, benign proliferative non-Langerhans cell histiocytic proliferation
  • Occurs predominantly in young children
  • Clinically, it presents as a solitary red-brown, yellowish papule or nodule, most often on the head and neck area
  • Systemic JXG is rare and may involve any organ system
  • Histologically characterized by the presence of histiocytes, foam cells and Touton giant cells
  • Generally follows a benign course with spontaneous resolution over a period of a few years
  • Cutaneous JXG is usually self limited
    • Cases of ocular JXG should be referred to an ophthalmologist
    • Symptomatic systemic JXG may require treatment
Terminology
  • Nevoxanthoendothelioma, xanthoma multiplex, juvenile xanthoma, multiple eruptive xanthoma in infancy, congenital xanthoma tuberosum, xanthoma neviforme and juvenile giant cell granuloma
ICD coding
  • ICD-10: D76.3 - Other histiocytosis syndromes
Epidemiology
  • Incidence is unknown
  • Between 40 - 70% of cases arise in the first year of life
  • More than 15 - 20% of patients have lesions at birth
  • Adult onset accounts for 10% of cases
  • M:F = ~1.4:1 (Actas Dermosifiliogr 2020;111:725)
  • Has been associated with juvenile chronic myelogenous leukemia and neurofibromatosis type I (Br J Dermatol 2017;176:481)
Sites
  • Presents as a solitary cutaneous nodule on the head, neck or trunk
  • Occasionally extracutaneous presentation of nodules occurs within the eye (most common), soft tissues or visceral organs
    • Ocular JXG is usually unilateral and most often develops before the age of 2
    • Hyphema and glaucoma are serious complications of ocular JXG
  • Systemic involvement is rare, occurring in less than 5% of cases
  • Reference: Actas Dermosifiliogr 2020;111:725
Pathophysiology
  • Unknown at this time
Etiology
  • Cause is unknown
  • Suggested that histiocytes possibly react to a traumatic or infectious stimuli by progressive lipidation
Clinical features
  • 2 clinical variants have been described: a small and a large nodular form; both present as red-brown, dome shaped papules
    • Small nodular form usually consists of multiple 2 - 5 mm sized papules, whereas the large nodular form is characterized by 1 or a few nodules 1 - 2 cm in diameter; these 2 forms can coexist
  • Has been associated with insulin dependent diabetes mellitus, aquagenic pruritus, urticaria pigmentosa and cytomegalovirus infection
  • Most adult patients have solitary lesions
  • Reference: Actas Dermosifiliogr 2020;111:725
Diagnosis
  • Diagnosis is made clinically based on the typical appearance of lesions on physical examination in most cases
  • On dermoscopy, the setting sun sign is described, which corresponds to a yellow-orange, central area surrounded by a peripheral border with branched, linear telangiectasias (An Bras Dermatol 2018;93:138)
  • In older lesions, white and yellow globules are most commonly seen on dermoscopy because of the more lipidized cells (An Bras Dermatol 2018;93:138)
Prognostic factors
  • Has a good prognosis; lesions usually regress spontaneously
  • Cases of liver or central nervous system involvement may rarely be fatal
Case reports
  • 31 week preterm newborn with multiple skin lesions whose clinical, histological and immunohistochemical findings were consistent with juvenile xanthogranuloma (Rev Paul Pediatr 2019;37:257)
  • 14 month old and 14 year old girls with oral JXG affecting the lower lip and the soft palate, respectively (Pediatr Dent 2017;39:238)
  • 12 year old girl with a slowly growing yellowish polypoid lesion on the posterior scalp (Am J Dermatopathol 2017;39:773)
Treatment
Clinical images

Contributed by Lamiaa Hamie, M.D., M.Sc. and Ossama Abbas, M.D.

Red-brown papule

Microscopic (histologic) description
  • Demarcated dense dermal infiltrate of foamy histiocytes that can extend to the subcutis in large lesions
  • Scattered Touton giant cells are characteristic
  • Can have loss of the rete ridges and ulceration
  • Lymphocytes, plasma cells, eosinophils and some neutrophils can be observed
  • Early lesions usually have monomorphous histiocytes with abundant eosinophilic cytoplasm
  • In older lesions, the histiocytes become more xanthomatous by developing lipid in their cytoplasm, creating a foamy appearance
  • Reference: Actas Dermosifiliogr 2020;111:725
Microscopic (histologic) images

Contributed by Lamiaa Hamie, M.D., M.Sc. and Ossama Abbas, M.D.

Monomorphic histiocytic infiltrate

Foamy histiocytic infiltrate

Scattered Touton giant cells

Foamy histiocytes and a Touton giant cell



Contributed by Jijgee Munkhdelger, M.D., Ph.D. and Andrey Bychkov, M.D., Ph.D.

Low power

Inflammatory cells

Fibrohistiocytic proliferation with giants

Touton giant cell

Juvenile xanthogranuloma immunoprofile

Virtual slides

Images hosted on other servers:
Missing Image

Mononuclear histiocytes and multinucleated giant cells

Missing Image

CD68 demonstrates histiocytic origin of proliferation

Negative stains
Videos

Brief review of JXG

Sample pathology report
  • Right arm, excision:
    • Xanthogranuloma (see comment)
    • Comment: Dense sheets of histiocytes infiltrating the dermis and the upper portion of the subcutaneous fat. Numerous multinucleated giant cells of the Touton cell type. In addition, clinicopathologic correlation may be helpful.
Differential diagnosis
  • Langerhans cell histiocytosis:
    • LCH cells are positive for S100 protein, CD1a and langerin (CD207)
    • Intracytoplasmic Birbeck granules are seen on electron microscopy
    • Systemic manifestations include osteolytic bone lesions, diabetes insipidus, hepatosplenomegaly and lymphadenopathy
  • Papular xanthoma:
    • Multiple yellow-red papules
    • Mainly on the trunk
    • Fewer Touton cells than JXG
    • Factor XIIIa is more likely to be negative
  • Tuberous xanthoma:
    • Occurs in areas of pressure
    • Associated with hypercholesterolemia and high LDL
    • Consists of dermal aggregates of foam cells, no Touton cells or inflammatory cells
  • Spitz nevus:
    • Nests of spindle and epithelioid cells within a uniformly hyperplastic epidermis
    • Melanocytic immunostaining with S100, MelanA (MART1) and tyrosinase
  • Mastocytoma:
    • Reddish golden-brown papules
    • Can display Darier sign
    • Can be pruritic
    • Mast cells in the papillary dermis
  • Dermatofibroma:
    • Brownish papules
    • Mainly on the extremities
    • May arise after trauma
    • Older age group
    • Dermal spindle shaped proliferation with collagen trapping
  • Reticulohistiocytoma:
    • More prominent nucleoli
    • Few to no Touton giant cells
    • More likely to be punctuated by inflammatory foci
    • Ground glass cytoplasm (can be seen in some JXGs)
  • Erdheim-Chester disease:
    • Non-Langerhans cell histiocytosis that usually affects adults
    • Skin involvement is rare but can present with red-brown to yellow nodules / plaques
    • Fever, bone pain, exophthalmos, diabetes insipidus
    • CD68+, CD163+, CD1a- foamy histiocytes
    • Touton giant cells also often present
Board review style question #1

A 1 year old girl was brought to the clinic for an asymptomatic red-yellow papule on her face with the above histological findings. Which is the most common extracutaneous site of involvement?

  1. Eyes
  2. Central nervous system
  3. Lungs
  4. Bone
  5. Liver
Board review style answer #1
A. This is a juvenile xanthogranuloma. Its most common site of extracutaneous presentation is the eyes.

Comment Here

Reference: Juvenile xanthogranuloma
Board review style question #2
A 2 year old girl was brought to the clinic for an asymptomatic red-yellow papule of a few months' duration on the arm. On histological examination, there was a dermal infiltrate of foamy histiocytes and multinucleated giant cells with a ring of nuclei surrounded by a foamy cytoplasm. Which of these markers would be negative on immunohistochemical staining?

  1. HAM56
  2. CD207
  3. Factor XIIIa
  4. CD163
  5. CD68
Board review style answer #2
B. This is a juvenile xanthogranuloma, which is negative for CD207 (Langerin).

Comment Here

Reference: Juvenile xanthogranuloma

Kaposi sarcoma

Keloid
Definition / general
  • Abnormal fibroblast reaction to injury
  • Usually in people of African descent, often in earlobe
  • High rate of recurrence after excision
Treatment
  • Corticosteroid injection
Clinical images

Contributed by Mark R. Wick, M.D.
Breast skin

Breast skin

Microscopic (histologic) description
  • Wide bands of collagen with large, brightly eosinophilic, glassy fibers
  • Also parallel fibroblasts and myofibroblasts
  • Mucinous pools after steroid injection
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D.
Molecular / cytogenetics description
Differential diagnosis
  • Complication of acne
  • Hyperplastic scar
  • Keloidal dermatofibroma

Keloid
Definition / general
  • Result of abnormal fibroblast activity in response to skin trauma or inflammation resulting in increased production of collagen and growth factors
  • Nodule or mass forming scar that extends beyond the site of initial injury
Essential features
  • Benign and potentially disfiguring fibrous tumor caused by increased fibroblast activity in response to skin trauma, causing increased collagen and growth factor production
  • Common in individuals with darker skin and most common in African Americans (Cureus 2022;14:e23503)
  • In comparison to hypertrophic scars, keloids extend beyond the site of initial injury
  • Persist until treated, commonly by a combination of intralesional corticosteroids and surgical excision
  • Recurrence is common despite treatment (Am Fam Physician 2009;80:253)
Terminology
  • Keloid scar
ICD coding
  • ICD-10: L91.0 - hypertrophic scar
  • ICD-11
    • EE60 - keloid or hypertrophic scars
    • EL50.0 - keloidal surgical scar
    • EE60.1 - hypertrophic scar
    • EL501.1 - hypertrophic surgical scar
Epidemiology
  • Common in < 30 years of age (Am Fam Physician 2009;80:253)
  • Commonly seen after ear piercings
  • Most common in Black and darker pigmented individuals
  • Positive family history increases risk of development
Sites
  • Any site of trauma or injury to the skin can be affected
  • Typically occurs in the head and neck region, including upper part of the back, deltoids and presternal areas
  • Characteristically occurs on the earlobes from ear piercings
Pathophysiology
  • Exact pathophysiology is unknown
  • Commonly accepted to be due to a dysregulated fibroblastic phase of wound healing, in which fibroblasts have increased proliferative activity and decreased apoptosis resulting in increased collagen, extracellular matrix and cytokine production (J Clin Aesthet Dermatol 2020;13:33, StatPearls: Keloid [Accessed 28 July 2023])
  • Primary drivers of keloid formation are considered to be transforming growth factor beta (TGFβ) and platelet derived growth factor (PDGF) (StatPearls: Keloid [Accessed 28 July 2023])
    • TGFβ promotes chemotaxis of fibroblasts to the site of inflammation and produces collagen
    • Abnormalities in this pathway lead to fibrosis and an abnormal scar response
Etiology
  • Caused by genetic and environmental factors
  • Can occur as a result of increased tension in a wound
  • Occurs in susceptible individuals after any type of skin trauma including surgery, piercings, acne, tattooing, insect bites, burns, lacerations, abrasions, vaccinations and any other process resulting in cutaneous inflammation (StatPearls: Keloid [Accessed 28 July 2023])
Diagrams / tables
Not relevant to this topic
Clinical features
Diagnosis
  • Diagnosis is made clinically but lesions may be biopsied for confirmation or excised for cosmetic purposes
Laboratory
Not relevant to this topic
Radiology description
Not relevant to this topic
Radiology images
Not relevant to this topic
Prognostic factors
  • Benign but may cause symptoms including pain and pruritus
  • Persists unless treated; most commonly removed for cosmetic reasons and if symptomatic
  • Despite treatment, recurrence is common (Scars Burn Heal 2021;7:2059513120980320)
Case reports
Treatment
  • Complete surgical excision
  • In conjunction with complete surgical excision, intralesional corticosteroid injections imiquimod, bleomycin or radiotherapy can be used to decrease the risk of recurrence (Dermatol Ther 2022;35:e15425)
  • Compression bandages, silicone dressings, cryotherapy, laser therapy and intralesional 5-fluorouracil are also treatment options
Clinical images

Images hosted on other servers:
Keloids before and after treatment Keloids before and after treatment

Keloids before and after treatment

Keloids before and after treatment Keloids before and after treatment Keloids before and after treatment

Keloids before and after treatment


Recurrent keloid formation on earlobe Recurrent keloid formation on earlobe

Recurrent keloid formation on earlobe

Vertical growth

Vertical growth

Horizontal growth

Horizontal growth

Characteristic shape

Characteristic shape


Erythema Erythema

Erythema

Elevation

Elevation

Gross description
Gross images

Images hosted on other servers:
Trabeculated, whitish fibrous appearance

Trabeculated, whitish fibrous appearance

Frozen section description
Not relevant to this topic
Frozen section images
Not relevant to this topic
Microscopic (histologic) description
  • Characteristic keloidal collagen is composed of broad, homogeneous, brightly eosinophilic bands of hyalinized collagen bundles
  • Keloidal collagen in often described as bubble gum-like collagen
  • Collagen bands are haphazardly arranged or form a nodular lesion in the reticular dermis sparing the papillary dermis (Am J Dermatopathol 2004;26:379)
  • Focal aggregates of fibroblasts can be seen in between or around the collagen bands (Scars Burn Heal 2021;7:2059513120980320)
Microscopic (histologic) images

Contributed by Monika Bhatt, M.D.
Nodular dermal lesion

Nodular dermal lesion

Keloidal collagen

Keloidal collagen

Dermal lesion

Dermal lesion

Fibroblasts

Fibroblasts

Left ear nodule

Left ear nodule

Keloidal collagen

Keloidal collagen

Virtual slides

Images hosted on other servers:

Thick collagen bundles in the dermis

Cytology description
Not relevant to this topic
Cytology images
Not relevant to this topic
Immunofluorescence description
Not relevant to this topic
Immunofluorescence images
Not relevant to this topic
Positive stains
Not relevant to this topic
Negative stains
Not relevant to this topic
Electron microscopy description
Not relevant to this topic
Electron microscopy images
Not relevant to this topic
Molecular / cytogenetics description
Not relevant to this topic
Molecular / cytogenetics images
Not relevant to this topic
Videos

Dr. Gardner explains histological findings of keloids

Sample pathology report
  • Skin, ear lobe, excision:
    • Keloid
Differential diagnosis
  • Hypertrophic scar:
    • Does not grow outside of wound margins
    • Can regress on its own
    • Does not contain keloid type collagen bundles on histological exam
  • Dermatofibroma:
    • Positive dimple sign on physical exam; lesion develops a central depression when lateral pressure is applied
    • More common on the lower legs
    • No history of preceding trauma
  • Dermatofibrosarcoma protuberans:
    • No history of trauma
    • Harbors translocation t(17;22)(q22;q13) that results in a COL1A1::PDGFB fusion gene
    • Most commonly found on the chest, shoulders and limbs
  • Lobomycosis:
    • Fungal infection caused by Lacazia loboi
    • Usually found on the distal extremities
    • History of exposure to dolphins or rural soil in Central and South America
    • Microscopically granulomatous inflammation with giant cell reaction is seen
    • PAS and silver stain highlight the fungal organisms
Board review style question #1

A mass forming nodule is excised from a patient's ear. Histological findings are shown in the image above. What is the diagnosis?

  1. Dermatofibroma
  2. Dermatofibrosarcoma protuberans
  3. Hypertrophic scar
  4. Keloid
Board review style answer #1
D. Keloid. The image above shows a nodular bundle of keloidal collagen within the reticular dermis. The collagen bundles are thick eosinophilic bands arranged in a nodular or haphazard pattern, a characteristic finding in keloids. Also, the ear is the most common site for keloid formation. Answer C is incorrect because hypertrophic scars would have fewer thick collagen fibers. Answer A is incorrect because dermatofibromas are more common on the lower extremities. Answer B is incorrect because dermatofibrosarcoma protuberans are most commonly found on the chest, shoulders and limbs.

Comment Here

Reference: Keloid
Board review style question #2
Which of the following commonly occurs after excision of a keloid?

  1. Complete resolution
  2. Increase in propensity for development of squamous cell carcinoma
  3. Melanoma in situ
  4. Recurrence
Board review style answer #2
D. Recurrence. Keloids are a benign entity that are known to have the potential to recur after surgical excision. Therefore, intralesional steroids or radiotherapy in conjunction with surgical management is usually the treatment of choice to prevent recurrence. Answers B and C are incorrect because keloids are not known to have malignant transformation or an increased propensity for cancer. Answer A is incorrect because recurrence is common despite treatment.

Comment Here

Reference: Keloid
Board review style question #3
Which of the following is a key difference between keloids and hypertrophic scars?

  1. Keloids grow beyond the wound site
  2. Keloids have an increased propensity for malignant transformation
  3. Keloids never recur after treatment
  4. Keloids occur exclusively in pigmented skin
Board review style answer #3
A. Keloids grow beyond the wound site. On gross examination, keloids grow beyond the site of initial injury, while hypertrophic scars remain confined to the wound margins. Answer D is incorrect becuase keloids can occur in all skin types although individuals with darker pigmented skins are more prone to keloid formation after skin trauma. Answer C is incorrect because keloids can recur after treatment. Answer B is incorrect because keloids are benign skin lesions and do not have propensity for malignant transformation.

Comment Here

Reference: Keloid

Keratoacanthoma / SCC keratoacanthoma type
Definition / general
  • Keratoacanthoma (KA) is a well differentiated, cutaneous squamous cell carcinoma, which often spontaneously regresses
  • Regression is thought to be due to immune mediated destruction of squamous cells
  • For lesions that are entirely resected, can diagnose as "well differentiated squamous cell carcinoma, keratoacanthoma type”
  • For lesions that are not entirely histologically examined, can diagnose as "well differentiated squamous cell carcinoma with features of keratoacanthoma”
Essential features
  • Common, represents approximately up to 30% of cases of cutaneous squamous cell carcinoma (SCC)
  • Dome shaped tumor with central keratinous plug in sun exposed areas
  • May become large, up to 10 cm (called giant KA)
  • Good prognosis; vast majority of cases spontaneously regress
Terminology
  • Agglomerate KA (variant)
  • KA centrifugum (variant)
  • Giant KA (variant)
  • Subungual KA (variant)
  • Intraoral KA (variant)
ICD coding
  • ICD-O: 8071/3 - keratoacanthoma, NOS
  • ICD-10: L85.8 - other specified epidermal thickening
  • ICD-11: 2F72 - neoplasms of uncertain behavior of skin
Epidemiology
Sites
Pathophysiology
Clinical features
Diagnosis
  • Excisional biopsy preferred to show suggestive histopathology
Prognostic factors
  • Nonfatal tumor
  • Can regress; however, final size prior to regression is difficult to predict (J Am Acad Dermatol 2016;74:1220)
  • Recurrence is uncommon with surgical removal
  • Self limited proliferation with low probability of malignant transformation (more often associated with perineural invasion) (Acta Biomed 2019;90:580)
Case reports
Treatment
  • Monitoring for spontaneous regression; often leaves atrophic hypopigmented scar (Indian J Dermatol 2011;56:435)
  • Typically treated with complete surgical excision; Mohs surgery performed dependent on location (Ann Dermatol 2011;23:357)
  • Electrosurgery, cryosurgery, laser surgery, systemic or topical chemotherapy occasionally utilized (J Dent (Shiraz) 2014;15:91)
  • Radiotherapy can be utilized with reoccurrence or potential large cosmetic disfigurement with surgery; typically not utilized in young adult patients (J Am Acad Dermatol 1990;23:489)
Clinical images

Images hosted on other servers:

Large face tumor

Palm tumor

Forehead lesion

Gross description
  • Typically large, scaly, dome shaped tumor with central keratinous plug
Microscopic (histologic) description
  • Low power magnification shows large, well differentiated squamous tumor with central keratin filled crater
  • Surrounding epidermis forms a lip around the invaginating crateriform tumor
  • Tumor is composed of bland squamous cells with abundant eosinophilic or glassy cytoplasm and enlarged hyperchromatic to vesicular appearing nuclei
  • Mitotic activity and cellular atypia are usually seen at the periphery of tumor
  • Intraepidermal neutrophilic microabscesses and keratin horn pearls are seen within the tumor
  • Regressing tumors show epidermal atrophy, bland cytologic features, dermal inflammation and fibrosis at the periphery of tumor
Microscopic (histologic) images

Contributed by Poonam Sharma, M.B.B.S.

Central crater

Bland squamous cells

Whorl of laminated keratin

Virtual slides

Images hosted on other servers:

Invaginated, keratin filled tumor

Positive stains
Negative stains
Molecular / cytogenetics description
  • Transforming growth factor beta receptor 1 can cause multiple self healing squamous epithelioma (MMSE); most common cause of multiple KA
  • Germline mutations of hMSH2 and hMLH1 mismatch repair genes causing Muir-Torre syndrome have been implicated in solitary and multiple KA formation
  • BRAF kinase inhibitors utilized to treat KA has been reported to induce reactive KA by paradoxical activation of the ERK MAPkinase pathway
  • Reference: Exp Dermatol 2016;25:85
Videos

Keratoacanthoma

Sample pathology report
  • Skin, left cheek (clinical keratoacanthoma), excision:
    • Well differentiated squamous cell carcinoma, keratoacanthoma type (see comment)
    • Comment: Well differentiated hyperplastic squamous epithelium with crater-like depression with keratin horn pearls present centrally. Enlarged hyperchromatic vesicular nuclei present along with glassy appearing cytoplasm.
Differential diagnosis
  • Conventional, well differentiated squamous cell carcinoma:
    • Shows greater cellular atypia and mitotic activity
    • Intraepidermal neutrophilic microabscesses and tissue eosinophilia less common
    • May be difficult to distinguish in superficial biopsies
  • Verrucous carcinoma:
    • Shows prominent endophytic and exophytic growth
    • Lacks central crateriform architecture
    • Also has bland cytologic features
    • May be difficult to distinguish in superficial biopsies
Board review style question #1

A 62 year old man presents with a single, solitary, skin colored tumor that has a crater-like appearance with central keratin buildup on his left forearm. It appeared spontaneously and has grown rapidly over the past 3 weeks. It is painless to the touch but is cosmetically displeasing. He has no other skin conditions. The tumor is excised and a microscopic image is shown above. Which of the following is true concerning this skin lesion?

  1. It is a variant of basal cell carcinoma
  2. The underlying cause for this tumor is typically due to a genetic mutation
  3. The malignant potential of this tumor is high and it must be removed immediately
  4. This tumor has the potential to completely spontaneously regress
Board review style answer #1
D. This tumor has the potential to completely spontaneously regress. This is a well differentiated squamous cell carcinoma, keratoacanthoma type.

Comment Here

Reference: Keratoacanthoma
Board review style question #2
A 70 year old male farmer presents with a skin tumor on his cheek that has grown quickly over the past month. It has a crater-like appearance without arborizing telangiectasias on dermatoscopy. Biopsy shows that the tumor is mainly composed of bland squamous cells with intraepidermal neutrophilic microabscesses and keratin horn pearls under the microscope. Which of the following is true concerning this skin tumor?

  1. Peripheral palisading and clefting are also likely to be seen under the microscope
  2. It has overlapping features of squamous cell carcinoma
  3. Due to its rapid growth, this tumor is likely to metastasize
  4. Excessive UV damage plays no role in pathogenesis
Board review style answer #2
B. It has overlapping features of squamous cell carcinoma. The lesion is a keratoacanthoma.

Comment Here

Reference: Keratoacanthoma

Langerhans cell histiocytosis

Large cell acanthoma
Definition / general
  • Benign intraepidermal lesion composed of large keratinocytes
  • Debate as to whether this represents a distinct entity or a reaction pattern within a solar lentigo (Int J Dermatol 2003;42:36)
  • Recent study suggests large cell acanthoma is a solar lentigo with cellular hypertrophy (J Cutan Pathol 2014;41:733)
Case reports
Gross description
  • Light-tan to dark brown, well circumscribed macule or patch
  • Few millimeters to centimeters in diameter
  • Typically on sun exposed skin
  • Clinically resembles a seborrheic or actinic keratosis
Microscopic (histologic) description
  • Atrophic or acanthotic epidermis containing larger than usual keratinocytes (~2x normal epidermal keratinocytes)
  • Usually well demarcated from adjacent epidermis
  • Basal layer may appear hyperpigmented; sometimes conspicuous rete ridges
  • Orthokeratosis; may have hypergranulosis
  • Minimal nuclear pleomorphism
  • Basal mitoses only
Microscopic (histologic) images

Images hosted on other servers:
Features

Features

Large keratinocytes, hyperkeratosis

Large keratinocytes, hyperkeratosis

Differential diagnosis

Leiomyoma
Definition / general
  • Divided into lesions of nipple or scrotum, pilar leiomyoma or solitary angioleiomyoma (vascular leiomyoma) usually in subcutis
  • May be very painful
  • Familial cutaneous leiomyomatosis (Reed syndrome):
Clinical features
Case reports
Treatment
Clinical images

Images hosted on other servers:

Multiple asymptomatic, hyperkeratotic nodules on leg

Gray-brown macules on face

Trunk

Microscopic (histologic) description
  • Intersecting smooth muscle fascicles
  • May have scattered bizarre hyperchromatic nuclei (symplastic leiomyoma)
  • No atypia, no mitotic activity, no necrosis
  • Pilar leiomyoma:
    • Dermal intersecting fascicles of eosinophilic spindle cells with plump, cigar shaped nuclei with dermal collagen bundles
Microscopic (histologic) images

Contributed by Angel Fernandez-Flores, M.D., Ph.D.

Various images

Positive stains
Negative stains
Molecular / cytogenetics description
Differential diagnosis

Leiomyoma
Definition / general
  • Cutaneous leiomyomas are benign tumors originating from arrector pili (piloleiomyoma), vascular (angioleiomyoma), periareolar and genital smooth muscle cells (genital leiomyoma)
  • Cutaneous piloleiomyomas may be sporadic or associated with the autosomal dominant syndrome multiple cutaneous and uterine leiomyomatosis (also known as Reed syndrome or hereditary leiomyomatosis and renal cell cancer)
Essential features
  • Cutaneous leiomyomas are benign dermal tumors of smooth muscle cells arranged in interlacing fascicles
  • Leiomyomas may be differentiated from other spindle cell tumors due to their positivity for both SMA and desmin
  • Cutaneous piloleiomyomas can be sporadic or inherited in association with multiple cutaneous and uterine leiomyomatosis (also known as Reed syndrome or hereditary leiomyomatosis and renal cell cancer)
Terminology
Not relevant to cutaneous leiomyomas
ICD coding
  • ICD-10: D21.9 - benign neoplasm of connective and other soft tissue, unspecified
Epidemiology
  • Leiomyomas are typically diagnosed in young and middle aged adults; however, they can occur in children
  • Frequency of piloleiomyomas is about equal in males and females and it is unknown whether a sex predominance exists for genital leiomyomas; there is no known racial predilection for cutaneous leiomyomas (StatPearls: Cutaneous Leiomyomas [Accessed 10 April 2024])
  • Angioleiomyomas have been further split by some authors into solid (more common in females), cavernous and venous subtypes (cavernous and venous being more common in males) (Laryngoscope 2004;114:661)
Sites
  • Cutaneous leiomyomas can occur anywhere on the body
Pathophysiology
Repetitive with definition and epidemiology headings
Etiology
  • Germline heterozygous mutations of the fumarate hydratase gene are found in cutaneous piloleiomyomas associated with the autosomal dominant syndrome multiple cutaneous and uterine leiomyomatosis (also known as Reed syndrome or hereditary leiomyomatosis and renal cell cancer)
  • Fumarate hydratase missense mutations are associated with decreased enzyme activity suggesting a tumor suppressor role (J Mol Diagn 2005;7:437)
  • Little is known about the etiology of sporadic cutaneous leiomyomas (Int J Mol Med 2004;13:13, Cancer Genet Cytogenet 2006;170:58, Cancer Genet Cytogenet 2010;199:21)
    • Recurrent losses in chromosome 22 and recurrent gains in Xq have been reported in few cases of sporadic angioleiomyomas
    • Few studies have examined cytogenetics of genital leiomyomas
    • No consistent mutations have been identified in sporadic piloleiomyomas to date
Diagrams / tables
Not relevant to this topic
Clinical features
  • Cutaneous leiomyomas can be solitary or multiple; when multiple, they may be clustered or linear in distribution
  • They present as skin colored or red-brown, firm papules and nodules
  • Pain and cold sensitivity are common complaints
  • Genital leiomyomas may be pedunculated and are less likely to be painful compared to other subtypes
Diagnosis
  • Diagnosis is often made by histopathology
  • Imaging including computed tomography (CT), magnetic resonance imaging (MRI) and ultrasound can be performed
  • Germline mutations of the fumarate hydratase gene are diagnostic of multiple cutaneous and uterine leiomyomatosis (also known as Reed syndrome or hereditary leiomyomatosis and renal cell cancer)
Laboratory
Not relevant to cutaneous leiomyomas
Radiology description
  • Imaging is not routinely performed for cutaneous leiomyomas
  • Characteristic imaging findings of angioleiomyomas have been described
  • On ultrasound, angioleiomyomas are often described as well defined, solid, round and homogenous; echogenicity and vascularity are variable; Doppler ultrasound can be used to evaluate vasculature and presence of feeding vessels (Korean J Radiol 2018;19:752)
  • Signal intensity of angioleiomyomas on MRI can be similar or hyperintense compared to skeletal muscle on T1 weighted images; a dark reticular signal on T2 weighted images has been proposed as a characteristic feature of angioleiomyomas (Skeletal Radiol 2022;51:837)
  • Calcifications due to degenerative change can be seen on CT
Radiology images

Images hosted on other servers:
MRI findings of angioleiomyoma

MRI findings of angioleiomyoma

Prognostic factors
  • Cutaneous leiomyomas are slow growing and can increase in number over time
  • Excised solitary, sporadic cutaneous leiomyomas have an excellent prognosis with low recurrence rates (J Oral Maxillofac Pathol 2013;17:281)
  • Malignant transformation is rare in cutaneous leiomyomas
  • Leiomyosarcomas have been described in patients with multiple cutaneous and uterine leiomyomatosis (also known as Reed syndrome or hereditary leiomyomatosis and renal cell cancer); subcutaneous extension is a poor prognostic factor in leiomyosarcoma (JAAD Case Rep 2015;1:150, J Am Acad Dermatol 2014;71:919)
  • Presence of multiple cutaneous piloleiomyomas is cause for further work up due to the association of the following malignancies with multiple cutaneous and uterine leiomyomatosis (also known as Reed syndrome or hereditary leiomyomatosis and renal cell cancer)
    • Renal cell cancers, most commonly type II papillary renal cell carcinoma
    • Cutaneous and uterine leiomyosarcomas
    • Cases have been reported of breast cancer, bladder cancer, gastrointestinal stromal tumors, adrenal tumor, testicular and ovarian tumors; however, it is unknown whether these are coincidental or related (GeneReviews: FH Tumor Predisposition Syndrome [Accessed 10 April 2024])
Case reports
Treatment
  • Surgical excision can be performed when practical for symptomatic lesions or for elective removal
  • Conservative but complete excision should be advised if atypical features are present on histopathology
  • Alternative destructive interventions include CO2 laser, electrodessication and cryosurgery
  • Topical, intralesional and systemic treatments for symptomatic relief may be indicated (J Am Acad Dermatol 2017;77:149)
    • Topical options include nitroglycerin, lidocaine and capsaicin
    • Use of intralesional botox has been reported
    • Systemic treatment options include gabapentin, pregabalin, NSAIDs, calcium channel blockers, doxazosin, nitroglycerin, phenoxybenzamine and duloxetine
Clinical images

Images hosted on other servers:
Multiple asymptomatic, hyperkeratotic nodules on leg

Multiple asymptomatic, hyperkeratotic nodules on leg

Gray-brown macules on face

Gray-brown macules on face

Trunk

Trunk

Gross description
N/A
Gross images

Images hosted on other servers:
Well circumscribed mass

Well circumscribed mass

Excised tumor

Excised tumor

Frozen section description
Not relevant to cutaneous leiomyomas
Frozen section images
Not relevant to cutaneous leiomyomas
Microscopic (histologic) description
  • Smooth muscle cells have elongated, thin nuclei with blunt ends; they are often described as cigar shaped with pink, eosinophilic cytoplasm
  • Well circumscribed tumor consisting of fascicles and interlacing smooth muscle bundles
  • Piloleiomyomas are nonencapsulated while angioleiomyomas can be encapsulated
  • Angioleiomyomas have prominent vessels described as slit-like or round; smooth muscle layers surrounding the vessels interlace with surrounding smooth muscle fascicles
  • Overlying epidermal changes such as acanthosis and hyperkeratosis can be seen
  • Cytologic atypia is usually absent and mitotic activity is negligible
  • Presence of nuclear atypia and degenerative features have been described in symplastic leiomyomas (Dermatol Surg 2004;30:1249)
Microscopic (histologic) images

Contributed by Elaine Kunzler, M.D. and Angel Fernandez-Flores, M.D., Ph.D.
Epidermal hyperplasia

Epidermal hyperplasia

Fascicles

Fascicles

Elongated nuclei

Elongated nuclei

Well circumscribed

Well circumscribed


Slit-like vessels

Slit-like vessels

Muscle specific actin

Muscle specific actin

Desmin

Desmin

Virtual slides

Images hosted on other servers:
piloleiomyoma

Piloleiomyoma

angioleiomyoma

Angioleiomyoma

Cytology description
N/A
Cytology images
Not relevant to cutaneous leiomyomas
Immunofluorescence description
Not relevant to cutaneous leiomyomas
Immunofluorescence images
Not relevant to cutaneous leiomyomas
Negative stains
Electron microscopy description
Not relevant to cutaneous leiomyomas
Electron microscopy images
Not relevant to cutaneous leiomyomas
Molecular / cytogenetics description
  • For piloleiomyomas, fumarate hydratase gene mutations can be identified using a variety of methods including sequencing and copy number analyses; a variant specific test can be used if there is a known germline pathogenic variant
  • Fumarate hydratase activity can be measured in fibroblasts or leukocytes (GeneReviews: Fumarate Hydratase Deficiency [Accessed 10 April 2024])
Molecular / cytogenetics images
N/A
Videos

Pilar leiomyoma video
by Dr. Jerad Gardner

Angioleiomyoma video
by Dr. Jerad Gardner

Sample pathology report
  • Skin, right shoulder, shave biopsy:
    • Piloleiomyoma (see comment)
    • Comment: This is a dermal tumor consisting of interlacing fascicles of smooth muscle cells. Cellular atypia and mitoses are not identified. The tumor is positive for SMA.
Differential diagnosis
Board review style question #1

A 25 year old man presents with multiple red-brown, painful papules on the right upper back. On histopathology, there is a dermal nodule with fascicles of spindle cells with eosinophilic cytoplasm. Tumor cells are positive for SMA and desmin and are negative for S100. What is the diagnosis?

  1. Atypical fibroxanthoma
  2. Dermatomyofibroma
  3. Leiomyoma
  4. Neurofibroma
Board review style answer #1
C. Leiomyoma. Answer D is incorrect because neurofibromas are positive for S100 and negative for SMA and desmin. Answer B is incorrect because the spindled cells are horizontal, not arranged in fascicles as in dermatomyofibroma. Answer A is incorrect because the well circumscribed dermal tumor and patient age point towards a benign diagnosis. In atypical fibroxanthoma, cytologic atypia is more apparent.

Comment Here

Reference: Leiomyoma
Board review style question #2

A 36 year old woman presents for excision of a painful, slowly enlarging nodule on the thigh present for many years. Her father has history of papillary type 2 renal cell carcinoma. Mutation in which of the following enzymes is implicated in this genetic syndrome?

  1. Alpha galactosidase A
  2. Ferrochelatase
  3. Fumarate hydratase
  4. Uroporphyrinogen decarboxylase
Board review style answer #2
C. Fumarate hydratase. Fumarate hydratase is mutated in multiple cutaneous and uterine leiomyomatosis (also known as Reed syndrome or hereditary leiomyomatosis and renal cell cancer). Answer A is incorrect because alpha galactosidase A is mutated in Fabry disease. Answer B is incorrect because ferrochelatase is mutated in erythropoietic protoporphyria. Answer D is incorrect because decreased uroporphyrinogen decarboxylase function is associated with porphyria cutanea tarda.

Comment Here

Reference: Leiomyoma

Leukemia cutis
Definition / general
  • Skin involvement ("leukemia cutis") occurs in 5% with CML, 8% with CLL, 10% with monocytic leukemia
  • Myeloid leukemia with monocytic differentiation more commonly involves the skin than other types of myeloid leukemia
  • Usually is abnormal peripheral blood count at diagnosis
  • Skin involvement is rarely initial manifestation of recurrence (Am J Clin Pathol 2008;129:130)
  • May also have accompanying vasculitis (Am J Clin Pathol 1997;107:637)
  • Aggressive behavior and short survival (J Am Acad Dermatol 1999;40:966)
Case reports
Treatment
  • Systemic chemotherapy directed at eradicating the leukemic clone
Clinical images

Contributed by Mark R. Wick, M.D.

Chronic lymphocytic type

Myeloid type

Gross description
  • Multiple nodules / papules
Microscopic (histologic) description
  • In CLL, may be perivascular, periadnexal, nodular or band-like dermal infiltrate
  • Infiltrate in leukemic patients is often NOT neoplastic, but reactive
  • AML: dermis and superficial subcutaneous fat are diffusely infiltrated by a monotonous population of large cells with a high nuclear to cytoplasmic ratio, round to slightly irregular nuclear contours, finely dispersed chromatin and prominent nucleoli
Microscopic (histologic) images

Case #140

AML


CD45

CD43

CD117

CD68



Contributed by Mark R. Wick, M.D.

Myeloid type, granulocytic sarcoma


Myeloid type

CD43

Myeloperoxidase

Leder

Positive stains
  • Myeloblasts: chloroacetate esterase (Leder stain), myeloperoxidase
Additional references

Lobular capillary hemangioma
Definition / general
  • Very common; rapidly growing polypoid red mass surrounded by thickened epidermis, often in finger or lips
  • Also called granuloma pyogenicum, lobular capillary hemangioma
  • May be associated with keratinous cyst
  • Benign, often regresses spontaneously
  • May be disseminated, occur within port wine stains, be in deep dermis / subcutis or be intravenous

Variants:
  • Classic polypoid, dermal, subcutaneous, intravenous, eruptive, with multiple satellites
Treatment
  • None or excision (may recur as multiple satellites)
Clinical images

Contributed by Mark R. Wick, M.D. and @JMGardnerMD on Twitter

Pyogenic granuloma

Lobular capillary hemangioma

Lobular capillary hemangioma

Gross description
  • Fleshy cutaneous tumor
Microscopic (histologic) description
  • Lobular pattern of vascular proliferation with inflammation and edema resembling granulation tissue
  • Thin epidermis at top with variable ulceration
  • Acanthosis and hyperkeratosis at sides
  • Central branching vessel is called capillary or vascular lobule, with no / rare red blood cells, surrounded by endothelial cells
  • Variable mitotic activity
  • Deep lesions often lack edema and inflammation
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D. and @JMGardnerMD on Twitter

Pyogenic granuloma

Lobular capillary hemangioma Lobular capillary hemangioma Lobular capillary hemangioma

Lobular capillary hemangioma



Images hosted on other servers:

Low magnification

Differential diagnosis

Lymphoepithelioma-like carcinoma
Definition / general
  • Exceptionally rare and poorly differentiated cutaneous carcinoma with prominent reactive inflammatory infiltrate
    • Mimics undifferentiated nasopharyngeal carcinoma (Rare Tumors 2013;5:e47)
    • WHO defines these tumors (in nasopharynx/sinonasal cavity) as lymphoepithelial carcinoma
    • Importantly, cutaneous LELC is NOT associated with Epstein Barr virus (EBV) infection
Epidemiology
Sites
  • Head and neck most common site
Pathophysiology
Etiology
  • Uncertain origin (Dermatol Online J 2008;14:12)
  • Unclear whether a poorly / undifferentiated squamous cell carcinoma versus poorly differentiated adnexal neoplasm
Clinical features
Case reports
Treatment
  • Wide local excision or Moh's microsurgery to ensure complete removal
  • Radiation reserved for recurrence or lymph node involvement (Case Rep Oncol Med 2012;2012:241816)
  • Generally low recurrence and metastatic potential with complete excision (in contrast to nasopharyngeal counterpart)
  • Rare lymph node metastasis (Am J Dermatopathol 2006;28:211) or death from disease
Clinical images

Images hosted on other servers:
Missing Image Missing Image

Forehead tumor (pre- and post-excision)

Microscopic (histologic) description
  • With Diagnostic criteria:
    • Well circumscribed lobules, nests or small aggregates of large, cohesive, epithelioid cells closely associated with a dense, mixed T and B lymphocytic and plasmacytic infiltrate (Rare Tumors 2013;5:e47)
      • Biphasic nature may be more apparent at high magnification
    • The epithelioid component generally lacks connection with the epidermis
      • Cells are often polygonal and display poorly defined eosinophilic cytoplasm, vesicular nuclei with prominent nucleoli and increased mitotic activity, including atypical mitotic figures (Dermatol Online J 2008;14:12)
      • Focal ductular differentiation or trichilemmal keratinization rarely reported (J Cutan Pathol 1991;18:93)
    • Mandatory dense inflammatory infiltrate (polymorphous and polytypic) surrounding and intermingling with epithelial tumor cells
      • Lymphoid infiltrate may obscure the epithelial component
      • When inflammation predominates, may mimic lymphoproliferative disorder
Microscopic (histologic) images

Contributed by Sara Shalin, M.D., Ph.D.
Missing Image

LELC



Images hosted on other servers:
Missing Image

Aggregates of atypical epithelial cells

Missing Image Missing Image Missing Image

Vulvar infiltrate with nodal micrometastasis

Missing Image

No epidermal infiltration


Missing Image Missing Image

Atypical, epithelioid cells in syncytial pattern

Missing Image

Round to polygonal cells with eosinophilic cytoplasm

Missing Image Missing Image

Proliferation of epithelial tumor cells and numerous small lymphocytes

Missing Image

H&E, CK5/6, EBV


Missing Image Missing Image

AE1/AE3, EMA

Missing Image Missing Image

CD20, CD3

Missing Image

CK5/6+

Positive stains
  • CK AE1/AE3 (pancytokeratin), p63, EMA, +/- CK5/6
  • Intraepithelial lymphocytes are typically a mixture of CD20+ B cells and CD3+ T cells with no loss of T cell antigens (i.e. preserved CD2, CD5 and CD7), polytypic plasma cells (both kappa and lambda expressing subsets)
Negative stains

Lymphomatoid papulosis
Definition / general
Essential features
ICD coding
  • ICD-O: 9718/1 - lymphomatoid papulosis
Epidemiology
Sites
Etiology
  • No etiologic factors identified (e.g., not associated with EBV)
  • One case report associated with HTLV-1 (Eur J Dermatol 2016;26:194)
  • Some cases that show progression to anaplastic large cell lymphoma show resistance to CD30 ligand and mutations of TGF-β (Semin Diagn Pathol 2017;34:22)
Clinical features
Diagnosis
Prognostic factors
Case reports
Treatment
Clinical images

Contributed by Roberto N. Miranda, M.D.
Sites of involvement

Sites of involvement

Dermoscopy evolution

Dermoscopy evolution

Single nodular lesion 6 mm in diameter

Fully developed lesion with multiple papules in leg

Papule with central crust

Post inflammatory macule

Microscopic (histologic) description
  • Lymphomatoid papulosis is characterized by a wedge shaped pattern, with a wide superficial base and the tip at the bottom, usually deep dermis and less frequently into the subcutaneous tissue
  • The most characteristic appearance is the presence of few to numerous large cells with a Hodgkin or Hodgkin-Reed Sternberg admixed with a reactive background of small lymphocytes and less frequently eosinophils, plasma cells and histiocytes
  • The microscopic appearance and immunophenotype of large cells is variable and led to subclassification of lymphomatoid papulosis into different categories (J Am Acad Dermatol 2016;74:59, Blood 2019;133:1703, Semin Diagn Pathol 2017;34:22, J Am Acad Dermatol 2013;68:809, J Am Acad Dermatol 2012;66:928, Am J Clin Pathol 2003;119:731, Am J Surg Pathol 2010;34:1168, Am J Surg Pathol 2013;37:1)
    • Type A: wedge shaped and extensive lymphoid infiltrate with neutrophils, eosinophils and histiocytes; CD30+ cells are scattered and the overall histologic appearance mimics classic Hodgkin lymphoma
    • Type B: epidermotropism and band-like distribution of small to medium atypical lymphocytes with cerebriform nuclei, without CD30+ expression and mimics mycosis fungoides, patch stage
    • Type C: sheets of large cells, uniformly positive for CD30, with or without epidermotropism and few admixed inflammatory cells; the lesion mimics primary cutaneous anaplastic large cell lymphoma
    • Type D: atypical small to medium lymphoid infiltrate with epidermotropism; the neoplastic cells express CD30 and CD8 and mimic pagetoid reticulosis
    • Type E: angiocentric and angiodestructive pleomorphic lymphoid infiltrate of small to medium size lymphocytes; there are scattered large cells, positive for CD30 and the overall appearance is that of an aggressive lymphoma such as extranodal T/NK cell lymphoma, nasal type; EBER is negative
    • Lymphomatoid papulosis with DUSP22-IRF4 rearrangement: biphasic growth pattern with pagetoid reticulosis-like epidermotropism of small to medium size cerebriform lymphocytes that lack CD30; the second component is dermal or periadnexal and the atypical lymphocytes express CD30+
    • Rare forms
      • Folliculotropic: perifollicular infiltrate of atypical lymphocytes, cystic dilatation of hair follicle, rupture of hair follicle, hyperplasia of the follicular epithelium, intrafollicular pustules (neutrophil collections) and follicular mucinosis
      • Syringotropic: eccrine units with periglandular infiltrate
      • Granulomatous: mononuclear infiltrate with perivascular, eccrinotropic and neurotropic distribution associated with noncaseating granulomas
Microscopic (histologic) images

Contributed by Roberto N. Miranda, M.D.

Higher cellularity towards upper dermis

LyP type A with RS-like cells

LyP type A and CD30+

LyP type C

LyP type C with sheets of large cells


LyP type C and anaplastic morphology

LyP type C and CD3+

LyP type C and CD4+

LyP type C and CD8-

LyP type C and CD30+


LyP type C and TIA1+

LyP type E and angiocentricity

LyP type E and CD30+

LyP with DUSP22-IRF4 rearrangement

Positive stains
Negative stains
Immunohistochemistry & special stains
Immunohistochemical Summary of LyP Types
Type of LyP Reference CD2 CD3 CD4 CD5 CD7 CD8 CD30 Other markers
Type A Blood 2019;133:1703 +/- + + +/- +/- - + TIA1+
Type B Blood 2019;133:1703 +/- + + +/- +/- - - TIA1+
Type C Blood 2019;133:1703 +/- + + +/- +/- - + TIA1+
Type D Am J Surg Pathol 2010;34:1168 + + - - - + + TIA1+, βF1+, CD56-, EBER-
Type E Am J Surg Pathol 2013;37:1 + + -/+ + +/- + + CD45RA-, CD45RO+/-, TIA1+, βF1+, EBER-
LyP with DUSP22-IRF4 rearrangement Am J Surg Pathol 2013;37:1173 +/- + - +/- +/- +/- + CD15+, TIA1-, Ki67 (> 80%), TCR-β+
Folliculotropic LyP J Am Acad Dermatol 2013;68:809 + + + +/- - - +
Granulomatous LyP Am J Clin Pathol 2003;119:731 + + + + - - +
*All cases are anaplastic lymphoma kinase - 1 (ALK-1) negative
Molecular / cytogenetics description
  • Monoclonal rearrangement of the T cell receptor (TCR) is usually detected (Semin Diagn Pathol 2017;34:22)
  • Recurrent NPM-TYK2 gene fusion induces activation of STAT signaling pathway (Blood 2014;124:3768)
  • DUSP22-IRF4 rearrangement cases have characteristic features (Am J Surg Pathol 2013;37:1173)
    • 6p25.3 translocation
    • More common in older patients
    • Localized lesions and clinical presentation suggesting benign inflammatory dermatoses or epithelial tumors
    • Histologically has a bimodal pattern, with epidermotropic cells appearing small and uniform while dermal cells are large and CD30+
Sample pathology report
  • Skin of left ear, excisional biopsy:
    • CD30 positive T cell lymphoproliferative disorder, with extensive ulceration and necrosis, most consistent with lymphomatoid papulosis (see comment)
    • Comment: According to outside pathology report, the patient has a history of left ear abscess. No other clinical history is available at this time.
      One routinely stained slide shows four sections of squamous lined tissue with underlying lymphoid neoplasm and minimal remnants of skin that includes sebaceous glands and hair follicle. There is extensive ulceration and a dense diffuse infiltrate of large cells with vesicular nuclei, irregular nuclear outlines and occasional prominent nucleolus. Few mitotic figures are noted. Extensive coagulative necrosis, as well as suppurative necrosis is noted. Rare areas show perivascular viable cells surrounding by extensive necrosis.
      The immunohistochemical studies reveal the large neoplastic cells are positive for CD2, CD3, CD4, CD5, CD30 and a subset were positive for CD117 and TIA1. The aberrant cells are negative for CD10, CD20, ALK-1, PAX-5, cytokeratin, CD7, CD8, CD56, ALK and EBER.
      The features of this lesion are consistent with primary cutaneous CD30 positive T cell lymphoproliferative disorder, that encompasses lymphomatoid papulosis and cutaneous anaplastic large cell lymphoma. The final diagnosis of this relies on the clinical history. The differential diagnosis includes peripheral T cell lymphoma or ALK negative anaplastic large cell lymphoma with cutaneous involvement or transformation of mycosis fungoides. To exclude these possibilities, clinical correlation and staging studies are recommended.
Differential diagnosis

LyP Type A Primary cutaneous anaplastic large cell lymphoma
Reference Blood 2019;133:1703 Am J Dermatopathol 2017;39:877
Age Fifth decade Seventh decade
Gender (M:F) 2 - 3:1 2 - 3:1
Clinical Features Papular, papulonodular or nodular, < 2 cm Localized nodule or papule, usually > 2 cm
Microscopy Wedge shaped and large lymphoid infiltrate with neutrophils, eosinophils and histiocytes Diffuse and uniform dermal infiltrate of anaplastic large cells
Immunophenotype CD3+, CD4+, CD8-, CD30+ CD3+/-, CD4+, CD8-, CD30+
Treatment None for spontaneous regression; symptomatic treatment for relief Excision, radiation or chemotherapy, alone or combined
Outcomes Excellent Excellent

LyP Type B Early stage mycosis fungoides
Reference Blood 2019;133:1703 An Bras Dermatol 2018\;93:546
Age Fifth decade Sixth decade
Gender (M:F) 2 - 3:1 5:4
Clinical Features Papular, papulonodular or nodular Patches and plaques
Microscopy Epidermotropism and band-like distribution of small / medium atypical lymphocytes with cerebriform nuclei (MF-like) Cerebriform cells with epidermotropism, predominate along basal layer of epidermis
Immunophenotype CD4+, CD8-, CD30- CD4+, CD7-, CD8-, CD30-
Treatment Spontaneous regression or symptomatic treatment Ultraviolet light
Outcomes Excellent Good

LyP Type C Primary cutaneous anaplastic large cell lymphoma
Reference Blood 2019;133:1703 Am J Dermatopathol 2017;39:877
Age Fifth decade Seventh decade
Gender (M:F) 2 - 3:1 2 - 3:1
Clinical Features Papular, papulonodular or nodular; < 2 cm Localized nodule or papule, > 2 cm
Microscopy Sheets of large lymphocytes with or without epidermotropism and few admixed inflammatory cells Diffuse dermal infiltrate of anaplastic morphology
Immunophenotype CD4+, CD8-, CD30+ (uneven) CD3+/-, CD4+, CD8-, CD30+ (uniform)
Treatment Spontaneous regression or symptomatic treatment Radiation or chemotherapy, alone or combined
Outcomes Excellent Good

LyP Type D Primary cutaneous CD8+ aggressive epidermotropic cytotoxic T cell lymphoma
Reference Am J Surg Pathol 2010;34:1168 Mod Pathol 2017;30:761
Age Third decade Fourth decade
Gender (M:F) 6:3 5:2
Clinical Features Papules and small nodules Ulcerative patches or plaques
Microscopy Epidermotropism with pagetoid reticulosis-like Small to intermediate size lymphocytes with epidermotropism
Immunophenotype CD2+, CD3+, CD4-, CD8+, CD30+, TIA1+ CD2-, CD3+, CD7+, CD8+, CD30-, CD56-
Treatment Spontaneous regression or symptomatic treatment Chemotherapy, stem cell transplant
Outcomes Excellent Poor

LyP Type E Extranodal NK/T cell lymphoma, nasal type
Reference Am J Surg Pathol 2013;37:1 Blood 2019;133:1703, Curr Hematol Malig Rep 2016;11:514
Age Sixth decade Fifth and sixth decade
Gender (M:F) 3:1 2:1
Clinical Features Recurrent papular lesions with rapid progression to hemorrhagic necrotic ulcers Necrotic lesions
Microscopy Angiocentric and angiodestructive pleomorphic lymphoid infiltrate of small / medium size Atypical lymphocytes; angiocentric and angiodestructive in most cases
Immunophenotype CD2+, CD3+, CD4-/+, CD8+, CD30+, TIA1+ CD3-, CD4-, CD5-, CD8-, CD30+, CD56+, TIA1+, EBER+
Treatment Spontaneous regression or symptomatic treatment Progressive disease
Outcomes Excellent Poor

LyP with DUSP22-IRF4 rearrangement Transformed mycosis fungoides
Reference Am J Surg Pathol 2013;37:1173 Pathology 2014;46:610
Age Eighth decade Seventh decade
Gender (M:F) 9:2 1.1:1
Clinical Features One or multiple eruptive and papulonodular lesions in a single body area Patches, plaques and tumor
Microscopy Biphasic growth pattern and epidermotropism and periadnexal small to medium size cerebriform cells Cerebriform with increased large cells in dermis
Immunophenotype CD3+, CD4-, CD8+/- CD3+, CD4-, CD7-, CD8-, CD30+/-
Treatment Spontaneous regression or symptomatic treatment Radiation, chemotherapy
Outcomes Excellent Poor

Folliculotropic LyP Folliculitis
Reference J Am Acad Dermatol 2013;68:809 Folliculitis
Age Sixth decade Variable
Gender (M:F) 9:2 Variable
Clinical Features Follicular pustules Superficial folliculitis
Microscopy Perifollicular infiltrate of atypical lymphocytes and follicular mucinosis Moderate mixed inflammatory cells in the follicular ostium
Immunophenotype CD3+, CD4+, CD30+ Nonclonal
Treatment Spontaneous regression Topical treatment
Outcomes Excellent Excellent

Granulomatous LyP Discoid lupus erythematous
Reference Am J Clin Pathol 2003;119:731 Arch Dermatol 2009;145:249
Age Fifth decade Fifth decade
Gender (M:F) 5:4 1:2
Clinical Features Nodules Discoid lesions
Microscopy Noncaseating granulomas associated with mononuclear infiltrate with perivascular, eccrinotropic and neurotropic distribution Edema and a mild infiltrate of inflammatory cells (lymphocytes) in upper dermis
Immunophenotype CD3+, CD4+, CD5-, CD7-, CD8-, CD30+ CD3+, CD4+; no loss of T cell antigens
Treatment Spontaneous regression Systemic agents, Phototherapy
Outcomes Excellent Excellent
Board review style question #1
Which is the rearrangement that defines a type of lymphomatoid papulosis?

  1. ETV6-ABL
  2. EML4-ALK
  3. DUSP22-IRF4
  4. NPM-TYK2
Board review style answer #1
C. DUSP22-IRF4

Comment here

Reference: Lymphomatoid papulosis
Board review style question #2
Which LyP types are usually associated with a cytotoxic phenotype?

  1. Type A and B
  2. Type B and C
  3. Type C and D
  4. Type D and E
Board review style answer #2
D. Type D and E

Comment here

Reference: Lymphomatoid papulosis

Melanoacanthoma
Definition / general
  • Benign pigmented lesion composed of melanocytes and keratinocytes
  • Best regarded as a variant of seborrheic keratosis
  • Oral melanoacanthoma or melanoacanthosis is clinically distinct from cutaneous melanoacanthoma
    • Oral melanoacanthoma is likely reactive, possibly a form of postinflammatory hypermelanosis, and not a tumor-like proliferation
    • It is not discussed as part of this topic
Case reports
Clinical images

Images hosted on other servers:
Various images

Oval lesion of maximum transverse diameter 10 cm

Ulcerated plaque Ulcerated plaque

Ulcerated plaque

Gross description
  • Pigmented plaque, papule or nodule with dull surface
  • Usually solitary and slow growing lesion on head and neck or trunk of older individuals
  • Can clinically mimic a seborrheic keratosis or melanoma
  • Large variants have been described, up to 3 cm or more
Microscopic (histologic) description
  • Resembles a seborrheic keratosis with acanthotic or slightly verrucous epidermis containing small cuboidal keratinocytes and numerous dendritic melanocytes
  • The dendritic melanocytes are heavily pigmented with abundant melanin granules
  • Most melanin pigment is contained within the dendritic melanocytes with little transfer to adjacent keratinocytes
  • Dendritic melanocytes express usual markers of melanocytic differentiation (i.e. S100, HMB45, MelanA)
  • Often has overlying compact eosinophilic parakeratosis even when not irritated
  • No significant cytologic atypia present
Microscopic (histologic) images

Images hosted on other servers:
Hyperkeratosis and papillomatosis

Hyperkeratosis and papillomatosis

Hyperkeratosis and acanthosis

Hyperkeratosis and acanthosis

Basaloid cells

Basaloid cells

Masson-Fontana

Masson-Fontana

Electron microscopy description
  • Composed of benign keratinocytes and dendritic melanocytes
  • Incomplete transfer of pigment from melanocytes to keratinocytes, normal population of Langerhan cells (J Cutan Pathol 1978;5:127)
Differential diagnosis
  • Melanoma:
    • While the dendritic melanocytes in melanoacanthoma might be confused for intraepidermal spread of melanocytes, the melanocytes lack atypia and are part of a background keratinocytic proliferation
  • Pigmented hidroacanthoma / poroma:
    • Contains discrete smooth bordered intraepidermal cellular aggregates with ductal differentiation
    • Melanin pigmentation not restricted to pigmented melanocytes
  • Pigmented squamous cell carcinoma in situ:
    • Full thickness intraepidermal keratinocytic atypia with mitoses and pigment in keratinocytes
  • Seborrheic keratosis:
    • Melanoacanthoma is a variant of seborrheic keratosis so the distinction is not critical
    • Some might sign out melanoacanthoma as seborrheic keratosis, or might use melanoacanthoma with the additional descriptor of 'benign variant of seborrheic keratosis with prominent dendritic melanocytes'

Meningothelial hamartoma of scalp
Definition / general
  • Scalp nodule that often arises during infancy containing meningothelial cells; may be diagnosed later in life
  • Also called rudimentary meningocele or hamartoma of scalp with ectopic meningothelial elements
  • Related to meningioma of skin
Case reports
Microscopic (histologic) description
  • Whorls or dissecting pattern of meningothelial cells in dermis and subcutis
  • Often psammoma bodies and cartilage
  • Center of lesion may have long cleft
  • Adipose tissue, blood vessels, nerves admixed with lesion
Microscopic (histologic) images

Images hosted on other servers:

H&E and immunostains

Positive stains

Merkel cell carcinoma
Definition / general
  • Merkel cell carcinoma (MCC) is an uncommon and highly aggressive primary cutaneous neuroendocrine carcinoma
Essential features
  • Merkel cell carcinoma is a highly aggressive primary cutaneous neuroendocrine carcinoma primarily affecting elderly and immunosuppressed individuals
  • Diagnosis requires microscopic evaluation as the clinical appearance is nonspecific and can mimic a variety of benign and malignant skin lesions
  • There is emerging evidence of distinct Merkel cell polyomavirus associated and UV mediated oncogenetic pathways
  • Distinction between primary cutaneous Merkel cell carcinoma and metastatic neuroendocrine carcinoma in the skin requires immunohistochemical and clinical pathologic correlation
Terminology
  • Originally described as trabecular carcinoma by Toker in 1972
ICD coding
  • ICD-O: 8247 / 3 - Merkel cell carcinoma
  • ICD-10: C4A - Merkel cell carcinoma
Epidemiology
Sites
Pathophysiology
  • Emerging evidence of distinct oncogenetic pathways: majority of cases (up to ~80%) are associated with Merkel cell polyomavirus while minority are UV mediated (Eur J Cancer 2017;71:53)
Etiology
  • Risk factors: advanced age, chronic sun exposure, fair skin, immune suppression (e.g. organ transplantation, HIV infection, chronic lymphocytic leukemia, autoimmunity) (Head Neck Pathol 2018;12:31)
Clinical features
  • Painless and rapidly growing flesh colored or red-violaceous nodule
  • AEIOU acronym for clinical characteristics (J Am Acad Dermatol 2008;58:375):
    • Asymptomatic / lack of tenderness
    • Expanding rapidly
    • Immune suppression
    • Older than age 50
    • UV exposed site
Diagnosis
  • Biopsy or surgical excision
  • Distinction between primary cutaneous Merkel cell carcinoma and metastatic neuroendocrine carcinoma in the skin is based on the immunohistochemical profile of the tumor and clinical pathologic correlation (i.e. clinically exclude prior malignancy and potential noncutaneous sources of metastases)
Radiology description
  • CT: isoattenuating or hyperattenuating areas in soft tissue
  • MRI: enhances with contrast
  • Fluorodeoxyglucose PET / CT: avid hypermetabolic activity (AJR Am J Roentgenol 2013;200:1186)
Radiology images

Images hosted on other servers:

Fluorodeoxyglucose avid hypermetabolic lesion

Prognostic factors
  • Factors relevant to staging for Merkel cell carcinoma include tumor size, depth of invasion, locoregional nodal involvement and disseminated metastasis
  • Negative clinical prognostic indicators: advanced disease stage at diagnosis (most important), age > 60, male gender, head and neck site, immunosuppression, Merkel cell polyomavirus negative subtype (Curr Opin Oncol 2019;31:72)
  • ~60% present with local disease, ~30% with nodal disease and ~10% with distant disease (Ann Surg Oncol 2016;23:3564)
  • High mortality
  • Rarely, complete spontaneous regression of the tumor can occur (J Cutan Pathol 2016;43:1150)
Case reports
Treatment
  • Multidisciplinary approach with wide local excision and sentinel lymph node biopsy in clinically node negative patients with or without adjuvant radiotherapy
  • Emergence of immune checkpoint blockade therapy, targeting PDL1 / PD1 pathway, for advanced disease (N Engl J Med 2016;374:2542, Lancet Oncol 2016;17:1374)
  • Chemotherapy generally ineffective and reserved for palliation
  • Single modality radiotherapy for inoperable tumors
Clinical images

Contributed by Thai Yen Ly, M.D.

Violaceous,
lobulated, polypoid
cutaneous nodule

Gross description
  • Fleshy to tan-brown lesion with nodular or ill defined silhouette
  • Variable ulceration
Gross images

Contributed by Thai Yen Ly, M.D.

Exo / endophytic tumor on ear

Multinodular tumor of skin

Microscopic (histologic) description
  • Expansile, nodular or diffusely infiltrative tumor within the dermis, variably in subcutis
  • Variable mixture of nodules, sheets, nests and trabeculae of neoplastic cells
  • Intraepidermal tumor (or component) is occasionally present
  • Generally, small round blue cell tumor with high N:C ratio, round / oval nuclei, finely dispersed chromatin (salt and pepper), indistinct nucleoli and scant cytoplasm
  • Conspicuous mitoses and apoptotic bodies
  • Variable nuclear molding and crush artifact
  • Majority of cases display pure neuroendocrine morphology (pure Merkel cell carcinoma)
  • Minority of cases feature neuroendocrine and other elements (combined Merkel cell carcinoma) such as divergent differentiation (e.g. squamous, sarcomatoid) or intimate association with other cutaneous neoplasms (most commonly in situ or invasive squamous cell carcinoma)
Microscopic (histologic) images

Contributed by Thai Yen Ly, M.D.

Undifferentiated small blue round cell tumor

Numerous mitoses and apoptotic bodies

Combined Merkel cell carcinoma

Intraepidermal involvement


CK20

Neurofilament

Chromogranin

Synaptophysin

MCPyV

Virtual slides

Images hosted on other servers:

Merkel cell carcinoma

Cytology description
  • Hypercellular specimen featuring small to medium sized neoplastic cells, dispersed and arranged in cohesive, disorganized groups with nuclear molding
  • Neoplastic cells display uniform, round / oval nuclei with fine stippled chromatin, inconspicuous nucleoli, scant cytoplasm and high N:C ratio
  • Apoptotic bodies, necrotic debris and mitoses are frequent
Cytology images

Contributed by Thai Yen Ly, M.D.

Hypercellular specimen

Cellular crowding and necrosis

Atypical small blue cells

Positive stains
Negative stains
Electron microscopy description
  • Intracytoplasmic membrane bound, dense core neurosecretory granules
  • Paranuclear aggregates of keratin filaments
Electron microscopy images

Images hosted on other servers:

Intracytoplasmic neurosecretory granules

Molecular / cytogenetics description
Sample pathology report
  • Left temple skin, lesion, shave excision:
    • Merkel cell carcinoma (see relevant parameters below)
      • Tumor size: ___ cm (maximum clinical tumor diameter; if unavailable use maximum gross or microscopic measurement)
      • Depth of involvement (dermis only, involvement of fascia, muscle, cartilage or bone or unclear, due to superficial sampling)
      • Morphology (pure or combined)
      • MCPyV status (positive or negative)
      • Lymphovascular involvement (present or absent)
      • Margin status (involved or uninvolved)
  • Left ear, tumor, wide excision:
    • Merkel cell carcinoma (see synoptic report)
  • Refer to the International Collaboration on Cancer Reporting (ICCR) Histopathology Reporting Guide (1st Edition, 2019) and CAP reporting guidelines for Merkel cell carcinoma and Merkel cell carcinoma staging
Differential diagnosis
Board review style question #1
This is a tumor from a 65 year old man with a history of a cutaneous malignancy. The tumor expresses paranuclear AE1 / AE3, neuroendocrine markers and MCPyV. Which is true about the lesion?



  1. Clinical course is indolent
  2. Middle aged females are primarily affected
  3. Morphological features alone cannot distinguish between primary cutaneous disease and metastasis from a noncutaneous source
  4. Nuclear pattern of CK20 expression is classic
  5. Site of predilection is the trunk
Board review style answer #1
C. This is Merkel cell carcinoma. Morphological features alone cannot distinguish between primary cutaneous disease and metastasis from a noncutaneous source. Accurate diagnosis requires immunohistochemistry, clinical investigation and clinical-pathologic correlation. Merkel cell carcinoma primarily affects the sun exposed skin of elderly males and preferentially the head and neck region. The classic pattern of CK20 expression is dot-like and paranuclear. Merkel cell carcinoma is aggressive and associated with high mortality.

Comment Here

Reference: Merkel cell carcinoma
Board review style question #2
Which of the following is typically expressed in Merkel cell carcinoma?

  1. CD45
  2. CK7
  3. CK20
  4. S100
  5. TTF1
Board review style answer #2
C. CK20. The classic pattern of CK20 expression is dot-like and paranuclear. Lack of expression for CK7, TTF1, S100 and CD45 help exclude other entities such as pulmonary neuroendocrine carcinoma, melanoma and lymphoma.

Comment Here

Reference: Merkel cell carcinoma

Metastatic
Definition / general
  • Cutaneous metastases from many internal organ sites or soft tissue sarcomas are uncommon (J Surg Case Rep 2014 Sep 9;2014)
  • Men: usually from lung (25%), colon, melanoma, kidney, upper aerodigestive tract
  • Women: usually from breast (69%), lung, melanoma, kidney, ovary
  • Metastasis is often close to site of primary
Sites
  • Local recurrence / regional metastasis most commonly occurs upon anterior chest wall
  • Scalp is tmost commonly observed distant metastatic site
Case reports
  • 73 year old woman with cutaneous metastases as only sign of extranodal tumor spread (ISRN Surg 2011;2011:902971)
  • 83 year old woman with herpetiform cutaneous metastases from transitional cell carcinoma of bladder (J Clin Pathol 2006;59:1331)
  • 91 year old woman with seven month history of redness and swelling around her right eye (BMJ 2007;334:1228)
Treatment
  • Treat underlying malignancy, possible excision of metastasis
Clinical images

Contributed by Mark R. Wick, M.D.

Metastatic breast carcinoma



Images hosted on other servers:

Cutaneous metastases

Gross description
  • Often multiple, firm, nonulcerated nodules, but may be solitary
Microscopic (histologic) description
  • Poorly differentiated adenocarcinoma within dermis and often underlying subcutaneous tissue
  • Tumor cells in sheets or clusters or as single detached cells
  • Either abortive gland formation or "signet ring" cells (with central, cleared mucin-filled space which compresses and marginalizes the cell nucleus)
  • Tumor cells may show a "single file" appearance (metastatic lobular breast carcinoma)
  • Cytologic atypia in small to large epithelioid cells, often with striking nuclear pleomorphism
  • Tumor cells may produce mucin, or mucinous debris may separate collagen bundles
  • Variable desmoplasia or sclerosis of dermal collagen
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D.

Metastatic breast carcinoma



Images hosted on other servers:

Metastatic bladder urothelial carcinoma

Renal cell carcinoma - metastatic

p63- metastatic adenocarcinoma

Positive stains
Negative stains
  • Negative immunostains for metastatic breast adenocarcinoma are S100 (positive in melanoma), TTF1 (positive in lung adenocarcinoma), CK20 and CDX2 (positive in colon adenocarcinoma)
Differential diagnosis

Microcystic adnexal carcinoma
Definition / general
  • Microcystic adnexal carcinoma is a rare malignant adnexal tumor with sweat duct and follicular differentiation
Essential features
  • Malignant adnexal carcinoma with sweat duct and follicular differentiation
  • Most commonly occurs on the face of elderly White patients
  • Histopathology shows deep tumor extension with perineural invasion and may show the presence of lymphoid aggregates
  • Tumors may be locally aggressive and Mohs micrographic surgery is the mainstay of treatment
Terminology
  • Sclerosing sweat duct carcinoma
  • Malignant syringoma
  • Sweat gland carcinoma with syringomatous features
  • Locally aggressive adnexal carcinoma
  • Reference: Mod Pathol 2008;21:178
ICD coding
  • ICD-10: C44.9 - other and unspecified malignant neoplasm of skin, unspecified
  • ICD-11
    • 2C33 - adnexal carcinoma of the skin
    • XH17P2 - microcystic adnexal carcinoma
Epidemiology
Sites
Pathophysiology
  • Mutations
    • No signature mutation to date
    • TP53 is the most common mutation (22%)
    • JAK1 (17%)
    • Other oncogenes (JAK2, MAF, MAFB, JUN, FGFR1)
    • Individual cases may harbor ultraviolet pattern of mutations
  • References: Mod Pathol 2020;33:1092, JAMA Dermatol 2019;155:1059
Etiology
Clinical features
  • Firm, yellow or flesh colored papule, plaque or nodule
  • Mean size at diagnosis: 2.8 cm
  • Rarely metastasizes
  • Reference: JAMA Dermatol 2019;155:1059
Diagnosis
Prognostic factors
Case reports
Treatment
Clinical images

Images hosted on other servers:
White, irregular papule

White, irregular papule

Multiple yellow papulonodules

Multiple yellow papulonodules

Pink and yellow plaque

Pink and yellow plaque

Microscopic (histologic) description
  • Features of both eccrine and follicular differentiation
  • Poorly circumscribed and deeply infiltrating
  • Extension into the subcutaneous adipose tissue and skeletal muscle
  • Superficial pseudohorn cysts containing lamellar keratin
  • Small aggregates of epithelioid cells with curved and angulated shapes, sometimes referred to as having a tadpole-like morphology
  • Deeper components typically have smaller tumor aggregates and can be a single cell layer thick
  • Some islands of tumor cells will show ductal differentiation with luminal centers lined by a pink cuticle
  • Dense fibrous stroma consisting of thick collagen bundles that surround the tumor aggregates
  • Mild cytologic atypia with often bland appearing cells characterized by monomorphic nuclei and scant cytoplasm
  • Lymphoid aggregates adjacent to or in association with tumor
  • Incidental calcifications can be present
  • Mitotic figures are often absent
  • Perineural invasion is a common feature
  • References: Mod Pathol 2008;21:178, Mod Pathol 2006;19:S93
Microscopic (histologic) images

Contributed by Christopher Cullison, M.D. and Bethany R. Rohr, M.D.
Jagged Tumor Islands

Jagged tumor islands

Lymphoid Aggregate

Lymphoid aggregate

Follicular and Ductal Differentiation

Follicular and ductal differentiation

Skeletal muscle invasion

Skeletal muscle invasion

Incidental Calcification

Incidental calcification

Perineural Invasion

Perineural invasion

Virtual slides

Images hosted on other servers:
Deeply invasive tumor islands

Deeply invasive tumor islands

Positive stains
Videos

Microcystic adnexal carcinoma by Dr. Jerad Gardner

Differential diagnosis
  • Skin, left cheek, punch biopsy:
    • Microcystic adnexal carcinoma (see comment)
    • Comment: The sections reveal a deeply infiltrating carcinoma with ductal and follicular differentiation. There are surrounding lymphoid aggregates and focal perineural invasion.
Differential diagnosis
Board review style question #1

Which immunohistochemical stain is likely to be positive in the tumor shown above?

  1. AR
  2. CK19
  3. CK20
  4. S100
Board review style answer #1
B. CK19. CK19 has a cytoplasmic staining pattern for eccrine glands and stains positive in 70 - 100% of microcystic adnexal carcinomas. Answer A is incorrect because androgen receptor does not stain positive in microcystic adnexal carcinoma; rather, it typically stains sebaceous glands and apocrine glands. Answer C is incorrect because CK20 is a stain for Merkel cells that is typically lost in microcystic adnexal carcinoma. Answer D is incorrect because S100 is not expressed in microcystic adnexal carcinomas.

Comment Here

Reference: Microcystic adnexal carcinoma
Board review style question #2
Microcystic adnexal carcinoma is most likely to have which of the following findings at diagnosis?

  1. Angiolymphatic invasion
  2. Distant organ metastasis
  3. Lymph node metastasis
  4. Perineural invasion
Board review style answer #2
D. Perineural invasion. Perineural involvement is a common feature in microcystic adnexal carcinomas. Answers A - C are incorrect because metastasis of microcystic adnexal carcinoma is exceedingly uncommon and is not reported to have angioinvasion on pathology.

Comment Here

Reference: Microcystic adnexal carcinoma
Board review style question #3
Which of the following features can differentiate microcystic adnexal carcinoma from syringoma and desmoplastic trichoepithelioma?

  1. Depth of invasion
  2. Presence of ductal differentiation
  3. Presence of follicular differentiation
  4. Sclerotic stroma
Board review style answer #3
A. Depth of invasion. Microcystic adnexal carcinoma typically has a depth of invasion to at least the deep reticular dermis and may invade into subcutaneous structures, including adipose tissue, skeletal muscle or bone. Answer B is incorrect because syringoma will also have presence of ductal differentiation. Answer C is incorrect because desmoplastic trichoepithelioma will also have presence of follicular differentiation. Answer D is incorrect because both syringoma and desmoplastic trichoepithelioma can have a sclerotic stroma.

Comment Here

Reference: Microcystic adnexal carcinoma

Mucoepidermoid carcinoma
Definition / general
  • Mucoepidermoid carcinoma
    • Very rare in skin (~30 cases reported)
    • Probable sweat gland origin
    • Resembles similar tumor of salivary gland
    • Usually low to intermediate grade, some higher grade tumors perhaps better classified as adenosquamous carcinoma
  • Adenosquamous carcinoma
Sites
  • Adenosquamous carcinoma
    • Frequent confusion in the literature regarding this entity in the head and neck and high grade mucoepidermoid carcinoma (Int J Clin Exp Pathol 2014;7:1809)
    • More common in organs where adenocarcinoma arises frequently, including stomach, intestines and uterus (Oncol Lett 2014;7:1941)
Pathophysiology / etiology
  • Adenosquamous carcinoma
    • Although its pathogenesis is largely unknown, 4 hypotheses have been proposed:
      • Malignant transformation of both squamous and glandular-like cells originating from pleiotropic epithelial stem cells
      • Tumorigenesis of squamous metaplasia in columnar epithelium
      • Transdifferentiation of adenocarcinoma to squamous cell carcinoma
      • Coexistence of both carcinomas (World J Surg Oncol 2013;11:124)
Diagnosis
  • Adenosquamous carcinoma
    • Neoplasm composed of an admixture or separate areas of squamous cell carcinoma and adenocarcinoma
    • Criteria for squamous cell carcinoma component are 2 or more of these features:
      • Intercellular bridging
      • Keratin pearl formation
      • Parakeratotic differentiation
      • Individual cell keratinization
      • Cellular arrangement showing a pavementing or mosaic pattern
    • Criterion for adenocarcinoma component is demonstration of intracytoplasmic mucin
    • World Health Organization Classification does not require intracytoplasmic mucin for diagnosis of adenocarcinoma in the presence of true glandular formation (Int J Clin Exp Pathol 2014;7:1809)
Case reports
Treatment
  • Adenosquamous carcinoma
    • Surgical excision or Mohs microsurgery are common treatment options (Head Neck Pathol 2011;5:108)
    • Locoregional recurrence is not uncommon
Clinical images

Images hosted on other servers:

Adenosquamous carcinoma

Gross description
  • Mucoepidermoid carcinoma
    • Up to 0.6 cm, ulcerated and nonencapsulated
    • Flesh colored nodules, painless
  • Adenosquamous carcinoma
    • White nodular infiltrate into subcutaneous tissue
Gross images

Images hosted on other servers:

High grade mucoepidermoid carcinoma

Microscopic (histologic) description
  • Mucoepidermoid carcinoma
    • Circumscribed tumor, may not be attached to surface
    • Multilobulated nodulocystic tumor extending throughout dermis, exhibiting glandular and squamoid differentiation
    • Dermal lobules or cystic growth of low grade epidermoid, intermediate, mucinous cells and clear cells
    • Cribiform nests of epidermoid cells contain glandular spaces with mucin
    • Nuclei are mildly atypical and contain scattered mitotic figures
    • Peritumoral fibrosis is common
    • May have focal perineural invasion
  • Adenosquamous carcinoma
    • Infiltrative islands of squamous cell carcinoma with admixed mucin containing glandular structures, adenomatous changes and acinar formation
    • Glandular structures lined by low columnar epithelium, sometimes lined by an eosinophilic cuticle (ductular differentiation)
    • Perineural invasion relatively common (Arch Dermatol 2009;145:1152)
Microscopic (histologic) images

Images hosted on other servers:

Mucoepidermoid carcinoma, site unknown

Mucoepidermoid carcinoma, Alcian blue-PAS

Mucoepidermoid carcinoma, salivary gland

Adenosquamous carcinoma

Cytology images

Images hosted on other servers:

Mucoepidermoid carcinoma in salivary gland

Positive stains
Negative stains
Molecular / cytogenetics description
  • Mucoepidermoid carcinoma
    • CRTC1 rearrangements have been detected in cutaneous MEC (like salivary gland MEC) but translocation t(11,19) or MAML2 rearrangements not seen (unlike salivary gland MEC) (Br J Dermatol 2009;161:925)
Differential diagnosis
Mucoepidermoid carcinoma
  • Adenosquamous carcinoma:
    • High grade tumor, often involves epidermis, adenocarcinoma component is well differentiated
  • Metastatic salivary gland tumor:
    • Usually high grade
  • Mucinous metaplasia

Adenosquamous carcinoma

Multicentric reticulohistiocytosis
Definition / general
  • Rare disorder of women ages 40 - 50, with widespread cutaneous papules and nodules (eMedicine)
  • Often associated with a destructive arthritis and internal malignancy
  • Tumor cells are histiocytes and multinucleated giant cells containing abundant eosinophilic cytoplasm with a "ground glass" appearance
Terminology
  • Formerly called lipoid dermatoarthritis
Epidemiology
  • Usually women 40 - 50 years
Etiology
Clinical features
  • Skin lesions on the hands, especially at the base of the nails
  • Lesions may also be on the face, ears, arms, scalp or mucosal surfaces
  • "Coral beads" and vermicular erythematous lesions bordering nostrils are pathognomonic (J Eur Acad Dermatol Venereol 2001;15:524)
  • Lesions vary from small papules to lesions several centimeters across, and are usually skin colored, yellow or reddish brown
  • Recommended to screen patients for malignancy (Rheumatology 2008;47:1102), since accompanied by neoplasm in 28% of cases
  • May be a paraneoplastic process (J Am Acad Dermatol 1998;39:864), or association with neoplasm may be due to reporting bias (eMedicine)
  • Associated with destructive arthritis
Case reports
Treatment
Clinical images

Images hosted on other servers:

Fingers

Hands

Ear


Various images

Destruction of knee articular surface

Erythematous patches on back

Microscopic (histologic) description
  • Prominent oncocytic histiocytes and multinucleated giant cells with eosinophilic, “ground glass” cytoplasm
Microscopic (histologic) images

Case #153

Low power

Medium / high power

PAS



Images hosted on other servers:

Low power

Medium / high power

H&E, CD68 and vimentin

Positive stains
Differential diagnosis
  • Solitary reticulohistiocytoma: younger age, solitary lesions usually not on digits or face (Am J Surg Pathol 2006;30:521)
  • Epithelioid fibrous histiocytoma: usually < 1 cm on extremities, usually no giant cells, primarily myofibroblastic, not histiocytic
  • Epithelioid sarcoma: deep seated tumor with markedly atypical cells that form granuloma-like clusters with central necrosis; tumor cells are EMA+, keratin+, CD68-
  • Granulomatous inflammation: well formed granulomas and prominent lymphocytes, no large epithelioid histiocytes with eosinophilic glassy cytoplasm
  • Histiocytic sarcoma: typically forms a large mass of epithelioid histiocytes with significant nuclear atypia and mitotic activity
  • Juvenile xanthogranuloma: usually children, has scattered Touton type histiocytic giant cells and numerous eosinophils, but large epithelioid histiocytes are not prominent
  • Rosai-Dorfman disease: associated with adenopathy; histiocytes are pleomorphic and S100+

Multinucleate cell angiohistiocytoma
Definition / general
  • Uncommon cutaneous lesion
  • Vascular and fibrohistiocytic lineage are hypothesized
  • Characterized by odd multinucleated fibroblasts, superficial fibrosis and thickening of superficial papillary dermal vessels
Essential features
  • Presence of odd multinucleated fibroblasts
  • Presence of superficial parallel fibrosis
  • Presence and thickening of superficial papillary dermal vessels
  • Absence of perifollicular fibrosis
ICD coding
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
  • ICD-11: 2F23 - benign dermal fibrous or fibrohistiocytic neoplasms
Epidemiology
Sites
  • Dorsal hands or fingers most frequent; when multiple lesions are found, they are most likely to affect the hands or fingers (J Cutan Pathol 2019;46:563)
  • Lower extremities
  • Trunk or back
  • Head and neck (rare, most often these are fibrous papules with multinucleated stromal cells, not multinucleate cell angiohistiocytoma)
Pathophysiology
  • Currently unknown
  • Role for increased mast cells had been postulated but when mast cell counts were compared with counts of normal skin, no difference was found (J Cutan Pathol 2019;46:563)
  • Role for fibroblasts, given the increased intralesional elastic fibers in generalized multinucleate cell angiohistiocytoma, has been postulated (J Dermatol 2021;48:114)
  • Role for estrogen receptor alpha overexpression has been proposed (Am J Dermatopathol 2010;32:655)
Etiology
  • Unknown
Clinical features
Diagnosis
  • Made on H&E stain with light microscopy in conjunction with clinical findings
Prognostic factors
Case reports
Treatment
Clinical images

Contributed by Simon F. Roy, M.D. and Jennifer M. McNiff, M.D.

Erythematous papules on the trunk

Flesh colored papules

Gross description
Microscopic (histologic) description
Microscopic (histologic) images

Contributed by Simon F. Roy, M.D. and Jennifer M. McNiff, M.D.
Superficial papillary dermal fibrosis

Superficial papillary dermal fibrosis

Prominent papillary dermal vessels

Prominent papillary dermal vessels

Odd multinucleated cells

Odd multinucleated cells

Superficial dermal vessels

Superficial dermal vessels

Positive stains
Negative stains
Sample pathology report
  • Skin, left dorsal hand, punch biopsy:
    • Multinucleate cell angiohistiocytoma (see comment)
    • Comment: Sections show superficial parallel fibrosis, thickened and abundant papillary dermal vessels and stellate multinucleated cells consistent with multinucleate cell angiohistiocytoma.
Differential diagnosis
  • Fibrous papule of the nose:
    • Can also show prominent superficial papillary dermal vessels and multinucleated cells but the fibrosis is not parallel to the epidermis
    • Shows concentric perifollicular fibrosis in most instances, unlike multinucleate cell angiohistiocytoma
    • Much more commonly seen on the face / nose (J Cutan Pathol 2019;46:563)
  • Dermatofibroma:
    • More common on lower extremities
    • Atrophic vascular variant of dermatofibroma may mimic multinucleate cell angiohistiocytoma
    • Fibrosis in dermatofibroma is more storiform, rather than parallel to the epidermis
    • May also show multinucleated cells and factor XIIIa positivity
    • Follicular induction and beta catenin positivity in underlying dermal fibroblasts is frequent in dermatofibroma but absent in multinucleate cell angiohistiocytoma (J Cutan Pathol 2019;46:563)
  • Telangiectasia macularis eruptive perstans:
    • Eosinophils are frequent in the inflammatory infiltrate of telangiectasia macularis eruptive perstans but very rare in multinucleate cell angiohistiocytoma
    • Multinucleated cells in telangiectasia macularis eruptive perstans are slightly different, showing < 3 nuclei, contrarily to multinucleate cell angiohistiocytoma
    • Mast cell counts are elevated in telangiectasia macularis eruptive perstans compared with multinucleate cell angiohistiocytoma or normal skin (J Cutan Pathol 2019;46:563)
Board review style question #1

There are multiple red papules on the dorsal hands with multinucleated stromal cells, thickened superficial papillary dermal vessels and superficial dermal fibrosis arrayed in parallel to the epidermis. What is the best diagnosis?

  1. Dermatofibroma
  2. Fibrous papule
  3. Multinucleate cell angiohistiocytoma (MCAH)
  4. Telangiectasia macularis eruptive perstans (TMEP)
Board review style answer #1
C. Multinucleate cell angiohistiocytoma (MCAH)

Comment Here

Reference: Multinucleate cell angiohistiocytoma
Board review style question #2

Which histologic feature is absent in multinucleate cell angiohistiocytoma?

  1. Concentric perifollicular fibrosis
  2. Multinucleated stromal cells
  3. Prominent and thickened superficial papillary dermal vessels
  4. Superficial dermal fibrosis arrayed in parallel
Board review style answer #2
A. Concentric perifollicular fibrosis. Concentric perifollicular fibrosis is rather a feature of fibrous papule of the nose and is the main histopathological criteria to distinguish these entities, along with knowledge of the anatomical site.

Comment Here

Reference: Multinucleate cell angiohistiocytoma

Mycosis fungoides
Definition / general
  • Peripheral T cell lymphoma derived from mature, post-thymic T lymphocytes
  • Presents as cutaneous patches and can progress to plaques, tumors and erythroderma
  • While most patients experience an indolent course, the lymph nodes, bone marrow and viscera can become involved in advanced stage disease
Essential features
  • Mycosis fungoides (MF) is a clinical diagnosis that requires strong correlation with histopathologic and sometimes molecular findings to exclude benign inflammatory diseases, more aggressive primary cutaneous lymphomas, and extracutaneous lymphomas that can involve the skin
  • Conventional MF begins as eczematoid or psoriasiform patches and plaques that arise in a "bathing trunk" distribution
  • MF often behaves as an indolent lymphoma with excellent overall survival; however, a minority of patients experience disease progression with fungating tumors, erythroderma and extracutaneous spread
  • Biopsies from developed patches and plaques of MF show band-like papillary dermal lymphoid infiltrates with background fibroplasia, lymphocytes tagging the junction and migrating to the epidermis as single cells and Pautrier microabscesses, lymphoid cytologic atypia and an absence of findings that would otherwise suggest a spongiotic, psoriasiform or interface dermatitis; there are many histologic variants
  • Immunohistochemistry is generally more useful to exclude other lymphomas than to confirm a diagnosis of MF
  • Clonality testing can sometimes solidify a diagnosis when findings from pairs of temporally or anatomically separate samples are compared together
Terminology
  • Mycosis fungoides (MF)
  • Cutaneous T cell lymphoma
ICD coding
Epidemiology
  • Higher incidence among men than women
  • Median age at diagnosis in 50s
  • Wide age range, predominantly older adults
  • Children more likely to have the hypopigmented variant
Sites
  • Photoprotected "bathing trunk" distribution involving the buttock, trunk and proximal limbs is found in most cases; however, mycosis fungoides can arise anywhere on the body
Pathophysiology
  • Pathophysiology is incompletely understood due to the clinical and immunophenotypic heterogeneity of this entity
  • Cells of origin are skin honing, effector memory T cells in contrast with the central memory T cell phenotype encountered in Sézary syndrome
  • In skin limited disease, the cells most often show a tissue resident memory T cell phenotype and less frequently a migratory memory T cell phenotype (Sci Transl Med 2015;7:308ra158)
  • Immunological milieu of mycosis fungoides (MF) is enhanced for Th2 gene expression and Th2 associated cytokine production (Am J Hematol 2016;91:151)
  • NFkB has been associated with B cell lymphomagenesis and is constitutively activated in MF (Am J Hematol 2016;91:151)
  • STAT activation, cyclin upregulation and loss of RB1 have been identified in some MF cohorts (Am J Hematol 2016;91:151)
  • Cytogenetic and comparative genomic hybridization have demonstrated loss of 9p21 (including the CDKN2a - CDKN2B locus) in some cases of large cell MF and tumor stage MF (Am J Hematol 2016;91:151)
  • Recurrent chromosomal translocations are rare in comparison with B cell lymphomas (Am J Hematol 2016;91:151)
Etiology
  • Must exclude HTLV1 infection because some cases of adult T cell leukemia / lymphoma closely mimic mycosis fungoides (MF) at presentation
  • Human leukocyte antigen (HLA) type associations and rare familial cases suggest genetic predisposition in some patients
  • Some medications are associated with the development of T cell dyscrasias that resemble MF and resolve when the offending drug is withdrawn
  • Infrequently, drug associated T cell infiltrates persist and behave as MF following agent withdrawal, suggesting that some medications can actually induce MF
Clinical features
  • Must avoid overdiagnosis if equivocal findings, but underdiagnosis may lead to treatments that suppress antitumor immunity and foster disease development
  • Can mimic spongiotic or psoriasiform dermatitis, wax and wane and elude a definitive diagnosis for years to decades
  • Begins as a skin limited disease
  • Classic patches are erythematous and heterogeneous in size and shape with fine scale and variable atrophy
  • Increased risk of other malignancies including synchronous lymphoma
  • Usually indolent, but may progress to infiltrative plaques, tumors and erythroderma reminiscent of Sézary syndrome
  • Lymph node involvement, visceral involvement and rarely bone marrow involvement can occur with disease progression
  • Advanced stage disease is associated with loss of T cell repertoire causing infection and death
  • Variants include poikilodermatous, hypopigmented, granulomatous, folliculotropic, syringotropic and palmoplantar mycosis fungoides
Diagnosis
  • Mycosis fungoides (MF) is a clinical diagnosis and a diagnosis of exclusion, using clinical, histologic and molecular data
  • Must exclude inflammatory dermatitis (in particular spongiotic, psoriasiform and lichenoid diseases),drug induced reactions and dyscrasias, cutaneous dissemination of extracutaneous peripheral T cell lymphoma, indolent cutaneous lymphoproliferative disorder or aggressive primary cutaneous lymphoma
Laboratory
  • Elevated lactate dehydrogenase (LDH) is an independent prognostic marker of worse survival
  • Smear, flow cytometry and clonality testing can detect involvement in the peripheral blood
  • HTLV1 serologic studies may be required to exclude adult T cell leukemia / lymphoma
Prognostic factors
  • Independent prognostic markers of worse survival include clinical stage IV disease, age older than 60 years, large cell transformation and elevated lactate dehydrogenase (LDH) (J Clin Oncol 2015;33:3766)
  • T category is based on the percentage of skin surface involvement and the lesional morphology (patchy, plaque, tumor or erythroderma)
  • N category is based on the presence of dermatopathic lymphadenopathy, lymphoma cells and evidence of clonality in addition to the extent of nodal involvement by lymphoma
  • M category is based on the presence or absence of visceral involvement
  • Peripheral blood staging is based on the quantity of abnormal lymphocytes (blood tumor burden) and presence or absence of clonality
  • Median survival for stage IA (< 10% skin surface involvement by patches or plaques) is ≥ 20 years
  • Median survival beyond stage III (> 80% skin surface confluence / erythroderma) is approximately 5 years
Case reports
Treatment
Clinical images

Contributed by Mark R. Wick, M.D.

Various images

Breast skin



Images hosted on other servers:

Classic distribution

Patches

Plaques

Tumors


Erythroderma

Hypopigmentation

Erythematous plaques

Irregular erythematous patch

Microscopic (histologic) description
  • Early lesions are sometimes histologically indistinguishable from more common inflammatory skin diseases
  • Band-like papillary dermal lymphoid infiltrate
  • Intraepidermal lymphocytes out of proportion with spongiosis (epidermotropism) ± Pautrier microabscesses
  • Lymphocytes tagging the junction and within the epidermis may have haloes and can show variable nuclear pleomorphism, nuclear contour irregularity (cerebriform cytomorphology can be difficult to appreciate on biopsies) and hyperchromasia
  • Dermal reticular fibroplasia with space between single lymphocytes
  • Absences of features that would otherwise suggest a diagnosis of spongiotic dermatitis, drug hypersensitivity reaction, psoriasis or interface dermatitis
  • Tumors can show an absence of epidermotropism and can involve the fibroadipose tissue
  • Large cell transformation should always be reported when present and is defined as cells ≥ 4x size of a small lymphocyte comprising > 25% of total infiltrate
Microscopic (histologic) images

Contributed by Robert E. LeBlanc, M.D. and Mark R. Wick, M.D.
>H&E >H&E

H&E



CD4



Contributed by @DrAldehyde on Twitter
Mycosis fungoides Mycosis fungoides Mycosis fungoides Mycosis fungoides

Mycosis fungoides



Mycosis fungoides Mycosis fungoides Mycosis fungoides Mycosis fungoides Contributed by @DrAldehyde on Twitter (see original post here)"> Mycosis fungoides

Mycosis fungoides

Virtual slides

Images hosted on other servers:

72 year old woman with plaque phase MF

Positive stains
  • In general, immunohistochemistry is more helpful to exclude other lymphomas than to confirm a diagnosis of mycosis fungoides
  • Immunophenotype is variable and can change with large cell transformation and stage progression
  • Mature T cell phenotype (CD45RO+, TCR β+, CD2+, CD3+, CD4+ [frequent], CD8+ [infrequent], CD5+, CD7+) is most commonly observed
  • Loss of CD2 or CD5 favors mycosis fungoides over inflammatory diseases such as eczema and psoriasis
  • Partial loss of CD7 is common; however, this equivocal finding is also present in benign dermatoses
Negative stains
  • TCR γ staining should raise consideration for gamma delta T cell lymphoma; however, there are clinically annotated cases of mycosis fungoides that have exhibited this immunophenotype
  • CD8 and cytotoxic marker (TIA1, granzyme) expression should raise consideration for primary cutaneous CD8+ aggressive epidermotropic T cell lymphoma
  • EBER staining of lesional cells should raise consideration for extranodal NK / T cell lymphoma, nasal type
Molecular / cytogenetics description
  • Clonal rearrangement of the T cell receptor can be helpful if histology is nonspecific and clinical findings are strongly suggestive, but identification of clonality in a single biopsy is considerably less specific than identification of the same clone in multiple biopsies taken at different times or from different sites
  • Clones can persist in pityriasis lichenoides spectrum disease and other T cell dyscrasias that do not behave as lymphoma
Sample pathology report
  • Skin, left flank, punch biopsy:
    • Epidermotropic T-cell infiltrate, suspicious for mycosis fungoides (see comment)
    • Comment: This lesion has Pautrier-type microabscesses comprised of CD4+ T-cells that express TCR-beta and show a loss of CD2 expression. Background features of clinically considered psoriasis are not identified, and a PASd stain is negative for intracorneal fungal elements. No folliculotropism is identified. No evidence of large cell transformation is identified. Correlations with clinical and laboratory findings are essential to confirm the above impression.
Differential diagnosis
  • Erythroderma:
    • Acute graft versus host disease: subtle interface dermatitis with satellite cell necrosis
    • Adult T cell leukemia / lymphoma: distinction often requires correlations with clinical history and serologic studies
    • Atopic dermatitis: lymphocyte exocytosis proportionate to the degree of spongiosis; however, intraepidermal edema with eosinophils also can be present in mycosis fungoides so paired clonality studies may be helpful
    • Chronic actinic dermatitis: clinical history, paired clonality studies
    • Erythrodermic drug reaction: clinical correlation is necessary
    • Paraneoplastic erythroderma: clinical correlation is necessary
    • Pemphigus: an unusual cause of erythroderma; however, an absence of acantholysis and direct / indirect immunofluorescence findings could exclude this unlikely possibility
    • Pityriasis rubra pilaris: clinical correlations can be extremely helpful and additionally, PRP can show alternating ortho and parakeratosis, thickening of suprapapillary plates, follicular plugs and focal acantholysis
    • Psoriasis: dilated and tortuous papillary dermal vessels, attenuation of the suprapapillary plates, confluent parakeratotic crust with neutrophils and attenuation of the granular layer are helpful features but close clinical correlations and paired clonality testing are sometimes necessary
    • Sézary syndrome: clinical correlations are necessary
    • T cell prolymphocytic leukemia: clinical correlations would exclude mycosis fungoides (MF)
  • Patch / plaque:
    • Adult T cell leukemia / lymphoma: clinical correlations, HTLV1 serology
    • Chronic actinic dermatitis: photodistribution should raise doubts about MF; however, paired clonality studies may be necessary in some cases
    • Extranodal NK / T cell lymphoma nasal type: EBER+, angiodestructive
    • Interface dermatitis: vacuolar degenerative changes, keratinocyte necrosis
    • Lichenoid interface dermatitis: colloid body formations
    • Mycosis fungoides-like drug reaction: clinical correlations are required
    • Pityriasis rubra pilaris: clinical correlations can be extremely helpful and additionally, PRP can show alternating ortho and parakeratosis, thickening of suprapapillary plates, follicular plugs and focal acantholysis
    • Primary cutaneous CD8+ aggressive epidermotropic cytotoxic T cell lymphoma: ulcerated plaques and tumors; furthermore, histology will show dense epidermotropic infiltrates of enlarged CD8 positive cells filling the epidermis without Pautrier microabscesses
    • Primary cutaneous gamma delta T cell lymphoma: violaceous and sometimes ulcerated plaques and tumors comprised of TCR gamma+ lymphocytes
      • Some show a predominantly epidermotropic pattern reminiscent of MF
      • Another, potentially more aggressive variant shows adipotropism mimicking tumor stage / large cell MF and subcutaneous panniculitis-like T cell lymphoma
    • Psoriasis: dilated and tortuous papillary dermal vessels, attenuation of the suprapapillary plates, confluent parakeratotic crust with neutrophils and attenuation of the granular layer are helpful features but close clinical correlations and paired clonality testing are sometimes necessary
    • Secondary cutaneous involvement by an extracutaneous peripheral T cell lymphoma: clinical correlations are required
    • Secondary syphilis: vacuolar interface changes, conspicuous endothelial swelling and a preponderance of plasma cells; immunohistochemistry or silver stain may highlight treponemes
    • Spongiotic dermatitis: lymphocyte exocytosis proportionate to the degree of spongiosis; however, intraepidermal edema with Langerhans cells also can be present in mycosis fungoides so paired clonality studies may be helpful
    • T cell prolymphocytic leukemia: clinical correlations would exclude MF
    • Dermatophyte: PASd+ hyphae and yeast present in the stratum corneum
  • Tumor:
Board review style question #1
Which of the following studies best distinguishes mycosis fungoides from adult T cell leukemia / lymphoma?

  1. Biopsy revealing Pautrier microabscesses
  2. History of cutaneous patches at disease onset
  3. HTLV1 serology
  4. Immunohistochemistry with CD25
Board review style answer #1
C. HTLV1 serology. A clinical history, positive HTLV1 serology and a subsequent clinical staging could potentially distinguish mycosis fungoides from adult T cell leukemia / lymphoma. These diseases can show significant clinical and histologic overlap. Both can show CD25 expression.

Comment Here

Reference: Mycosis fungoides
Board review style question #2
Which of the following findings best distinguishes mycosis fungoides from Sézary syndrome?

  1. History of cutaneous patches at disease onset
  2. Involvement of the bone marrow
  3. Presence of atypical lymphocytes on peripheral smear
  4. Presence or absence of epidermotropism from the biopsy
Board review style answer #2
A. History of cutaneous patches at disease onset. The history of patch stage disease preceding the erythroderma distinguishes mycosis fungoides from Sézary syndrome. The remaining options can be seen with both diseases, although involvement of the bone marrow is rare.

Comment Here

Reference: Mycosis fungoides

Mycosis fungoides special variants
Pagetoid reticulosis
  • Indolent, T cell cutaneous proliferative disorder related to mycosis fungoides
  • Also called Woringer-Kolopp disease; disseminated lesions called Ketron-Goodman disease
  • 2:1 male to female incidence
  • Usually solitary lesion on distal extremities
  • Treatment may include electron beam, pharmacological intervention, psoralen plus ultraviolet A (PUVA) photochemotherapy, radiotherapy, or surgery (J Clin Aesthet Dermatol 2010;3:46)
  • Solitary erythematosquamous patch with monomorphic intraepidermal infiltrate of mycosis fungoides-like cells, in Paget's disease type pattern
Other variants
  • Special variants to be added include the WHO-EORTC variants including folliculotropic/adnexotropic MF and granulomatous slack skin syndrome. Rarer variants like pigmented purpura-like etc can be mentioned
Case reports
Clinical images

Images hosted on other servers:

Pagetoid reticulosis: 10 × 10mm plaque

Microscopic (histologic) images

Contributed by Mark R. Wick, M.D.

Pagetoid reticulosis



Images hosted on other servers - pagetoid reticulosis:

Psoriasiform hyperplasia

CD8, CD4

Positive stains
  • Pagetoid reticulosis: either CD4+ CD8+ or CD4- CD8-

Myeloid sarcoma

Myoepithelioma
Definition / general
  • Tumors with myoepithelial cells but no epithelial cells (Hum Pathol 2004;35:14)
  • Usually benign lesions of extremities, but should be completely excised
  • May recur locally, rarely metastasize
  • Usually male, mean age 22 years (range 10 - 63 years)
  • Part of a continuum with mixed tumors (ductal structures but few myoepithelial cells)
  • Mitotic activity may predict more aggressive tumor
Gross description
  • Mean 1 cm, range 0.5 to 2.5 cm
Microscopic (histologic) description
  • Well circumscribed dermal lesions, not connected to epidermis, may extend into superficial subcutis
  • Either composed of:
    • Solid proliferation of ovoid, spindled, histiocytoid or epithelioid cells with abundant eosinophilic syncytial cytoplasm and little stroma; or
    • Reticular architecture with epithelioid, plasmacytoid or spindle cells in myxoid or hyalinized stroma
  • Cells have ovoid / spindled nuclei, mild pleomorphism, small necrotic areas, fatty metaplasia, minimal mitotic figures (0 - 6 / 10 HPF), no ductal differentiation
Positive stains
Differential diagnosis
  • Benign mixed tumor
  • Cellular neurothekeoma: nested architecture, sclerotic dermal collagen, NKI-C3+, S100-, EMA-, keratin-
  • Epithelioid benign fibrous histiocytoma: lower limbs, circumscribed, polypoid, plump and often binucleate epithelioid cells, may entrap dermal collagen, keratin-, myogenic markers-, S100-
  • Epithelioid sarcoma: distal extremities of young adults, infiltrates along fibrous septa and fascial planes, discontinuous growth, S100-, GFAP-
  • Leiomyoma
  • Myoepithelial carcinoma
  • Spitz nevus: large epithelioid melanocytes with prominent nucleoli, junctional component, downward maturation, HMB45+, S100+, EMA-, keratin-, myogenic markers-

Myopericytoma / myofibroma

Neurothekeoma
Definition / general
  • Superficial tumor, first described in 1980
  • Originally, derivation thought to be from nerve sheath; now thought to derive from fibroblasts with ability to differentiate into myofibroblasts and to recruit histiocytes (Am J Surg Pathol 2007;31:1103)
Sites
  • Usually head, upper extremities or shoulder girdle
Clinical features
  • Solitary, superficial, slow growing mass up to 2 cm
  • 60% women, mean age 17 years (range 2 - 85 years), 80% are < age 30 at initial diagnosis
Case reports
Treatment
  • Excision, may recur
Clinical images

Images hosted on other servers:

Thumb nodule

Lower back

Lower eyelid

Microscopic (histologic) description
  • Cellular, myxoid or mixed subtypes
  • Involves dermis or subcutis
  • Multinodular mass with myxoid matrix and peripheral fibrosis
  • Whorled or focally fascicular patterns of spindled and epithelioid mononuclear cells with abundant cytoplasm, indistinct cell borders
  • Margins usually positive; usually occasional multinucleated giant cells
  • Variable nuclear atypia
  • Median 4 MF / 25 HPF, may have 10+ MF / 25 HPF, may be atypical
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D.

MiTF

Cellular type


Cellular type, skin



Images hosted on other servers:

Left thumb nodule of 5 month old boy

Lower eyelid of elderly man

Positive stains
Negative stains
Differential diagnosis

Nevus comedonicus (pending)
[Pending]

Nevus lipomatosus superficialis
Definition / general
  • Nodules of mature fat tissue within dermis
  • Either solitary or multiple
Terminology
  • Also called nevus lipomatosus cutaneous superficialis and dermolipoma
  • Not a WHO diagnosis
  • First described in 1921 by Hoffman and Zurhelle
  • Considered to be a developmental anomaly or hamartoma
Epidemiology
  • Uncommon
  • At birth or by age 20 years
Clinical features
  • Buttocks or upper thighs
  • Either solitary (more common) or multiple type of Hoffmann-Zurhelle (Tunis Med 2006;84:800)
Case reports
Treatment
  • Excision - does not recur
Clinical images

Images hosted on other servers:

Lumbar region

Brown-red nodules

Gross description
  • Soft yellow papules
Microscopic (histologic) description
  • Mature adipocytes in dermis
  • No encapsulation or connection with subcutaneous fat
  • No distinct epidermal changes
Microscopic (histologic) images

Images hosted on other servers:

Dermal adipose tissue is continuous with underlying subcutis

Ectopic fat in mid to lower dermis

Nodule formed by clusters of mature fat cells


Nevus sebaceus of Jadassohn
Definition / general
  • Nevus sebaceus of Jadassohn (sebaceous nevus, organoid nevus) is a hamartoma that is histologically characterized by a complex and abnormal proliferation of epidermis and adnexal structures
  • Clinically manifests as a congenital, yellow-orange mamillated patch / plaque with alopecia typically occurring on the scalp and face
Essential features
  • Hamartomatous proliferation of epidermis and adnexal structures that commonly presents at birth as a single yellow patch of alopecia on the scalp, becomes verrucous during childhood and puberty and shows variable enlargement during adulthood (due to development of a range of often benign and occasionally / rarely malignant tumors)
  • Unusual anatomical presentation and appearance as well as rare association with systemic syndromes (often with neurologic and developmental abnormalities) can occur
  • New genomic data suggest that nevus sebaceus may represent a mosaic RASopathy, often originating from a portion of skin with HRAS / KRAS mutations; this further explains the rare association with other developmental syndromes and intralesional growth of neoplasms
  • The common types of secondary neoplasm include syringocystadenoma papilliferum, trichoblastoma-like tumor, trichilemmoma and sebaceous neoplasms
  • Development of malignant neoplasms (mainly basal cell carcinoma) in nevus sebaceus is a rare phenomenon (< 1% of lesions in a large series) and almost always occurs during adulthood
Terminology
  • Organoid nevus; though sometimes used as a generic term for the group of cutaneous hamartomatous lesions (see epidermal nevus)
  • Nevus sebaceous; a common misspelling combining the Latin term nevus sebaceus with the English equivalent term sebaceous nevus
ICD coding
  • ICD-10: Q82.5 - congenital nonneoplastic nevus
Epidemiology
  • Present at birth or develops in early childhood, with a prevalence of 0.3% in newborns
  • F = M
  • Affects all races and ethnicities
  • Very low rate of malignant transformation, estimated to be ~0 - 0.8% (CMAJ 2019;191:E765)
  • Can be associated with the following syndromes:
    • Phacomatosis pigmentokeratotica
    • SCALP syndrome (sebaceus nevus syndrome, CNS symptoms, aplasia cutis, limbal dermoid and pigmented nevus with neurocutaneous melanosis)
    • Linear sebaceus nevus syndrome, i.e Schimmelpenning–Feuerstein–Mims syndrome (organoid nevi, abnormalities of the central nervous system and eyes, oral lesions and skeletal defects)
  • Up to 19% are associated with a secondary benign lesion (J Am Acad Dermatol 2014;70:332)
  • Reference: Calonje: McKee's Pathology of the Skin, 5th Edition, 2019
Sites
Pathophysiology
  • Nevus sebaceus is now considered as a mosaic RASopathy with postzygotic somatic mutations of the Ras protein family, mainly due to HRAS and rarely KRAS mutations, in a section of the skin
  • RAS mutations are also present in secondary tumors in nevus sebaceus (e.g., trichoblastomas)
  • Reference: Calonje: McKee's Pathology of the Skin, 5th Edition, 2019
Etiology
Clinical features
  • Clinical appearance of the lesion can change over time
    • In infancy, nevus sebaceus is usually a smooth hairless patch
    • During puberty (under hormonal influence), it can undergo rapid growth becoming more verrucous with tightly arranged yellowish waxy papules
    • In adulthood, further enlargement can occur due to the development of intralesional neoplasms
  • Nevus sebaceus, often in the form of a giant lesion involving large areas of head and neck, can be associated with a range of ophthalmic and neurologic anomalies (nevus sebaceus syndrome); seizures and neurologic anomalies have also been reported as part of epidermal nevus syndrome (CMAJ 2019;191:E765, Pediatr Dermatol 1999;16:211)
  • Linear and zosteriform presentations have been described
Diagnosis
  • Usually clinically suspected and diagnosed
  • Skin biopsy may be required for confirmation or assessment of secondary benign or rarely malignant neoplasms developing in the lesion
Prognostic factors
  • Generally, nevus sebaceus will follow a benign course
  • Incidence of development of malignant neoplasms (e.g., basal cell carcinoma, squamous cell carcinoma, sebaceous carcinoma) in nevus sebaceus is extremely rare (< 1%); a significantly higher rate (up to 20%) reported in some older studies is likely due to misclassification and inclusion of trichoblastoma-like tumors (J Dermatol 2016;43:175, CMAJ 2019;191:E765)
Case reports
Treatment
  • Generally safe to observe
  • Surgical excision as a cosmetic procedure or to remove a secondary neoplasm
  • Superficial procedures such as shave removal, dermabrasion or laser resurfacing are typically unsuccessful due to incomplete removal of the lesion (Am J Clin Dermatol 2015;16:197)
Clinical images

Images hosted on other servers:

Yellow-orange plaque with alopecia

Soft elastic, painless formations

Newborn cerebriform nevus sebaceus

Scalp vertex lesion

Gross description
Microscopic (histologic) description
  • Well circumscribed and complex proliferation and alteration of epidermis and adnexal structures that often changes with age
  • Prepubertal lesions are broad with primitive hair follicles and markedly decreased terminal hairs (hair follicles are usually vellus)
  • Sebaceous glands can be increased or decreased based on the age and associated hormonal effect and can show an abnormal distribution and configuration; they can also be located higher than normal in the dermis without connection to a hair follicle and with direct opening onto the epidermal surface
  • Additional features are variably seen and include:
    • Increased acanthosis, papillomatosis and basal epidermal melanin pigmentation
    • Presence of an inflammatory infiltrate
    • Ectopic apocrine glands (up to half of cases and occasionally dilated)
    • Anomalous ductal sweat gland structures resembling eccrine hyperplasia
  • Secondary neoplasms occur, namely trichoblastoma-like tumor, trichilemmoma, syringocystadenoma papilliferum and sebaceous gland neoplasms
  • References: Int J Dermatol 2016;55:193, Calonje: McKee's Pathology of the Skin, 5th Edition, 2019
Microscopic (histologic) images

Contributed by Ronan Knittel, M.D. and Sara C. Shalin, M.D., Ph.D.
Abnormal epidermal / pilosebaceous alterations

Abnormal epidermal / pilosebaceous alterations

Abnormal pilosebaceous units

Abnormal pilosebaceous units

Abnormal sebaceous glands

Abnormal sebaceous glands

Anomalous sweat glands

Anomalous sweat glands

Epidermal acanthosis / papillomatosis Epidermal acanthosis / papillomatosis

Epidermal acanthosis / papillomatosis


Ectopic apocrine glands

Ectopic apocrine glands

Abnormally configured apocrine glands

Abnormally configured apocrine glands

Sebaceous hyperplasia

Sebaceous hyperplasia

Syringocystadenoma papilliferum Syringocystadenoma papilliferum

Syringocystadenoma papilliferum

Trichoblastoma like tumor

Trichoblastoma-like tumor


Basaloid proliferations

Basaloid proliferations

Syringocystadenoma papilliferum

Syringocyst-
adenoma papilliferum

Basaloid buds

Basaloid buds

Immature sebocytes Immature sebocytes

Immature sebocytes

Apocrine glands

Apocrine glands

Virtual slides

Images hosted on other servers:

Nevus sebaceus

Nevus sebaceus

Molecular / cytogenetics description
Videos

Nevus sebaceus in 5 minutes

Nevus sebaceus

Nevus sebaceus with associated benign hair follicle tumors

Nevus sebaceus (not SebaceOus!)

Sample pathology report
  • Skin, scalp, excision:
    • Nevus sebaceus (see comment)
    • Comment: The sections of scalp skin demonstrate a well circumscribed broad area of epidermal and adnexal proliferation. The lesion is characterized by marked epidermal acanthosis, papillomatosis and hyperkeratosis overlying lobules of hyperplastic and abnormally configured sebaceous glands. There are dilated apocrine glands in the dermis with a superficial dermal lymphoplasmacytic inflammation. There is no evidence of an associated neoplastic growth or malignancy in the material examined.
Differential diagnosis
  • Epidermal nevus:
    • Often clinically presents on the extremities, trunk and neck as linear lesions following Blaschko lines
    • Histologically similar epidermal changes but without sebaceous gland alterations
    • Mesa sign (i.e., the tips of the epidermal papillations are flattened)
    • Normal dermis, usually with no associated adnexal structures
  • Sebaceous hyperplasia:
    • Increased sebaceous glands, usually with no primary epidermal changes
  • Seborrheic keratosis:
    • Unusual diagnosis in children / young adults
    • Some overlapping epidermal changes without adnexal / sebaceous alterations
    • Presence of keratin horn cysts and squamous eddies
  • Acanthosis nigricans:
    • Usually in flexural areas of the body, particularly the axilla
    • Associated with an underlying metabolic disorder or malignancy
    • Hyperkeratosis and papillomatosis but mild acanthosis and no dermal inflammation or adnexal / sebaceous alterations
Board review style question #1

A histologic image from an excisional biopsy from the scalp of a 17 month old child is shown above. What is the most likely diagnosis?

  1. Dilated pore of Winer
  2. Nevus sebaceus
  3. Sebaceous adenoma
  4. Sebaceous hyperplasia
  5. Seborrheic keratosis
Board review style answer #1
B. Nevus sebaceus

Comment Here

Reference: Nevus sebaceus of Jadassohn
Board review style question #2

An incisional biopsy from the scalp of a 2 year old boy was performed; the histology is shown above. Which of the following alterations is most likely to occur in the lesion during puberty if not completely excised?

  1. Development of secondary malignant neoplasms
  2. Enlargement due to sebaceous gland hyperplasia
  3. Epidermal atrophy
  4. Spontaneous regression
Board review style answer #2
B. Enlargement due to sebaceous gland hyperplasia

Comment Here

Reference: Nevus sebaceus of Jadassohn

Nevus with “skyline” basal celllayer (PENS) (pending)
[Pending]

Normal nail histology and grossing (pending)
[Pending]

Onychocytic carcinoma (pending)
[Pending]

Onychocytic matricoma (pending)
[Pending]

Onychomatricoma (pending)
[Pending]

Onychopapilloma (pending)
[Pending]

Other cysts
Definition / general
Bronchogenic cyst
  • Bronchogenic cysts presenting in the skin are very rare, with fewer than 70 cases reported

Cutaneous ciliated cyst
  • Also called cystadenoma, cutaneous Müllerian cyst
  • Usually extremities of teenage girls
  • May have Müllerian derivation in females, distinct fetal eccrine duct origin in males
Epidemiology
Cutaneous ciliated cyst
  • Solitary lesion which presents shortly after menarche on limb of young females (12 - 42 years)
  • Also described in males and at atypical sites including back, shoulder, scalp, cheek
Sites
Bronchogenic cyst
  • Most are present at birth on the precordium or overlying the suprasternal notch
  • Occasionally occur near shoulder, back, scapula, neck, abdomen or chin or present at a later age

Cutaneous ciliated cyst
  • Thigh > buttock > calf > foot
Pathophysiology
Bronchogenic cyst
  • Believed to form from buds or diverticula that separate from foregut during development of the tracheobronchial tree
  • May be intrapulmonary or peripheral
  • Cutaneous bronchogenic cysts may result from subsequent sequestration outside the chest cavity following fusion of the mesenchymal bars of the sternum or from active migration prior to fusion

Cutaneous ciliated cyst
  • Lesions on limbs of young females are generally thought to be of Müllerian (paramesonephric) derivation, representing a migration abnormality of fetal development (heterotopia)
  • Cysts arising at other sites and in males may represent metaplasia of lining of a pre-existent simple cyst of sweat duct derivation or have an entirely different histogenesis
Clinical features
Bronchogenic cyst
  • 80% males
  • Variable presentation as cutaneous cystic nodule, sinus or papillomatous growth
  • Usually asymptomatic but may be tender or painful
  • Rarely are multiple
  • Treatment is surgical resection, if clinically indicated

Cutaneous ciliated cyst
  • Located in deep dermis or subcutaneous tissue
  • Usually asymptomatic
Case reports
Bronchogenic cyst
Cutaneous ciliated cyst
Treatment
Cutaneous ciliated cyst
  • Surgical excision
Clinical images

Images hosted on other servers:

Bronchogenic cyst
Sinus in suprasternal notch

Sinus in suprasternal notch

Sinus opening

Sinus opening

Gross description
Bronchogenic cyst
  • Skin nodule

Cutaneous ciliated cyst
  • Soft to cystic, solitary, movable, nontender, fluctuant swelling
Gross images

Images hosted on other servers:

Bronchogenic cyst
Scapular bronchogenic cyst

Scapular bronchogenic cyst


Cutaneous ciliated cyst
Round cyst with blue hue

Round cyst with blue hue

Microscopic (histologic) description
Bronchogenic cyst
  • Cutaneous bronchogenic cyst occurs within dermis or subcutaneous tissue
  • Lining is usually thrown into small folds
  • Epithelium is invariably ciliated, pseudostratified cuboidal or columnar, with mucus secreting goblet cells in 50% of cases
  • May have nonciliated cuboidal, columnar and stratified squamous epithelium
  • Smooth muscle supports the mucosa in 8% of cases
  • Lymphoid follicles are found in 25% of cases and appear to be part of a secondary inflammatory response
  • Occasionally seromucinous glands or cartilage are present
  • Cutaneous lung tissue heterotopia, in which fully developed bronchioles and alveoli are present, is considered a variant

Cutaneous ciliated cyst
  • Unilocular or multilocular cyst with intraluminal papillary projections of lining resembling fallopian tube
  • Cuboidal to columnar ciliated epithelium with frequent pseudostratified foci
  • Deep to the epithelium lie well vascularized parallel bundles of collagen but smooth muscle is not present
  • Occasional: squamous metaplasia, intercalated dark cells
  • Rare: mucin secreting cells, apocrine-like features
Microscopic (histologic) images

Images hosted on other servers:

Bronchogenic cyst
Pseudostratified ciliated columnar epithelium

Pseudostratified ciliated columnar epithelium

Ciliated respiratory epithelium

Ciliated respiratory epithelium

Respiratory and squamous epithelium

Respiratory and squamous epithelium



Cutaneous ciliated cyst
Epithelium-lined cyst in subcutaneous tissue

Epithelium lined cyst in subcutaneous tissue

Cilia on luminal border

Cilia on luminal border

Thin-walled cyst

Thin walled cyst

Focal squamous metaplasia

Focal squamous metaplasia

Cytokeratin+, EMA+

Cytokeratin+, EMA+


PanCK

PanCK

CEA-

CEA-

PR+, ER+

PR+, ER+

ER+

ER+

PR+

PR+

Cytology description
Bronchogenic cyst
Positive stains
Bronchogenic cyst
Cutaneous ciliated cyst
Electron microscopy description
Cutaneous ciliated cyst
  • Ultrastructurally, cilia have characteristic morphology with a central pair of microtubules, 9 radially orientated pairs of microtubules, basal bodies, cross striated rootlets
Differential diagnosis
Bronchogenic cyst
Cutaneous ciliated cyst

Palisaded encapsulated neuroma
Definition / general
  • Also called solitary circumscribed neuroma
  • Common, often on face; often due to trauma or surgery
Terminology
  • Also known as solitary circumscribed neuroma
Gross description
  • Small solitary papule
Microscopic (histologic) description
  • Spindle lesion with palisading, occasionally epithelioid cells, lesion is not truly encapsulated
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D.

Positive stains
Differential diagnosis

Papillary eccrine adenoma
Definition / general
Essential features
  • Solitary nodule, mostly in lower extremity of young black women
  • Well circumscribed in the dermis, composed of tubules with intraluminal papillary, micropapillary or cribriform and peripheral flat myoepithelial cells
  • Nuclear hyperchromasia, single cell necrosis and mitotic figures present
Terminology
ICD coding
  • ICD-10: D23.9 - Other benign neoplasm of skin, unspecified
  • ICD-11: 2F22 - Benign neoplasms of epidermal appendages
Epidemiology
  • Usually occurs in young people
  • Blacks > whites
  • Women > men
Sites
  • Usually extremities, lower more than upper
Pathophysiology
Etiology
  • Unknown
Clinical features
Diagnosis
  • Incisional or excisional biopsy should be performed with histologic examination for a definite diagnosis
Prognostic factors
  • Excellent prognosis; no recurrence following simple conservative complete excision
  • Can have recurrence / persistence after incomplete removal
Case reports
Treatment
  • Simple surgical excision
Microscopic (histologic) description
  • Nodular, well circumscribed and often asymmetrical from scanning magnification
  • Tubular or cystic structures with intraluminal papillary, micropapillary or cribriform growth (common)
  • Ductal lining is bilayered with luminal cuboidal cells and peripheral flat myoepithelial cells
  • Occasional findings
Microscopic (histologic) images

Contributed by Masoud Asgari, M.D. and Sheng Chen, M.D., Ph.D.
Well circumscribed

Well circumscribed

Tubules with micropapillae Tubules with micropapillae

Tubules with micropapillae

Epithelial and myoepithelial layers Epithelial and myoepithelial layers

Epithelial and myoepithelial layers


Necrosis

Necrosis

Mitosis

Mitosis

p63

p63

Ki67

Ki67

Positive stains
Negative stains
Electron microscopy description
  • Cells of the inner layer have features of eccrine secretory (clear) cells
  • Cells of the outer layer have characteristics of both ductal basal cells and glandular myoepithelial cells (J Dermatol Sci 1990;1:65)
Sample pathology report
  • Skin, right foot, excision:
    • Papillary glandular neoplasm (see comment)
    • Margins negative for tumor
    • Comment: The findings are consistent with a papillary eccrine adenoma (also called papillary adenocarcinoma in situ).
Differential diagnosis
Board review style question #1


A 41 year old African American woman with a solitary, 1 cm nodule localized on the right leg comes to the office. Incisional biopsy was performed and the findings are illustrated above. Which of the following statements regarding this neoplasm is correct?

  1. Identical features may be seen in nipple adenoma
  2. It is composed of solid proliferation of both myoepithelial cells and epithelial cells
  3. Many plasma cells are present
  4. Myoepithelial cell layer may be absent at the periphery of some tubular structures
  5. This is an aggressive neoplasm with a high rate of metastasis (15%)
Board review style answer #1
A. Identical features may be seen in nipple adenoma. This is a papillary eccrine adenoma. Nipple adenoma and papillary eccrine adenoma have identical histopathologic features and therefore choice A is correct. So called papillary eccrine adenoma is not an aggressive neoplasm (choice E) but it may recur if it is not completely excised. The lesion was initially considered benign and called adenoma by Rulon and Helwig on the basis of available follow up information. However, adenocarcinoma in situ, when completely removed, behaves in the same way, namely, no recurrence or metastasis. Choice B is a cardinal feature of so called aggressive digital papillary adenocarcinoma. Choice C is a feature of syringocystadenoma papilliferum. Choice D may suggest an invasive focus within the neoplasm, given the absence of myoepithelial layer around some tubular structures.

Comment Here

Reference: Papillary eccrine adenoma
Board review style question #2
Which of the following gene mutations is usually seen in papillary eccrine adenoma?

  1. BRAF V600E
  2. CYLD1
  3. FLCN
  4. MLH1
  5. PTCH
Board review style answer #2
A. BRAF V600E. Mutation in BRAF V600E is frequently seen in papillary eccrine adenoma, similar to its analogue nipple adenoma. Mutation in CYLD1 is usually seen in cylindroma. Mutation in PTCH is usually seen in adenoid basal cell carcinoma. Mutations in MMR genes (such as MLH1, MSH2, MSH6 and PMS2) are associated with sebaceous carcinoma. FLCN may be mutated in fibrofolliculoma / trichodiscoma.

Comment Here

Reference: Papillary eccrine adenoma

Papillary intralymphatic angioendothelioma

Penile intraepithelial neoplasia

Pilomatricoma
Definition / general
  • Pilomatricoma is a benign skin adnexal tumor originating from hair matrix and cortex
Essential features
  • Benign skin adnexal tumor originating from hair matrix and cortex
  • Most commonly occurs in head and neck, extremities and trunk in children
  • Grossly appears as circumscribed hard dermal tumor with cheesy material
  • Most cases behave indolently without recurrences
Terminology
  • Pilomatrixoma
  • Calcifying epithelioma of Malherbe (not recommended)
ICD coding
  • ICD-O: 8110/0 - pilomatrixoma, NOS
  • ICD-11: 2F22 & XH9E37 - benign neoplasms of epidermal appendages & pilomatricoma, NOS
Epidemiology
  • Pilomatricoma, being the most common adnexal neoplasm in children, favors young age (Actas Dermosifiliogr 2010;101:771)
    • However, a wide age range has been described from 3 months to 93 years
  • Females are affected slightly more than males
  • Usually solitary but multiple lesions can occur (Int J Dermatol 2017;56:1032)
Sites
Pathophysiology
  • Pilomatricoma development is associated with mutations in CTNNB1 gene located in the exon 3 region encoding beta catenin, a 92 kDa molecule involved in cell - cell adhesion
  • As an integral component of Wnt signaling pathway, beta catenin monitors cell proliferation, differentiation and survival
  • Beta catenin expression in hair matrix during human embryogenesis signifies its importance in hair follicle development and thus pilomatricoma origin
  • BCL2 oncoprotein is also implicated in its development (Arch Pathol Lab Med 2014;138:759)
Etiology
Clinical features
Diagnosis
  • Generally very hard, owing to calcification; skin lesions that are calcified tend to display positive teeter totter and tent signs
    • Teeter totter sign is observed when applying downward pressure to one end of the lesion causes the other end to spring upwards in the skin
    • Tent sign appears when the skin overlying the lesion is stretched, revealing multiple facets and angles, resembling a tent
  • Clinical examination alone may not be diagnostic (Am J Dermatopathol 2018;40:631)
    • Imaging studies ultrasound, computed tomography and magnetic resonance imaging are helpful; ultrasonography has a high positive predictive value (95.56%) (Arch Craniofac Surg 2020;21:288)
Radiology description
  • Ultrasound
    • Pilomatricoma present as heterogeneous, well demarcated, hypoechoic mass between the deep dermis and the subcutis on ultra high frequency ultrasonography
    • Most common features are internal echogenic foci (calcifying), hypoechoic rim and posterior shadowing (J Ultrasound 2008;11:76, Front Med (Lausanne) 2021;8:673861)
  • CT scan
    • Appear well demarcated, subcutaneous masses with various levels of calcification
    • Inflammatory cell infiltration into fat may result in peritumoral fat stranding on fat suppressed, T2 weighted images (Radiol Med 2019;124:1049)
Radiology images

Contributed by Nasir Ud Din, M.B.B.S.

Plain Xray of leg



Images hosted on other servers:

Well defined, heterogeneous, hyperechoic nodule

Axial and coronal CT scans of cheek

Prognostic factors
  • Rare tumor recurrence of ~1 - 2% is reported with incomplete primary excision (Arch Craniofac Surg 2020;21:288, Am J Dermatopathol 2018;40:631)
  • In rare cases, can undergo malignant transformation
  • Most of the reported cases of malignant transformation have occurred in adults, typically in their 40s or 50s
    • However, there have been rare cases reported in children as well
  • Risk of malignant transformation appears to be higher in large or longstanding pilomatricoma and in those that have been previously treated with radiation therapy (J Dermatol 2010;37:735)
Case reports
Treatment
  • Surgical excision is curative
Clinical images

Contributed by Mark R. Wick, M.D.

Breast skin



Images hosted on other servers:

Bullous pilomatricoma

Giant pilomatricoma

Preoperative appearance of face pilomatricoma

Calcified pilomatricoma

Gross description
  • Hard calcified, may be nodulocystic
  • Circumscribed edges, lobulated
  • Keratinous material appears cheesy while calcified areas are gritty
  • Rare ossified area will be difficult to cut
Gross images

Contributed by Mark R. Wick, M.D.

Breast skin



Images hosted on other servers:

Well circumscribed,
lobulated mass
with cheesy material

Microscopic (histologic) description
  • Lobulated and circumscribed dermal based neoplasm
  • Islands of basaloid cells exhibiting abrupt keratinization, without intervening granular layer (trichilemmal keratinization)
  • Ghost / shadow cells, may predominate
  • Basaloid cells show mitotic activity; however, abnormal mitoses are absent
  • Intermediary cells progressively more eosinophilic cytoplasm, with pyknotic nucleus
  • Tumor keratin may elicit inflammatory response with foreign body giant cells, granuloma, cholesterol clefts and calcification
  • Uncommonly metaplastic bone formation is present along with extramedullary hematopoiesis (Cureus 2022;14:e30661)
  • Hemosiderin, melanin and amyloid deposition has been reported
  • Occasionally tumors exhibit some atypical features that suggest a potential for malignancy but does not fully meet the criteria for pilomatrix carcinoma; these features include a focal infiltrative pattern at the periphery, variable cytological atypia, increased mitotic rate (up to 5/high power field) and irregular foci of central necrosis (Case Rep Dermatol 2021;13:98)
  • Histological patterns: pigmented, proliferating, nodulocystic, perforating, bullous, pilomatricoma with osseous metaplasia, melanocytic matricoma (JAAD Case Rep 2023;33:51)
Microscopic (histologic) images

Contributed by Nasir Ud Din, M.B.B.S. and Andrey Bychkov, M.D., Ph.D.

Dermal based pilomatricoma

Basaloid and shadow cells

Trichilemmal keratinization

Foreign body giant cell reaction

Shadow cells

Pilomatricoma with ossification


Ossified pilomatricoma

Dermal nodule

Basaloid nests

Ghost cells

Basal cells, giant cells and ghost cells

Virtual slides

Images hosted on other servers:
Circumscribed islands of ghost and basaloid cells

Circumscribed islands of ghost and basaloid cells

Cytology description
  • Clusters of basaloid cells with round to ovoid nuclei, nucleated squamous cells and anucleated squames as well as clusters of shadow cells are consistent with the characteristics typically seen in pilomatricoma (Diagn Pathol 2018;13:65)
  • Foreign body giant cells may be present; markedly calcified lesion may yield scanty material (Int J Trichology 2012;4:280)
  • Shadow cells are specialized cells, the result of a process called shadow cell degeneration, in which the cells lose their nuclei and undergo a series of structural changes
  • Cytoplasm of shadow cells often contains basophilic (blue) granules, which are thought to represent remnants of the cellular organelles
Cytology images

Contributed by R. F. Chinoy, M.D. (Case #424)
Basaloid cell smear

Basaloid cell smear

Giant cell in smear

Giant cell in smear



Images hosted on other servers:

Basaloid epithelial cells with scant cytoplasm

Shadow cell with keratinous cytoplasm

Cluster of shadow cells

MGG, cluster of basaloid cells

MGG, abundant cytoplasm

Positive stains
Negative stains
Electron microscopy description
  • Cell differentiation and keratinization in shadow cells is similar to the process in the cortex of hair
  • Shadow cells are fully keratinized that form a mantle around nuclear remnants
Videos

Diagnostic pearls of pilomatricoma in superficial biopsy

Pilomatricoma: mini tutorial

Sample pathology report
  • Left cheek, excision biopsy:
    • Pilomatricoma (see comment)
    • Comment: There is a well circumscribed, lobulated, dermal based adnexal neoplasm comprising of many islands of basaloid cells with scattered mitoses. These cells exhibit abrupt keratinization transforming to shadow cells. Surrounding stroma has histiocytes, foreign body giant cell reaction and foci of calcification. There is no evidence of atypical mitoses, nuclear anaplasia or destructive infiltration into the surrounding tissue. Immunohistochemical stain beta catenin shows nuclear positivity in basaloid cells. Features are consistent with pilomatricoma, a benign adnexal tumor with hair matrix differentiation. The tumor is completely excised. Local recurrence is uncommon following complete excision.
Differential diagnosis
  • Basal cell carcinoma:
    • Sun exposed areas of skin
    • Most often seen in older individuals
    • May occur in children in association with basal cell nevus syndrome, xeroderma pigmentosum and organoid nevus
    • Nests of basaloid cells palisade at periphery
    • Stromal retraction and mucin
    • High mitotic activity
    • Most show connection to epidermis
    • Lacks anucleate shadow cells
    • BerEP4 positive
  • Basal cell carcinoma with matrical differentiation:
    • Typical features of basaloid cells predominate with foci of matrical differentiation
  • Pilomatrix carcinoma:
    • Older adults; case reports document occurrence in children and young adults, mostly de novo
    • Rare cases arise from existing pilomatricoma
    • Pleomorphic basaloid cells
    • Prominent nucleoli
    • High mitotic activity, necrosis, lymphovascular invasion (rare)
    • Locally aggressive behavior, infiltrating borders
    • Local recurrence common
    • Metastases may rarely occur, involving regional lymph nodes and lungs
  • Squamous cell carcinoma:
    • Intercellular bridges
    • Epidermoid keratinization
    • Keratin pearl formation
    • Lacks shadow cells
  • Merkel cell carcinoma:
    • Small cells hyper chromatic nuclei; stippled (salt and pepper) nuclear chromatin pattern
    • Lacks anucleate shadow cells
    • Immunoreactive for neuroendocrine markers, such as chromogranin, synaptophysin, CD56 and CD57
    • Dot positivity for CK20
  • Epidermal cysts of Gardner syndrome:
    • Changes of pilomatricoma can occur
  • Other follicular tumors showing matrical differentiation, including
    • Trichoblastoma:
      • Circumscribed, dermal basaloid tumor without surface connection
      • Stromal induction and formation of primitive hair bulbs
    • Trichoepithelioma:
      • Dermal based, focal epidermal connection, branching nests of basaloid cells
      • Aggregations of fibroblasts as abortive attempts to form papillary mesenchyme (papillary mesenchymal bodies)
      • Small keratinous cysts
    • Panfolliculoma:
      • Rare, advanced follicular differentiation
      • Overlap features between trichoblastoma and matricoma
      • All elements of follicular differentiation
  • Ghost cells may also be found in noncutaneous tumors, such as calcifying cystic odontogenic tumor (CCOT), dentinogenic ghost cell tumor, odontogenic ghost cell carcinoma and craniopharyngioma; occasionally, these cells are also seen in odontomas, ameloblastoma, adenomatoid odontogenic tumor and ameloblastic fibroma (J Pharm Bioallied Sci 2015;7:S142)
Board review style question #1
A 5 year old girl presented with a hard nodule on her arm. Fine needle aspiration yielded clusters of epithelial cells with scant cytoplasm, some anucleated and nucleated squamous cells. Focal calcified debris was noticed. Which immunohistochemical stain is helpful in diagnosis?

  1. Beta catenin
  2. CK5/6
  3. Ki67
  4. p40
  5. p53
Board review style answer #1
A. Beta catenin. Beta catenin is the defining molecular alteration in pilomatricoma and nuclear staining of basaloid cells is diagnostic. Answers B and D are incorrect because CK5/6 and p40 expression is also seen in squamous cell carcinoma and some basal cell carcinoma. Answer E is incorrect because p53 aberrant expression is significant for malignant behavior and does not help in the identification of pilomatixoma. Answer C is incorrect because Ki67 will show increased staining in basaloid cells and other inflammatory cells (if present in the adjacent stroma). Identification of abrupt keratinization and ghost cells is based on H&E section.

Comment Here

Reference: Pilomatrixoma
Board review style question #2

A 20 year old man had multiple gradually enlarging, itchy scalp tumors for the past 2 years. On examination it was hard nodule and overlying skin was stretched, revealing multiple facets and angles resembling a tent. The excision specimen revealed features as shown in the photomicrograph above. The patient's history is significant for total colectomy a year ago with more than 100 polyps. What is the patient suffering from?

  1. Gardner syndrome
  2. Gliomatosis cerebri
  3. Myotonic dystrophy
  4. Rubinstein-Taybi syndrome
  5. Turner syndrome
Board review style answer #2
A. Gardner syndrome. The photomicrograph shows a classic picture of basaloid cells and shadow cells, consistent with pilomatricoma. Multiple lesions and past history of total colectomy with more than 100 polyps favors familial adenomatous polyposis related syndrome, specifically Gardener syndrome. Multiple pilomatricoma (6 or more) are significant for association with various syndromes, most involving myotonic dystrophy and familial adenomatous polyposis related syndromes (including Gardner syndrome). Other syndromes include Turner syndrome, Rubinstein-Taybi syndrome, Sotos syndrome (relatively scant) and Kabuki syndrome.

Answer C is incorrect because myotonic dystrophy is part of a group of autosomal dominant disorders with high penetrance that show skeletal and respiratory muscle weakness and myotonia. Other findings may be cardiac muscle abnormalities and conduction defects, cataracts, daytime somnolence and endocrine disorders.

Answer E is incorrect because in Turner syndrome, patients are female and lack part or all of the second sex chromosome. Characteristic features include short stature, premature ovarian failure and high risk for various phenotypic abnormalities (e.g., high palate, nail dysplasia, low posterior hairline, widely spaced nipples, webbed neck and low set or malrotated ears), as well as other medical problems such as cardiovascular defects, skeletal anomalies and autoimmune disorders. It is suggested that growth hormone (or its effector molecule insulin-like growth factor 1) and estrogen may promote hair follicle growth and inhibit apoptosis, as well as produce pilomatricomas.

Answer D is incorrect because Rubinstein-Taybi syndrome is a rare genetic disorder characterized by mental retardation and multiple congenital anomalies. It involves broad thumbs and toes, short stature, distinctive facial features and varying degrees of intellectual disability. Low set ears, cataracts, microcephaly and cryptorchidism may also be seen.

Answer B is incorrect because gliomatosis cerebri is a diffuse growth pattern of glioma that invade multiple lobes of the brain. By definition, more than 2 brain regions have to be affected and symptoms include epilepsy, headaches and behavioral changes.

Comment Here

Reference: Pilomatrixoma

Pleomorphic dermal sarcoma
Definition / general
  • An undifferentiated pleomorphic tumor with overlapping features of atypical fibroxanthoma but a higher rate of local recurrence and metastasis
Essential features
  • Undifferentiated pleomorphic tumor involving the dermis that histologically looks like an atypical fibroxanthoma and has any of the following:
    • Size > 2 cm
    • Shows extensive involvement of deeper tissue (subcutis, skeletal muscle, fascia)
    • Necrosis
    • Perineural
    • Lymphovascular invasion
Terminology
  • Pleomorphic dermal sarcoma (PDS)
  • Undifferentiated pleomorphic sarcoma of the skin
  • Superficial malignant fibrous histiocytoma (terminology no longer used)
ICD coding
  • ICD-10: C49.9 - malignant neoplasm of connective and soft tissue, unspecified
Epidemiology
Sites
  • Head and neck, predilection for scalp
Etiology
  • Ultraviolet radiation induced damage and immunosuppression
Clinical features
Diagnosis
  • Dependent on clinical and tissue pathologic correlation
Prognostic factors
Case reports
Treatment
Clinical images

Images hosted on other servers:

Rapidly growing scalp mass

Well circumscribed, erythematous papule

Solitary protuberant mass on upper eyelid

Microscopic (histologic) description
  • Dermal based lesion composed of pleomorphic cells with vesicular nuclei and prominent nucleoli (Cancer 1973;31:1541)
    • Cells can be spindled or epithelioid, often with admixed multinucleated giant cells
    • Cells can be arranged in sheets and fascicles
  • Necrosis often present
  • Perineural and lymphovascular invasion can be seen
  • Infiltration into subcutis, skeletal muscle or fascia
  • Additional findings include myxoid change, pseudoangiomatous growth and storiform growth (Am J Surg Pathol 2012;36:1317)
Microscopic (histologic) images

Contributed by Anthony Martinez, M.D.

Dermal based neoplasm

Cytologic atypia

Necrosis

Extensive subcutis involvement

Sheet-like growth

HMWK

Positive stains
Negative stains
Molecular / cytogenetics description
Sample pathology report
  • Skin, scalp, excision:
    • Pleomorphic dermal sarcoma
    • Tumor measures 3.5 cm in greatest dimension and extensively involves the subcutaneous tissue
    • Necrosis is present: 30%
    • Lymphovascular invasion is not identified
    • Perineural invasion is not identified
    • Margins are negative
Differential diagnosis
Board review style question #1
The following image is from a scalp lesion in an 85 year old man. Immunostains for AE1 / AE3, 34betaE12 / HMWCK / high molecular weight, S100, actin - alpha smooth muscle, desmin and ERG are negative. Which is the best diagnosis?



  1. Atypical fibroxanthoma
  2. Leiomyosarcoma
  3. Pleomorphic dermal sarcoma
  4. Sarcomatoid squamous cell carcinoma
Board review style answer #1
C. Pleomorphic dermal sarcoma

Comment Here

Reference: Pleomorphic dermal sarcoma
Board review style question #2
An 80 year old man has a dermal based scalp lesion characterized by pleomorphic cells growing in fascicles. The lesion is < 2 cm, well circumscribed and completely confined to the dermis. Immunostains are negative for high and low molecular weight keratins, S100, actin - alpha smooth muscle and desmin. What is the best diagnosis?

  1. Atypical fibroxanthoma
  2. Leiomyosarcoma
  3. Pleomorphic dermal sarcoma
  4. Sarcomatoid squamous cell carcinoma
Board review style answer #2
A. Atypical fibroxanthoma

Comment Here

Reference: Pleomorphic dermal sarcoma

Pleomorphic fibroma
Definition / general
  • Polypoid or dome-shaped cutaneous nodule with sparse cellularity and cytologic atypia of fibroblasts
  • Not a WHO diagnosis
  • First described in 1989 (Am J Surg Pathol 1989;13:107)
  • Usually trunk, extremity or head (Clin Exp Dermatol 1998;23:22)
  • Related entities: may be a variant of skin tag (acrochordon) with cytologic atypia
Clinical features
  • Flesh-colored polyp resembling skin tag (acrochordon)
Case reports
Microscopic (histologic) description
Microscopic (histologic) images

AFIP images
Large pleomorphic cells

Large pleomorphic cells

Atypical cells

Atypical cells



Images hosted on other servers:
Anal skin

Anal skin

Stromal cells have atypical nuclei

Stromal cells have atypical nuclei

Cytology description
  • Single lying and few clusters of pleomorphic cells with some monster cells and scant cytoplasm (J Cytol 2013;30:71)
Cytology images

Images hosted on other servers:
Pleomorphic cells with very large nuclei

Pleomorphic cells with very large nuclei

Pleomorphic cells scattered between collagen bundles

Pleomorphic cells
scattered between
collagen bundles

Vimentin+

Vimentin+

Positive stains
Differential diagnosis

Pleomorphic rhabdomyosarcoma

Polymorphous sweat gland carcinoma
Definition / general
Essential features
  • Histologically, the lesions are characterized by a cellular proliferation showing a combination of growth patterns, including trabecular, solid, tubular, cribriform or adenoid cystic and pseudopapillary (Am J Dermatopathol 2018;40:580)
  • Marked predilection for the distal extremities
Terminology
  • Polymorphous sweat gland carcinoma
  • Polymorphous low grade sweat gland carcinoma
  • PSGC
ICD coding
  • ICD-O-3 topography code: C44 - Skin
Epidemiology
Sites
Clinical features
Case reports
Treatment
  • Complete excision with conservative margins has been recommended as the preferred modality of treatment
Microscopic (histologic) description
  • Characterized by an admixture of multiple growth patterns within the same lesion, including trabecular, solid, tubular, cylindromatous and pseudopapillary, as well as prominent stromal changes, including hyalinization, hemorrhage, calcification and cystic changes
  • Tumors grow within the dermis in a generally well demarcated, pushing border but rarely can show infiltration at the edges
  • Parts of the tumor also show glandular differentiation and adnexal differentiation, such as abortive hair follicle formation and numerous squamous eddies
  • Tumors are predominantly composed of large epithelioid cells with atypical nuclei and abundant eosinophilic cytoplasm
    • Smaller cells with darker, angulated nuclei are scattered throughout the tumor
  • Spindling of tumor cells can be seen focally
  • Mitoses are present and can vary from case to case
Microscopic (histologic) images

Contributed by Shira Ronen, M.D.
Abortive hair follicle formation

Abortive hair follicle formation

Combination of growth patterns

Combination of growth patterns

Combination of growth patterns

Combination of growth patterns

Cribriform growth pattern

Cribriform growth pattern

Glandular differentiation

Glandular differentiation


Infiltration of surrounding fibroadipose tissue

Infiltration of surrounding fibroadipose tissue

Numerous squamous eddies

Numerous squamous eddies

Spindle cell appearance

Spindle cell appearance

Trabecular growth pattern Trabecular growth pattern

Trabecular growth pattern


Androgen receptor

Androgen receptor (AR)

CD56

CD56

Chromogranin

Chromogranin

CK5/6

CK5/6

p16

p16

p63

p63

Positive stains
Molecular / cytogenetics description
  • Negative for MYB-NFIB fusion
Sample pathology report
  • Neck mass, excision:
    • Polymorphous sweat gland carcinoma (see comment)
    • Comment: Sections show a relatively well demarcated tumor involving the dermis without a connection to the epidermis. The tumor shows multiple growth patterns including solid, trabecular, tubular and focal cribriform. Prominent hemorrhage, cystic changes and calcifications are seen. The cells are mostly epithelioid with prominent nucleoli and eosinophilic cytoplasm. Scattered mitotic figures are also observed. There is no evidence of significant cytologic atypia or necrosis. Overall, in the absence of a known or occult primary malignancy, the tumor is most consistent with a polymorphous sweat gland carcinoma. A clinical workup to exclude the possibility of metastasis, as clinically indicated, is recommended.
Differential diagnosis
  • Adenoid cystic carcinoma (ACC) (Am J Dermatopathol 1986;8:2):
    • ACC is characterized by monomorphic basaloid cells, which are arranged in tubules, elongated nests and cords
    • There are typically numerous cystic and ductular spaces with a prominent cribriform pattern
    • In comparison, polymorphous sweat gland carcinoma is characterized by striking variegation of histologic growth patterns, including glandular, trabecular, solid, cylindromatous and cribriform areas within the same tumor mass
    • MYB overexpression
    • Consistently expresses EMA
    • Positive for CD117, CEA, S100 and vimentin
  • Basal cell carcinoma (Am J Dermatopathol 1986;8:2):
    • Retains striking peripheral nuclear palisading and stromal retraction
  • Metastases from internal malignancies:
    • Can look histologically identical to polymorphous sweat gland carcinoma; therefore, clinical history and correlation with imaging are necessary
    • Negativity for p63 and CK5/6 favors a diagnosis of cutaneous metastasis of adenocarcinoma from internal organs over a primary cutaneous adnexal carcinoma
  • Benign and malignant adnexal neoplasm, such as ductal eccrine carcinoma, microcystic carcinoma and papillary digital eccrine carcinoma:
    • Key difference is the striking variegation of histologic growth patterns seen in polymorphous sweat gland carcinoma that would not be seen in other adnexal neoplasms
Board review style question #1
Which of the following statements is true about polymorphic sweat gland adenocarcinoma?

  1. It is characterized by an admixture of multiple growth patterns within the same lesion
  2. The most common location is the abdomen
  3. The tumor is common
  4. This tumor shows MYB-NFIB fusion
  5. It is a fast growing tumor
Board review style answer #1
A. It is characterized by an admixture of multiple growth patterns within the same lesion. The other answer choices are incorrect, since the tumor is shown to favor the extremities (choice B), the tumor is uncommon and only a few case reports and case series have been described (choice C), polymorphic sweat gland adenocarcinoma is negative for the MYB-NFIB fusion (choice D) and it is a slow growing, skin colored, firm dermal nodule (choice E).

Comment Here

Reference: Polymorphous sweat gland carcinoma
Board review style question #2
Which of the following immunohistochemical stains will show strong diffuse positivity in polymorphic sweat gland adenocarcinoma?

  1. CD117 and CEA
  2. p63 and p16
  3. Vimentin and cytokeratin AE1 / AE3
  4. Chromogranin and S100
  5. DOG1 and p40
Board review style answer #2
B. p63 and p16. The other answer choices are incorrect, since CD117 and CEA are negative (choice A), vimentin is negative within this entity, while cytokeratin AE1 / AE3 shows strong diffuse positivity (choice C), chromogranin stain shows focal expression, while S100 is negative within the tumor (choice D) and DOG1 is negative within this entity, while p40 shows strong diffuse positivity (choice E).

Comment Here

Reference: Polymorphous sweat gland carcinoma

Porokeratosis
Definition / general
  • Skin disorder of abnormal clonal keratinization with various clinical expressions and subtypes
  • Characterized by 1 or more variably sized lesions on the skin that are rimmed by a hyperkeratotic border
Essential features
  • Keratinization disorder resulting from abnormal clonal expansion of keratinocytes
  • Histologic hallmark is cornoid lamella, a column of parakeratosis with underlying dyskeratosis with reduction of keratohyalin granules (Indian J Dermatol Venereol Leprol 2022;88:291)
  • Some subtypes premalignant
  • Subtypes include
    • Porokeratosis of Mibelli
    • Disseminated actinic porokeratosis (DSAP)
    • Disseminated superficial porokeratosis (DSP)
    • Linear porokeratosis
    • Eruptive disseminated porokeratosis (EDP)
    • Porokeratosis palmaris et plantaris disseminate
    • Punctate porokeratosis
    • Porokeratosis ptychotropica
    • Penoscrotal porokeratosis
    • Follicular porokeratosis
Terminology
  • Disseminated superficial actinic porokeratosis
  • Disseminated superficial porokeratosis
  • Porokeratosis of Mibelli
  • Linear porokeratosis
  • Eruptive disseminated porokeratosis
  • Porokeratosis palmaris et plantaris disseminata
  • Punctate porokeratosis
  • Porokeratosis ptychotropica
  • Penoscrotal porokeratosis
  • Follicular porokeratosis
  • Porokeratoma
ICD coding
  • ICD-10
    • L56.5 - disseminated superficial actinic porokeratosis (DSAP)
    • Q82.8 - other specified congenital malformations of skin
    • L98.8 - other specified disorders of the skin and subcutaneous tissue
  • ICD-11
    • ED52 - porokeratoses
    • EC20.32 - papular palmoplantar keratodermas
Epidemiology
Sites
Pathophysiology
Etiology
Clinical features
Diagnosis
Prognostic factors
  • Prognosis varies depending on subtype and severity
  • Often a chronic condition refractory to treatment
  • Rarely, lesions may spontaneously resolve
  • Malignant transformation is rare
Case reports
Treatment
  • No standard treatment
    • Long term follow up
    • Sun protection recommended for all patients
    • Reported treatment options include excision, cryosurgery, electrocautery, carbon dioxide laser, topical 5-fluorouracil, keratolytics, imiquimod, vitamin D analogues and topical retinoids (Patterson: Weedon's Skin Pathology, 5th Edition, 2020)
  • Topical cholesterol / lovastatin has been shown to be effective, particularly in DSAP (J Am Acad Dermatol 2020;82:123)
  • Screen for hepatobiliary or hematologic malignancy in EDP
Clinical images

Images hosted on other servers:
Disseminated superficial porokeratosis

Disseminated superficial porokeratosis

Porokeratosis of Mibelli

Porokeratosis of Mibelli

Porokeratosis ptychotropica

Porokeratosis ptychotropica

Linear porokeratosis following Blaschko lines

Linear porokeratosis following Blaschko lines


Punctate porokeratosis of palms and soles

Punctate porokeratosis of palms and soles

Penoscrotal porokeratosis

Penoscrotal porokeratosis

Eruptive disseminated porokeratosis on the thigh

Eruptive disseminated porokeratosis on the thigh

Follicular porokeratosis of the scalp

Follicular porokeratosis of the scalp

Microscopic (histologic) description
Microscopic (histologic) images

Contributed by Wei-Shen Chen, M.D., Ph.D.
Atrophic center

Atrophic center

Slanted columns of parakeratosis (cornoid lamellae) Slanted columns of parakeratosis (cornoid lamellae)

Slanted columns of parakeratosis (cornoid lamellae)

Central acanthosis

Central acanthosis


Cornoid lamellae arising from follicular unit Cornoid lamellae arising from follicular unit

Cornoid lamellae arising from follicular unit

Cornoid lamellae distributed throughout

Virtual slides

Images hosted on other servers:
Disseminated superficial actinic porokeratosis

Disseminated superficial actinic porokeratosis

Porokeratosis of Mibelli

Porokeratosis of Mibelli

Punctate porokeratosis

Punctate porokeratosis

Linear porokeratosis

Linear porokeratosis

Positive stains
  • PAS special stain reportedly highlights granules within the corneocytes of the cornoid lamella, rarely required
Videos

Short video discussing porokeratosis (pathology pearls)

Sample pathology report
  • Skin, right upper arm, skin shave:
    • Porokeratosis (see comment)
    • Comment: Sections show tiers of parakeratosis overlying dyskeratotic keratinocytes. If multiple lesions are present clinically, a diagnosis of DSAP should be considered.

  • Skin, right scrotum, skin shave:
    • Porokeratosis (see comment)
    • Comment: Sections show acanthosis with multifocal columns of parakeratosis overlying dyskeratotic keratinocytes. At this anatomic site, the findings are consistent with a diagnosis of penoscrotal porokeratosis.
Differential diagnosis
  • Porokeratoma:
  • Verrucae:
    • Epidermal acanthosis, papillomatosis with tiers of parakeratosis and koilocytosis
  • Actinic keratosis:
    • Gritty papules in sun exposed sites
    • Histology with lower keratinocyte atypia and disorder
    • Cornoid lamella uncommonly seen
  • Psoriasis:
    • Regular acanthosis with parakeratosis and intracorneal neutrophils (Munro microabscesses)
  • Porokeratotic eccrine ostial and dermal duct nervus:
    • An eccrine hamartoma with punctate hyperkeratotic papules affecting the palms and soles
    • Histologically and clinically could appear like punctate porokeratosis but cornoid lamellae are limited to eccrine ostia
  • Tinea corporis (Am J Clin Dermatol 2022;23:37):
    • Foci of parakeratosis with intracorneal hyphae
Board review style question #1


A 40 year old woman presented with an erythematous, annular macule with a thread-like, raised border on her arm. She had several similar lesions on her extensor arms. What is the most likely diagnosis?

  1. Disseminated superficial actinic porokeratosis
  2. Guttate psoriasis
  3. Macular seborrheic keratoses
  4. Nummular eczema
  5. Tinea corporis
Board review style answer #1
A. Disseminated superficial actinic porokeratosis. This patient has disseminated superficial actinic porokeratosis, which classically presents as small, annular macules with thread-like raised borders. It usually presents more diffusely than other porokeratosis subtypes and frequently on sun exposed areas (posterior neck, extensor surfaces of arms, legs). The histological finding of cornoid lamella is characteristic of porokeratosis.

Answer B is incorrect because guttate psoriasis is a variant of psoriasis that presents in children and adolescents following streptococcal infection (StatPearls: Guttate Psoriasis [Accessed 7 August 2023]). Although guttate psoriasis may also present as lesions over the extensor surfaces, biopsy would reveal a psoriasiform reaction pattern. There is less acanthosis than seen in psoriasis vulgaris. Other features of psoriasis include dilated superficial blood vessels, parakeratosis and neutrophils in the stratum corneum (microabscesses of Munro) (StatPearls: Guttate Psoriasis [Accessed 7 August 2023]).

Answer C is incorrect because seborrheic keratoses are benign lesions that classically present as waxy papules or plaques with a stuck on appearance. Histologic features include hyperkeratosis, acanthosis and papillomatosis (Dermatol Online J 2019;25:13030).

Answer D is incorrect because nummular eczema is a chronic skin condition characterized by oval or coin shaped, erythematous, eczematous plaques. These lesions are usually symmetrically distributed with a predilection for the lower and upper extremities (Recent Pat Inflamm Allergy Drug Discov 2020;14:146). Predominant features of eczema are spongiosis, acanthosis and exocytosis of inflammatory cells.

Answer E is incorrect because tinea corporis is a superficial dermatophyte infection that may clinically resemble porokeratosis due to its annular appearance. Histopathology shows hyphal forms in the stratum corneum.

Comment Here

Reference: Porokeratosis
Board review style question #2
A 28 year old man presented with a 7 month history of a persistent rash over the scrotum and shaft of the penis associated with erythema, burning and pruritis. Clinical examination revealed a well circumscribed, erythematous plaque with a raised border and atrophic center. Poorly defined patches were also seen on the penile shaft. A punch biopsy revealed multiple cornoid lamellae. What is the most likely diagnosis?

  1. Bowen disease
  2. Condyloma acuminatum
  3. Penoscrotal porokeratosis
  4. Verruciform xanthoma
  5. Zoon balanitis
Board review style answer #2
C. Penoscrotal porokeratosis. This patient's clinical pathologic presentation is consistent with a diagnosis of penoscrotal porokeratosis, a distinct variant of porokeratosis that is often self resolving. This variant is seen in young males typically during the third decade of life (Indian Dermatol Online J 2015;6:339). Histology demonstrates multiple cornoid lamellae involving a mildly hyperplastic epidermis.

Answer A is incorrect because Bowen disease is a form of in situ squamous cell carcinoma. Although Bowen disease may present similarly to porokeratosis, it is rare in patients younger than 30 years of age (Indian Dermatol Online J 2022;13:177). Histopathology would demonstrate full thickness keratinocyte atypia (Indian Dermatol Online J 2022;13:177).

Answer B is incorrect because condyloma acuminata are anogenital warts caused by the human papillomavirus (HPV), most commonly HPV strains 6 and 11 (StatPearls: Condyloma Acuminata [Accessed 19 January 2024]). Similar to porokeratosis, condyloma acuminata display hyperkeratosis on histopathological evaluation. However, distinctive koilocytes are also expected. Additionally, condyloma acuminata lesions are usually found in groups that may coalesce into larger lesions and most often present as fleshy papules that range in size.

Answer D is incorrect because verruciform xanthoma is a rare mucocutaneous verrucopapillary lesion that is not associated with dyslipidemia. Histopathology demonstrates epithelial hyperplasia with prominent parakeratinization. The histological hallmark is the presence of foam cells or xanthoma cells confined to the connective tissue papillae (J Oral Maxillofac Pathol 2019;23:43).

Answer E is incorrect because Zoon balanitis is a nonvenereal condition that most commonly affects middle aged to older uncircumcised men. It most commonly presents as a single shiny erythematous plaque on the glans penis. Early cases will reveal thickening of the epidermis, parakeratosis and a patchy lichenoid infiltrate of lymphocytes and numerous plasma cells. Atrophy of the epidermis, superficial erosions, scattered neutrophils in the upper epidermis, scant spongiosis and a denser plasmacytic infiltrate may be seen in later stages (Indian J Sex Transm Dis AIDS 2016;37:129).

Comment Here

Reference: Porokeratosis

Poroma
Definition / general
  • Benign glandular adnexal tumor that usually originates from cells of the outer layer of the acrosyringium and terminal eccrine duct
  • Has both eccrine and apocrine origin (AMA Arch Derm 1956;74:511)
  • Malignant counterpart is referred to as porocarcinoma
Essential features
  • Single, slow growing, asymptomatic, well circumscribed, smooth, skin colored to red, slightly scaly papule or nodule
  • Most commonly on palms and sole or sides of the foot
  • Well circumscribed broad anastomosing bands of poroma cells with sharp demarcation from adjacent keratinocytes
  • Excellent prognosis with simple excision
Terminology
  • Eccrine poroma / hidroacanthoma simplex / dermal duct tumor, apocrine poroma
ICD coding
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
Epidemiology
Sites
Pathophysiology
  • Falls under the broad category of poroid neoplasms or acrospiromas
  • Poroid neoplasms include the eccrine poroma, apocrine poroma, hidroacanthoma simplex and dermal duct tumor (Clin Exp Dermatol 2014;39:119)
    • Eccrine poroma: derived from cells of the outer layer of the acrosyringium and the upper dermal eccrine duct, both epidermal and dermal (AMA Arch Derm 1956;74:511)
    • Apocrine poroma: reflects the common embryological ancestry of the 3 units (the folliculosebaceous apocrine unit) (Pathologe 2014;35:456)
Etiology
Clinical features
Diagnosis
  • Clinical appearance confirmed by characteristic histologic findings
Prognostic factors
Case reports
Treatment
Clinical images

Contributed by Aayushma Regmi, M.B.B.S. and Jodi Speiser, M.D.

Pink, scaly nodule

Erythematous nodule

Pedunculated and pigmented nodule

Gross description
Microscopic (histologic) description
  • Eccrine poroma:
    • Well circumscribed
    • Replaces the epidermis and extends into the dermis in broad anastomosing bands
    • Poroma cells are monomorphic, small, cuboidal with basophilic round nuclei, inconspicuous nucleoli and compact eosinophilic cytoplasm
    • Sharp demarcation present between the normal keratinocytes and poroma cells
    • Devoid of peripheral palisading
    • Ductal lumina with single row of luminal cells covered by eosinophilic lining or cystic spaces devoid of any formal lining
    • Cells are united by conspicuous intercellular bridges and supported by a delicate fibrovascular stroma (J Dermatol 1980;7:263)
    • Poroma cells usually contain glycogen (Int J Dermatol 2014;53:1053)
    • Occasionally, pigmented variants with associated dendritic melanocytes and tumor cell melanin deposition (J Dermatol 2010;37:542, J Eur Acad Dermatol Venereol 2008;22:303)
    • Dystrophic calcification and transepidermal elimination of tumor nests are exceptional findings (J Dermatol Case Rep 2009;3:38, J Dermatol 1997;24:539)
  • Apocrine poroma:
    • Shows sebaceous differentiation with the occasional presence of follicular differentiation and foci of apocrine-like features (J Cutan Pathol 2001;28:101)
    • Anastomosing trabeculae, displaying multiple points of origin from the epidermis and located largely in the papillary and upper reticular dermis
    • Cells are small and uniform with scanty cytoplasm and round to oval nuclei united by inconspicuous intercellular bridges
    • Foci of ductal differentiation with a well developed eosinophilic cuticle
    • Follicular differentiation in the form of epithelial lobules (Am J Dermatopathol 1999;21:31)
    • Sebaceous cells, singly and in clusters with bubbly cytoplasm and crenated nuclei is an infrequent feature (Am J Dermatopathol 1996;18:1)
  • Eccrine porocarcinoma:
    • May remain completely intraepidermal (in situ porocarcinoma) but is more often associated with an invasive dermal component
    • Poroma cells, with typical ductal lumina, associated with cytological features of malignancy, including nuclear and cytoplasmic pleomorphism, nuclear hyperchromatism and mitotic activity (Am J Surg Pathol 2001;25:710)
    • Prone to have local recurrence (17%) and is occasionally associated with nodal metastases (19%); however, systemic spread is rare (11%) (Am J Surg Pathol 2001;25:710)
Microscopic (histologic) images

Contributed by Aayushma Regmi, M.B.B.S. and Jodi Speiser, M.D.

Tumor with epidermal connection

Tumor with epidermal connection

Pedunculated nodule


Pigmented poroma

Poroma cells and adjacent keratinocytes

Poroma cells with duct

Hyalinized stroma and blood vessels


Poroma cells

EMA

CK7

CEA

BerEp4

Virtual slides

Images hosted on other servers:

Poroma cells with epidermal connection

Positive stains
Negative stains
Electron microscopy description
  • Cells have numerous connecting desmosomes, cytoplasmic tonofilaments, glycogen granules and intracytoplasmic lumina (J Dermatol 1980;7:263)
Molecular / cytogenetics description
Sample pathology report
  • Skin, left palm, excision:
    • Eccrine poroma, extending to the deep margin
Differential diagnosis
  • Hidroacanthoma simplex:
    • Entirely intraepidermal
    • Discrete circumscribed populations of poroma cells within an irregularly acanthotic epidermis
  • Dermal duct tumor:
    • Entirely intradermal, the epidermis is unaffected
    • Large lobules of uniform poroma cells in the mid and lower dermis
  • Basal cell carcinoma:
    • Nodules, nests or infiltrative cords with proliferation of small basaloid cells and peripheral palisading
    • Stromal retraction artifact between tumor cells and mucinous stroma
    • AE1 / AE3, BerEP4 positive
  • Squamous cell carcinoma (SCC):
    • Lacks evidence of ductal differentiation
    • Squamous differentiation abundant, eosinophilic cytoplasm with keratin pearls, intercellular bridges and keratinization
    • Greater cytologic atypia, dyskeratotic cells
  • Irritated / clonal seborrheic keratosis (SK):
    • Shows follicular differentiation with keratinizing pseudohorn cysts, no ductal differentiation
    • Cells are typically larger than in poroma
  • Hidradenoma:
    • Nests and nodules of epithelial cells lacking epidermal connection
    • Shows both solid and cystic components
    • More commonly shows clear cell features
  • Eccrine syringofibroadenoma:
    • Benign eccrine proliferation
    • Thin anastomosing reticulated cords and strands of basaloid monomorphous cuboidal cells extending from the basal layer of epidermis into dermis
    • Loose fibrovascular stroma
Board review style question #1
Which of the following vascular patterns is observed in the dermatoscopic evaluation of eccrine poroma?

  1. Glomerular
  2. Hairpin
  3. Mosaic
  4. Hairpin and glomerular
  5. Hairpin and mosaic
Board review style answer #1
D. Hairpin and glomerular. The vascular patterns commonly seen in eccrine poroma are the polymorphic, glomerular, linear irregular, leaf and flower-like and looped or hairpin variants. The leaf and flower-like pattern appears to be relatively unique to the poroma. Mosaic pattern is not observed in eccrine poroma (Clin Case Rep 2021;9:1601).

Comment Here

Reference: Poroma
Board review style question #2

Which of the following is true about eccrine poroma?

  1. Most commonly occurs on central part of the body: frontal scalp, anterior chest and around umbilicus
  2. No distinct demarcation between the poroma cells and adjacent keratinocytes
  3. Presence of monomorphic, basaloid cells with peripheral palisading
  4. Presence of sheets and trabeculae of monomorphic, round basophilic cells containing scattered duct-like structures
Board review style answer #2
D. Presence of sheets and trabeculae of monomorphic, round basophilic cells containing scattered duct-like structures

Comment Here

Reference: Poroma

Primary cutaneous CD4+ small / medium

Primary cutaneous CD8+ aggressive epidermotropic T cell lymphoma

Primary cutaneous PTCL, NOS (pending)

Primary cutaneous acral CD8+

Primary cutaneous anaplastic large cell lymphoma
Definition / general
Essential features
Terminology
  • Primary cutaneous anaplastic large cell lymphoma is included under the umbrella term CD30+ T cell lymphoproliferative disorders (Am J Dermatopathol 2017;39:877)
ICD coding
  • ICD-O: 9718/3 - primary cutaneous anaplastic large cell lymphoma
Epidemiology
Sites
Clinical features
Prognostic factors
Case reports
Treatment
Clinical images

Contributed by Roberto N. Miranda, M.D.

Clinical presentation of C-ALCL



Images hosted on other servers:

Annular, erythematous
to violaceous indurated
plaques with fixed
nodules over arm
Gross description
Microscopic (histologic) description
  • Diffuse dermal infiltrates with cohesive sheets of tumor cells with anaplastic morphology (round to irregularly shaped nuclei, prominent eosinophilic nucleoli and abundant cytoplasm); nonanaplastic appearance (pleomorphic or immunoblastic) is seen in 20% of cases (Pathology 2020;52:100, Am J Dermatopathol 2017;39:877)
  • Epidermotropism is marked in cases with DUSP22-IRF4 rearrangement (Histopathology 2015;66:846)
  • Cerebriform lymphocytes are not identified
  • Variable degrees of inflammatory infiltrate, reactive lymphocytes are common in the periphery of the lesions (Br J Dermatol 2003;148:580)
  • Ulcerated lesions may have lymphomatoid papulosis-like histology with abundant reactive T cells, histocytes, eosinophils, neutrophils and a low number of CD30+ cells (Am J Surg Pathol 2014;38:1203; J Cutan Pathol 2015;42:610)
Microscopic (histologic) images

Contributed by Roberto N. Miranda, M.D.

Primary C-ALCL

Cytological features

CD4 positivity

Strong CD30 positivity

ALK negativity


Systemic ALCL ALK- in skin

Cytologic features

Anaplastic morphology

Strong CD30+

Positive stains
Negative stains
Molecular / cytogenetics description
Sample pathology report
  • Skin, punch biopsy:
    • CD30 lymphoproliferative disorder, mostly consistent with cutaneous ALK- anaplastic large cell lymphoma, in the appropriate clinical context (see comment)
    • Comment: The section shows an extensive dermal infiltrate composed predominantly of medium to large sized lymphocytes and admixed neutrophils and histiocytes. Scattered mitotic figures are noted. There is epidermal ulceration and extensive neutrophilic serum crust that prevents the evaluation of epidermotropism. Immunohistochemical studies show that the large lymphoid cells are diffusely positive for CD30. CD3 and CD20 label a mixed population of T and B small lymphocytes in the background; the large cells are only focally positive for CD3 and are negative for CD20. The large cells are also partially positive for CD4, while CD8 highlights very rare background lymphocytes. TCR beta labels background lymphocytes while TCR gamma is essentially negative. CD56, EBER and ALK1 are negative.
Differential diagnosis
Board review style question #1

    Which marker is typically negative in primary cutaneous ALCL?

  1. ALK
  2. CD30
  3. CD4
  4. CD45
  5. Perforin
Board review style answer #1
Board review style question #2

    The diagnosis of primary cutaneous ALCL requires that at least what percent of lymphoma cells express CD30?

  1. 20%
  2. 30%
  3. 75%
  4. 90%
  5. 100%
Board review style answer #2

Primary cutaneous diffuse large B cell lymphoma, leg type
Definition / general
  • Primary cutaneous large B cell lymphoma (PCLBCL) composed exclusively of centroblasts and immunoblasts, typically arising in the leg
Essential features
  • Aggressive B cell neoplasm predominantly affecting older women
  • Histologic evaluation shows large neoplastic B cell lymphoma composed exclusively of centroblasts and immunoblasts
  • Neoplastic cells are positive for CD20, CD79a, strong staining for MUM1 and BCL2, while negative for CD10 and EBER
  • Treatment involves R-CHOP (rituximab, cyclophosphamide, hydroxydaunomycin, oncovin, prednisone) or local radiotherapy (solitary lesions)
ICD coding
  • ICD-10: C83.39 - Diffuse large B cell lymphoma, extranodal and solid organ sites
  • ICD-11: 2A81.A - Primary cutaneous diffuse large B cell lymphoma, leg type
  • ICD-0: 9680/3 - Malignant lymphoma, large B cell, diffuse, NOS
Epidemiology
Sites
Pathophysiology
  • Not clearly understood
  • B cells undergo aberrant somatic hypermutation; process of affinity maturation of immunoglobulins by somatic hypermutation in regions of the genome that do not encode immunoglobulin genes
    • May occur in oncogene containing gene loci
  • Loss of high fidelity DNA repair mechanisms
Etiology
  • Not clearly understood
  • Arises de novo
  • Predisposing factors:
    • Chronic antigenic stimulation by viral and bacterial infections
  • Possible association:
Clinical features
Diagnosis
  • Extensive clinical history (e.g. presence of B symptoms, fever, weight loss, etc.)
  • Thorough physical examination
  • Tissue biopsy of skin lesion with ancillary testing (flow cytometry, FISH, mutation next generation sequencing studies)
Laboratory
  • Complete blood count with differential (CBC w/ diff):
    • Lymphocytosis, atypical lymphocytes (if leukemic involvement)
  • Comprehensive blood chemistry (CMP)
    • Including liver and kidney analysis
  • Lactate dehydrogenase (LDH):
    • Elevated (indicator of tumor lysis)
  • Serum Beta2 microglobulin:
Radiology description
Prognostic factors
  • Unfavorable prognosis:
    • Multiple skin lesions at diagnosis
    • Loss of CDKN2A and CDKN2B gene loci on chromosome 9p21.3 due to promotor methylation or gene deletion (up to 67% of cases)
    • MYD88 L265P mutation (Am J Surg Pathol 2018;42:1488)
  • Favorable prognosis:
    • Solitary skin lesion at diagnosis
    • Addition of rituximab to CHOP regimen
Case reports
  • 41 year old woman with longstanding history of systemic lupus erythematosus presents with an enlarging, well demarcated 3 x 2 cm firm, multilobed, red violaceous, nontender nodule over the left shoulder (J Cutan Med Surg 2016;20:579)
  • 72 year old woman with a solitary, 3 cm upper arm nodule for 1 month duration (J Dermatol 2017;44:608)
  • 74 year old woman presenting with a painful, left lower leg lesion that was enlarging over 5 months (Cutis 2018;102:E31)
  • 82 year old woman with asymptomatic, growing lesions over the past year (Actas Dermosifiliogr 2014;105:78)
  • 83 year old man with history of pleomorphic sarcoma treated with radiotherapy, presents with a large, partially ulcerated cutaneous lesion in the region of prior radiation (J Gen Intern Med 2015;30:371)
  • 85 year old woman with disseminated solid nodularities (1 - 2 cm) on the face and upper extremities (Am J Dermatopathol 2017;39:e44)
Treatment
  • Local radiation or surgery for solitary lesions (Dermatol Clin 2015;33:835)
  • R-CHOP: rituximab, cyclophosphamide, hydroxydaunomycin, oncovin, prednisone
  • Bortezomib (NFkB inhibitor)
  • Ibrutinib (oral tyrosine kinase pathway inhibitor)
Clinical images

Contributed by Matko Kalac, M.D., Ph.D., Thomas Jandl, M.D. and Deena Altman, M.D.

Papulonodular lesion

Ulceration

Papulonodular lesions

Exophytic Lesions

Necrosis

Flat lesions, left shin

Microscopic (histologic) description
  • Superficial and deep dermal involvement by intermediate to large, monotonous, diffuse sheets of centroblasts (round, noncleaved nuclei with vesicular chromatin and one to multiple small nucleoli) and immunoblasts (large, oval nuclei with vesicular chromatin and one prominent nucleolus) (Semin Diagn Pathol 2017;34:85)
  • Epidermis spared (may be ulcerated)
  • Brisk mitotic activity frequently seen (Histopathology 2019;74:1067)
  • Apoptosis, karyorrhexis, cytoplasmic fragments (Am J Surg Pathol 2018;42:1488)
  • Small B cells and follicular dendritic cell meshworks (CD21+ / CD35+) are often absent
  • Rare reactive, perivascular T cells may be present
Microscopic (histologic) images

Contributed by Christian Salib, M.D. and Julie Teruya-Feldstein, M.D.

Lymphocytic Infiltrate

Centroblast and immunoblasts

Apoptosis

Immunoblasts

Immunoblasts with mitotic activity seen


CD79a

CD20

BCL6

MUM1

BCL2

Ki67 / MIB1

Cytology description
  • Neoplastic B cells resemble normal B cells that give rise to them
    • Centroblasts and immunoblasts cells
Positive stains
Negative stains
Molecular / cytogenetics description
  • Similar gene expression profile as DLBCL, activated B cell-like type (Blood 2005;105:3671)
  • Interface FISH: translocation of IGH genes with MYC or BCL6
  • DNA amplification of 18q21.31-21.33, including BCL2 and MALT1 genes, are identified in 67% of cases by array comparative genomic hybridization and FISH (J Clin Oncol 2006;24:296)
  • Loss of CDKN2A and CDKN2B gene loci on chromosome 9p21.3 due to promoter methylation or gene deletion (up to 67% of cases)
  • Other mutations: MYD88 L265P (60%), TNFAIP3 (40%), CD79B (20%), CARD11 (10%)
Molecular / cytogenetics images

Contributed by Vesna Najfeld, Ph.D.

FISH studies

Sample pathology report
  • Left thigh, biopsy:
    • Primary cutaneous diffuse large B cell lymphoma, leg type; nongerminal center, double expressing (BCL2+ MYC+) involving superficial and deep dermis
    • Microscopic description and additional studies:
      • H&E sections reveal diffuse sheets of an atypical lymphocyte population infiltrating the dermis while sparing the epidermis. The atypical cells are medium to large in size with moderate amount of cytoplasm, enlarged round nuclei with vesicular chromatin. Some cells show a single prominent nucleolus (immunoblasts-like) while others display multiple smaller nucleoli (centroblasts-like). Brisk mitotic activity is noted (up to 4/hpf). No areas of geographic necrosis is present. No granulomas seen on sections examined.
      • Immunochemistry show atypical large lymphoid cells are positive for CD45 (LCA), CD20, CD79a, MUM1, BCL6, BCL2 (> 95%), MYC (weak 40%), MIB1/Ki67 proliferative fraction of 80 - 90% while negative for CD3, CD10, P53 (< 3%), CD99, CD30 (< 1%), CHR, KER903, S100, TTF1, CK20, SYNAP.
      • In situ hybridization for EBV encoded viral RNA (EBER) is NEGATIVE.
      • PAS, GMS and AFB stains are negative for microorganisms.
    • Karyotype: nuc ish(BCL6x2),(D8Z2x2)(MYC,IGH)x3(MYC con IGHx2),(IGHx3,BCL2x2),(MYCx2)(5'MYC sep 3'MYCx1).
    • Report: Multiplex FISH x4 was performed on paraffin embedded tissue: nuc ish(BCL6x2) [191/200], (D8Z2x2)(MYC,IGH)x3(MYC con IGHx2) [130/200], (IGHx3,BCL2x2) [123/200], (MYCx2)(5'MYC sep 3'MYCx1) [124/200].
    • Interpretation: Interphase FISH results are consistent with a 65% of cells showing MYC-IGH [t(8;14)] fusion POSITIVE signal pattern. Cells were IGH-BCL2 fusion negative and BCL6 (3q27) gene rearrangement was NOT detected. Although the hallmark of Burkitt lymphoma is the presence of MYC-IGH rearrangement, approximately 10% of DLBCL cases will contain a MYC rearrangement and an aggressive clinical course (Arch Pathol Lab Med 2015;139:602).
    • Next generation mutation studies show:
      • Biomarker findings
      • Microsatellite status - MS-stable
      • Tumor mutational burden - TMB-intermediate (7 Muts/Mb)
      • CDKN2A/B loss
      • ETV6 rearrangement exon 4
      • MED12 R817H
      • MLL2 H313fs*20
      • MYD88 L265P
      • PRDM1 splice site 291+1G > T
Differential diagnosis
Additional references
Board review style question #1

    A 78 year old woman with no known clinical history presents with a solitary, violaceous, 4.3 cm papule on her left shin. Patient denies any history of recent travel, allergies or sexually transmitted infections. Biopsy reveals sheets of large round cells with vesicular chromatin and 1 to multiple prominent nucleoli infiltrating the dermis. The tumor cells are positive for CD20, MUM1 and BCL6, while negative for EBV, CD4, CD5, CD30 and ALK. What additional immunohistochemical stain will help differentiating this diagnosis from primary cutaneous follicle center lymphoma?

  1. BCL1 (cyclin D1)
  2. BCL2
  3. CD19
  4. CD23
  5. CD79a
Board review style answer #1
B. BCL2. The clinical, morphologic and immunohistochemical presentation are most suggestive of a large B-cell lymphoma. Differential considerations include de novo diffuse large B-cell lymphoma, leg type vs. secondary involvement of DLBCL from a different site (less likely, given no prior history of lymphoma) vs. primary cutaneous follicle center lymphoma (PCFCL). Primary cutaneous follicle center lymphoma are essentially follicular lymphomas arising from/associated with skin hair follicles, with neoplastic B-cell expressing germinal center markers (i.e. CD10, BCL6). Unlike follicular lymphomas however, the classic t(14;18)(BCL2-IGH) translocation is seen in only 20-30% of cases (Mod Pathol 2001;14:913) and generally lack expression of BCL2 protein. These two markers (CD10+, BCL2-) can be used to differentiate PCFCL from DLBCL, leg type, which is typically CD10-, BCL2+.

Comment Here

Reference: Diffuse large B cell lymphoma - leg
Board review style question #2
    Which of the following factors can be used as a predictor of an unfavorable prognosis for the above lymphoma entity?

  1. EBV positivity
  2. Gender
  3. Lack of MYD88 L265P mutation
  4. Presence of multifocal lesions
  5. Presence of t(14;18)(q32;q21) translocation
Board review style answer #2
D. Presence of multifocal lesions

Comment Here

Reference: Diffuse large B cell lymphoma - leg

Primary cutaneous follicle center lymphoma
Definition / general
  • Low grade B cell lymphoma arising in skin composed of follicular center B cells without evidence of systemic / nodal involvement at the time of diagnosis
Essential features
  • Mainly centrocytes; predominantly large centrocytes
  • Follicular, diffuse or follicular and diffuse growth pattern
  • Presence of monoclonal B cells in the right clinical and pathologic context can support a diagnosis of primary cutaneous follicle center lymphoma (PCFCL)
  • Diffuse growth of centroblasts and immunoblasts excludes this diagnosis
  • Clinically and genetically distinct from nodal follicular lymphoma
Terminology
ICD coding
  • ICD-O: 9597/3 - primary cutaneous follicle centre lymphoma
  • ICD-10: C86.6 - primary cutaneous CD30 positive T cell proliferations
Epidemiology
  • ~10% of all primary cutaneous lymphomas
  • Most common (~50%) type of primary cutaneous B cell lymphomas
  • Middle aged adults (i.e. 50 - 60 years)
  • M:F = ~1.5:1
  • Reference: Blood 2005;105:3768
Pathophysiology
Etiology
Diagrams / tables

Table 1: differential diagnosis of primary cutaneous follicle center B cell lymphoma
Criteria Primary cutaneous follicle center lymphoma (PCFCL) Secondary follicular lymphoma Primary cutaneous marginal zone lymphoma Primary cutaneous diffuse large B cell lymphoma, leg type
Age 50 - 60 years ~60 years ~55 years 70 years
Gender M > F M > F M > F F > M
Stage Low (I - II) High (III - IV) in majority of cases Low, limited to skin Low to high
Most common location Head and neck Variable Trunk, arms or head Lower leg(s)
Extracutaneous spread Absent Present, variable Rare (< 10%) Present, ~30%
Histology Mixture of centrocytes and centroblasts (no grading required) in follicular, follicular and diffuse or diffuse growth patterns; background fibrosis and sclerosis with stromal reaction present Mixture of centrocytes and centroblasts in follicles (grading based on number of centroblasts); background fibrosis and sclerosis with stromal reaction present Polymorphous: small lymphocytes, small to medium sized marginal zone (centrocyte-like) cells with pale cytoplasm, lymphoplasmacytic cells, plasma cells and occasional large, transformed lymphoid cells Centroblasts or immunoblasts (large noncleaved cells) in diffuse growth pattern; no background stromal reaction
BCL2 (IHC) Negative to weak positive Usually positive Usually positive Positive
CD10 Positive in follicular pattern (< 25%); negative in diffuse pattern Positive (usually) Negative Negative
BCL6 (IHC) Positive (~100%) Positive (usually) Negative Positive or negative
Ki67 Variable, usually > 30% Low (except for high grade)
BCL2 rearrangement / t(14;18) Absent (10 - 40% can be positive) Positive (~85%) Absent Present (~50%)
BCL6 or MYC rearrangements Absent Variably present Rare cases with BCL6 rearrangement BCL6 (30%), MYC (30%)
Cytogenetic findings Deletion of 14q32.33; c-REL amplifications (~60%) Complex
  • t(14;18)(q32;q21);IGH-MALT1
  • t(11;18)(q21;q21);BIRC3-MALT1
  • t(3;14)(p14.1;q32);IGH-FOXP1
Inactivation / deletion of CDKN2a (9p21.3) & CDKN2B genes, deletion of 6q arm (BLIMP1; 60%)
Molecular findings Mutations in any chromatin modifying genes (CREBBP, EZH2, KMT2D, EP300) are seen in < 10% of cases; gene expression profiling similar to germinal center B cell-like type DLBCL Mutations in any chromatin modifying genes (CREBBP, EZH2, KMT2D, EP300) are common MYD88 (6%), FAS (63%), SLAMF1 (24%), SPEN (18%) and NCOR2 (13%) MYD88 (60%), CD79B (20%), CARD11 (10%), TNFAIP3 / A20 (40%); gene expression profiling similar to activated B cell-like type
Prognosis Favorable, 95% at 5 years (disease free survival) Variable, 20 - 75% at 5 years 95 - 100% at 5 years Variable, ~50% at 5 years
References: Blood Adv 2021;5:649, Front Oncol 2020;10:651, Am J Clin Pathol 2020;154:428
Clinical features
Diagnosis
  • Diagnosis made by:
    • Histology and immunohistochemical evaluation of a skin biopsy
      • Lymphoma involving the dermis, with follicular to diffuse growth pattern
      • Mixture of centrocytes and centroblasts
  • Diagnosis is supported by:
    • Flow cytometry immunophenotyping, if performed
    • Immunoglobulin heavy / light chain gene rearrangement studies, if performed
  • Reference: StatPearls: Primary Cutaneous Follicle Center Lymphoma [Accessed 7 June 2021]
Radiology description
  • PET / CT: negative for lymphadenopathy
  • Dissemination to extracutaneous sites is uncommon (< 10% of patients) (J Clin Oncol 2006;24:1376)
Prognostic factors
Case reports
Treatment
  • Single cutaneous lesion
    • Local therapy with:
      • Either surgical excision or local radiation
      • In case of cutaneous relapse: radiotherapy
    • Generalized skin lesions:
      • Systemic intralesional administration of rituximab or systemic rituximab
  • Reference: Blood 2008;11:1600
Clinical images

Images hosted on other servers:

PCFCL in forehead and scalp

Microscopic (histologic) description
  • Infiltrate is based in the superficial dermis:
    • Lymphoma may involve the entire dermis
    • Often follicles of the tumor extending into the subcutis
    • Perivascular, periadnexal, nodular and diffuse infiltrates
    • Follicles are ill defined; lack tingible body macrophages and mantle zone
  • Intact grenz zone
  • Growth pattern varies:
    • Follicular (mainly head and neck)
    • Follicular and diffuse
    • Purely diffuse (primarily trunk)
  • Mixture of centrocytes and centroblasts
  • Large cells are variable and can be polylobated
  • Predominance of large centrocytes (large cleaved cells), may be spindle shaped
  • Variable numbers of centroblasts
  • T cells are abundant
  • No WHO grading of lymphoma required as in nodal / systemic follicular lymphoma
  • Reference: Arch Pathol Lab Med 2018;142:1313
Microscopic (histologic) images

Contributed by Beenu Thakral, M.D.

Panoramic view of skin biopsy

Neoplastic follicle

CD20

BCL6


BCL2

CD10

CD21

Ki67

Cytology description
  • Cells resembling centrocytes and centroblasts (Jaffe: Hematopathology, 2nd Edition, 2016)
  • Large centrocytes: dispersed chromatin and small nucleoli
  • Large noncleaved cells: prominent and paracentral nucleoli
Positive stains
Negative stains
Flow cytometry description
Molecular / cytogenetics description
Sample pathology report
  • Skin, left cheek, biopsy
    • Low grade follicular lymphoma involving skin (see comment)
    • Comment: Histologic sections demonstrate skin involved by lymphoma. The lymphoma has a vague nodular pattern and is composed of a mixture of large noncleaved and small cleaved lymphoid cells with predominance of small lymphoid cells. The epidermis is unremarkable. No epidermotropism noted. Plasma cells are not increased. Immunohistochemical studies performed demonstrate that the neoplastic cells are positive for CD10, CD20, PAX5, BCL6 and are negative for BCL2, cyclin D1 and MUM1. CD3 and CD5 highlight small reactive lymphocytes in the background. Ki67 demonstrates a proliferation index of ~20%.
    • In the appropriate clinical setting, this neoplasm involving the skin is consistent with primary cutaneous follicle center cell lymphoma. Clinical correlation and staging studies may help to confirm this diagnosis. If the neoplasm is systemic, this follicular lymphoma can be classified as low grade.
Differential diagnosis
Board review style question #1

CD20

BCL6

BCL2



A 50 year old man presented with a solitary and erythematous lesion on the scalp for ~3 weeks. He does not recall any similar lesion(s) in the past. On physical exam, no splenomegaly or lymphadenopathy is noted. A skin biopsy is performed and a diagnosis of primary cutaneous follicle center B cell lymphoma is rendered. Which of the following statements regarding this lesion is most accurate?

  1. Deletions of a small region on chromosome 9p21.3 containing CDKN2A and CDKN2B gene loci are frequently reported
  2. They are generally BCL2 negative, have a good prognosis and do not require grading for prognosis
  3. Majority of these lesions have t(14;18) translocation or BCL2 gene rearrangement
  4. MYD88 mutation is frequently reported in this lesion
Board review style answer #1
B. They are generally BCL2 negative, have a good prognosis and do not require grading for prognosis

Comment Here

Reference: Primary cutaneous follicle center lymphoma
Board review style question #2
Which of the following findings best matches with the diagnosis of primary cutaneous follicle center lymphoma (PCFCL)?

  1. CD20+, CD10-, BCL2+, BCL6+ with IGH-BCL2 gene rearrangement
  2. CD20+, CD10-, BCL2-, BCL6+with IGH-BCL2 gene rearrangement
  3. CD20+, CD10-, BCL2-, BCL6+ in the absence of IGH-BCL2 gene rearrangement
  4. CD20+, CD10+, BCL2+, BCL6+ with IGH-BCL2 gene rearrangement
  5. CD20+, CD10-, BCL2-, BCL6- in the absence of IGH-BCL2 gene rearrangement
Board review style answer #2
C. CD20+, CD10-, BCL2-, BCL6+ in the absence of IGH-BCL2 gene rearrangement (see Positive stains, Negative stains and Molecular / cytogenetics description)

Comment Here

Reference: Primary cutaneous follicle center lymphoma

Primary cutaneous gamma delta

Primary cutaneous marginal zone lymphoma
Definition / general
  • B cell lymphoma derived from mature, predominantly postgerminal center B cells
  • Subsumed under the 2016 revised World Health Organization classification of extranodal marginal zone B cell lymphoma of mucosa associated lymphoid tissue (MALT lymphoma) (Blood 2016;127:2375)
  • Nodule or coalescing papules on the torso or upper extremities; lesions are often localized
  • Excellent overall survival with rare reports of transformation to a more aggressive phenotype
Essential features
  • Primary cutaneous marginal zone B cell lymphoma (CMZL) is an indolent lymphoma with a nonspecific postgerminal center B cell phenotype that often presents as coalescing papules or a nodule on the torso or upper extremity of middle aged adults
  • Excellent prognosis, rarely disseminates beyond skin and exceptionally rare cases show progression to an aggressive blastic or large cell appearance
  • Comprised of 2 subtypes with distinct histomorphologic findings, a nonclass switched subtype that shares many similarities with extracutaneous marginal zone lymphomas and a class switched subtype that may be accompanied by a preponderance of reactive T and B lymphocytes
  • Colonization of germinal center follicles by lymphoma is a helpful feature for making the diagnosis; however, CMZL should never exhibit a germinal center immunophenotype and should not be comprised of large and mitotically active cells
  • Demonstration of a light chain restriction by in situ hybridization (or immunohistochemistry) or a clonally rearranged immunoglobulin heavy chain gene by PCR is more common in CMZL than in reactive cutaneous lymphoid hyperplasias; however, clonality is not pathognomonic of lymphoma
Terminology
  • Extranodal marginal zone B cell lymphoma of mucosa associated lymphoid tissue (MALT lymphoma)
ICD coding
  • C88.4: extranodal marginal zone B cell lymphoma of mucosa associated lymphoid tissue (MALT lymphoma)
Epidemiology
  • Young to middle aged men and women, rare childhood cases
  • Approximately 30% of cutaneous B cell lymphoma
Sites
  • Variable but most frequently involves the trunk and upper extremities
  • Many lesions are solitary or localized / regional; generalized spread in a minority of cases
  • Extracutaneous dissemination is exceptional and should raise suspicion for cutaneous dissemination of an extracutaneous lymphoma (e.g. of splenic or nodal origin)
Etiology
  • Etiology is poorly understood, although thought to evolve in the setting of cutaneous lymphoid hyperplasia resulting from a variety of antigenic triggers
  • Chronic antigenic stimulation may lead to immune dysregulation (analogous to the role of chronic Helicobacter pylori infection in the pathogenesis of gastric MALT lymphoma) (J Cutan Pathol 1997;24:457)
  • Two distinct subcategories
    • Nonclass switched
      • More closely resembles extracutaneous MZL in that it arises in the setting of chronic Th1 type inflammation with increased production of Th1 associated cytokines including IFNƴ and IL2 (Blood 2008;112:3355)
      • Borrelia burgdorferi antigen has been identified in a subset of cases from Europe but not in cases from the U.S. or Japan (J Cutan Pathol 1997;24:457, Am J Surg Pathol 2000;24:1279)
    • Heavy chain class switched
      • Majority of cutaneous MZL, is histologically distinct from extracutaneous MZL and arises in the setting of chronic Th2 type inflammation
      • May arise in individuals with an allergic or atopic diathesis (Am J Surg Pathol 2010;34:1830)
  • Some cases associated with medications including methotrexate, cyclosporine, antidepressants and antihistamines (J Cutan Pathol 2006;33:1)
  • Infectious disease associations have also included Helicobacter pylori, Hepatitis C and Epstein Barr virus
  • Autoimmune disease associations include polyarteritis nodosa, Sjögren syndrome, rheumatoid arthritis, Hashimoto thyroiditis and ulcerative colitis (Appl Immunohistochem Mol Morphol 2004;12:216)
  • Recurrent mutations involving FAS that affected the death domain of the apoptosis regulating FAS / CD95 protein were recently identified in 24 of 38 cases (J Invest Dermatol 2018 Feb 23 [Epub ahead of print])
Clinical features
  • Erythematous to violaceous nodule or plaque comprised of coalescing papules
Diagnosis
  • Clinical correlation (complete history, review of systems, physical examination, complete blood count and imaging studies) are required to exclude skin involvement by lymphoma of extracutaneous origin
Laboratory
  • Serologic testing for Borrelia burgdorferi may be helpful for excluding reactive lymphoid hyperplasia within Europe (no demonstrable association between the organism and CMZL in U.S. or Japan)
Prognostic factors
  • Excellent 5 year overall survival of > 95%
  • Small minority (4%) experience dissemination beyond the skin (J Am Acad Dermatol 2013;69:357)
  • Relapse occurs in less than half of patients and is more common in patients with multifocal skin involvement (J Am Acad Dermatol 2013;69:357)
  • Rare cases undergo transformation to an aggressive, blastic or large cell phenotype analogous to what is reported in splenic B cell lymphoma (APMIS 2007;115:1426)
Case reports
Treatment
  • Excision, radiotherapy and intralesional steroids for skin limited localized disease
  • Withdrawal of any agent associated with this disease, often an antihistamine or antidepressant, could also be considered
Clinical images

Images hosted on other servers:

Cutaneous lymphoma differential diagnosis

Microscopic (histologic) description
  • In general, CMZL exhibits a nodular and diffuse architecture described as having a grenz zone and a bottom heavy appearance (no tapering with descent to the deep dermis and superficial subcutis)
  • Neoplastic B cells can have 3 appearances
    • Monocytoid appearance
    • Cleaved nuclei reminiscent of centrocytes
    • Small round cells reminiscent of CLL / SLL
  • Plasma cells and plasmacytoid cells may be conspicuous at the periphery of dermal nodules
  • Nonclass switched CMZL often has dense sheets and nodules filling the dermis
  • Class switched CMZL often has a preponderance of T cells and reactive germinal B cell follicles accompanying the neoplastic B cell population, which ranges in appearance from dense nodules to perivascular infiltrates or sometimes sparse, scattered cells
  • Class switched CMZL can show variable plasmacytic differentiation and extreme examples were formerly called immunocytoma (dense perivascular plasma cell aggregates and binucleated forms, nuclear pleomorphism and Dutcher bodies are helpful hints to this diagnosis)
Microscopic (histologic) images

Contributed by Robert E. LeBlanc, M.D.

Monotonous monocytoid B cells

IHC expression

BCL2

BCL6


CD3

CD20

CD21

Virtual slides

Images hosted on other servers:

Cutaneous marginal zone lymphoma

Positive stains
  • Kappa or lambda light chain restriction in plasma cells or plasmacytoid forms has greater sensitivity for the detection of B cell monoclonality in CMZL than PCR detection of clonal IGH rearrangement
  • B cells most often exhibit a mature, postgerminal center immunophenotype
  • CD20 (may not be detected in patients who have received rituximab)
  • CD79a
  • BCL2
  • CD43 (not always expressed)
  • Colonization of germinal center follicles by neoplastic B cells (lymphoma cells are present within former germinal center follicles delineated by CD21 positive follicular dendritic cell networks and may surround diminished germinal centers that express BCL6 and CD10)
  • T cells can may outnumber B cells and show a preponderance of CD4 positive forms with numerous cells expressing T follicular helper markers (PD-1 and BCL6)
  • CXCR3, while not necessary for diagnosis, is more likely to be expressed in nonclass switched CMZL (Th1 dominant) along with IgM (the same as extracutaneous MZL)
  • CXCR3 is less likely to be expressed in class switched CMZL (Th2 dominant) along with IgG (most cases) and or other isotypes in a minority of cases (Blood 2008;112:3355)
Negative stains
  • BCL6 (may highlight diminished germinal center constituents in colonized follicles)
  • CD10 (may highlight diminished germinal center constituents in colonized follicles)
  • CD5 (positive in rare cases including some with progressive disease)
  • Cyclin D1
Molecular / cytogenetics description
  • Clonal rearrangement of the immunoglobulin heavy chain supports the diagnosis; however, this test suffers from low sensitivity owing to the fact that a large proportion of the infiltrate can be comprised of reactive T and B cells
  • Trisomy 3 is present in at least 60% (Blood 1995;85:2000) with trisomy 7, 12 and 18 in a smaller subset of cases
  • Rare cases show t(11;18)(q21;q21) resulting in API2-MALT1 fusion
  • Rare cases show t(14;18) involving IGH / BCL2 or IGH / MLT1 (Am J Surg Pathol 2003;27:702)
  • Report of t(3;14) IGH - FOXP1 (Leukemia 2005;19:652)
  • 7q deletions, which can also be seen in some splenic lymphomas, have been described in a subset of cases including ones that underwent transformation to an aggressive blastic phenotype (Am J Dermatopathol 2013;35:319)
Sample pathology report
  • Skin, left upper chest, excision:
    • Clonal B cell infiltrate, suspicious for cutaneous marginal zone lymphoma (see comment)
    • Comment: The dense dermal infiltrate comprised of small, mature B cells colonizes germinal center follicles. In the provided context of a solitary nodule on the chest of an adult, the immunophenotype and evident lambda light chain restriction provide support for a diagnosis of primary cutaneous marginal zone lymphoma. Correlation with clinical features is essential in order to confirm this impression.
Differential diagnosis
Board review style question #1
Which of the following stains highlight only B cells and not the accompanying T cells in a case of T cell rich primary cutaneous marginal zone B cell lymphoma?

  1. BCL2
  2. BCL6
  3. CD79a
  4. PD-1
Board review style answer #1
C. CD79a. BCL2 can be challenging to evaluate as it will highlight both the neoplastic B cells comprising marginal zone lymphoma as well as accompanying T lymphocytes. Cases of T cell rich marginal zone lymphoma can contain a substantial population of T cells exhibiting a T follicular helper immunophenotype with variable expression of PD-1 and BCL6. These markers should not be expressed in the neoplastic B cells of marginal zone lymphoma.

Comment Here

Reference: Cutaneous marginal zone lymphoma / MALT
Board review style question #2
Which of the following findings favors the diagnosis of primary cutaneous follicle center lymphoma over primary cutaneous marginal zone B cell lymphoma?

  1. B cells with a germinal center phenotype extending beyond CD21+ dendritic cell networks
  2. Confluent Ki67 expression by neoplastic B cells throughout the germinal center follicles
  3. Presence of circumscribed dermal aggregates
  4. Presence of kappa light chain-restricted plasma cells
Board review style answer #2
A. B cells with a germinal center immunophenotype extend beyond CD21 positive dendritic cell networks in primary cutaneous follicle center lymphoma. This is distinct from colonization of germinal center follicles present in some cases of marginal zone lymphoma. Circumscribed, nodular dermal aggregates can be identified in both lymphomas. A kappa light chain-restricted plasma cell population could be seen in marginal zone lymphoma. Lastly, Ki67 staining appears decreased in neoplastic germinal centers of follicle center lymphoma.

Comment Here

Reference: Cutaneous marginal zone lymphoma / MALT

Primary cutaneous mucinous carcinoma
Definition / general
  • Also called adenocystic carcinoma
  • Scalp of elderly patients
  • Caucasian > Black > Asian
Case reports
Clinical images

Contributed by Mark R. Wick, M.D.
Gross description
  • Slow growing, solitary, asymptomatic, flesh colored nodule with a slightly translucent surface
Microscopic (histologic) description
  • Resembles mammary colloid carcinoma with lakes of mucin containing small tumor cell clusters
  • May also have an infiltrating ductal pattern
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D., Case #413 and @JMGardnerMD on Twitter

H&E images

CK5/6

CK7


CK20

ER

PR

GATA3


Primary cutaneous mucinous carcinoma

Primary cutaneous mucinous carcinoma



Images hosted on other servers:

Gluteal mass

Positive stains
  • Primary skin tumors: 100% CK7+, 40% showed rare p63+ cells, 20% had focal CK5/6+
Differential diagnosis

Proliferating pilar tumor
Definition / general
  • Proliferating pilar tumor (PPT) is an uncommon neoplasm within the dermis or subcutis that arises from the isthmus region of the outer root sheath of the hair follicle
  • Generally develops from a pilar cyst (sometimes following trauma) but may also arise de novo
Essential features
  • Most commonly arises on the scalp of elderly women
  • Histologically resembles a trichilemmal cyst but with central lobules of squamous proliferation and variable atypia and mitotic figures
  • Complete excision and clinical follow up is recommended due to the small risk for recurrence and metastasis
Terminology
  • Proliferating pilar cyst
  • Proliferating trichilemmal cyst
ICD coding
  • ICD-10: L72.11 - pilar cyst
  • ICD-11: EK70.1 - trichilemmal cyst
Epidemiology
Sites
Pathophysiology
Etiology
  • Genetic basis of hereditary pilar cysts has been reported as mutations in PLCD1; it is not known whether proliferating pilar tumors share this genetic alteration (Sci Rep 2020;10:6035)
Clinical features
  • Slowly growing nodule on the scalp
  • Lesions are generally well circumscribed, although they may be multinodular
  • Tend to shell out from surrounding tissue upon surgical excision, similar to pilar cysts
  • Average size is several centimeters; however, tumors up to 25 cm have been reported (Cancer 1971;28:701)
Diagnosis
  • Based on histopathologic findings following skin biopsy or excision of the nodule
Prognostic factors
  • Nonscalp location, recent rapid growth, size > 5 cm and histologic features of extensive necrosis, high mitotic activity, poor circumscription with areas of invasion and cytologic atypia are all concerning for more aggressive or malignant behavior (J Cutan Pathol 2003;30:492)
  • Local recurrence rate is 6.6%; risk of lymph node involvement is 2.6% (Am J Clin Pathol 2004;122:566)
Case reports
  • 33 year old woman with multiple proliferating pilar tumors and a porokeratotic adnexal ostial nevus (JAAD Case Rep 2020;6:344)
  • 46 and 54 year old women with proliferating pilar tumors treated with Mohs micrographic surgery (J Drugs Dermatol 2021;20:1346)
  • 76 year old woman with a proliferating trichilemmal cyst with delayed treatment due to COVID outbreak (Front Surg 2021;8:680160)
Treatment
  • Complete surgical excision, either via wide local excision or Mohs micrographic surgery (Dermatol Surg 2007;33:1102)
  • If the lesion was enucleated in its entirety, well circumscribed and without other concerning histologic features, additional surgical therapy is not required
  • Ongoing surveillance to monitor for recurrence or metastasis is recommended
Clinical images

Contributed by Viktoryia Kazlouskaya, M.D., Ph.D.

Large cystic lesion

Gross description
  • Encapsulated, pearly white nodule; may be multinodular
Microscopic (histologic) description
  • Features of a pilar cyst with abrupt trichilemmal keratinization with additional extensive epithelial proliferation within the center of the cystic space
  • Mitotic activity and cytologic atypia may be variable
  • Invasion into the surrounding soft tissue should not be present
  • Grading criteria as suggested by Ye et al. stratifies proliferating pilar tumors into 3 groups based on histologic characteristics (Am J Clin Pathol 2004;122:566):
    • Group 1:
      • Circumscribed silhouettes with pushing margins, modest nuclear atypia and an absence of pathologic mitoses, necrosis and invasion of nerves or vessels
      • Behaves in a benign manner
    • Group 2:
      • Similar to group 1 but manifest irregular, locally invasive silhouettes with involvement of the deep dermis and subcutis
      • Small risk of local recurrence
    • Group 3:
      • Invasive growth patterns, marked nuclear atypia, pathologic mitotic forms and geographic necrosis, with or without the involvement of nerves or vascular structures
      • Potential for regional recurrence and metastasis
Microscopic (histologic) images

Contributed by Viktoryia Kazlouskaya, M.D., Ph.D. and Colleen J. Beatty, M.D.

Lesion with trichilemmal differentiation

Proliferating lesion, trichilemmal differentiation

Trichilemmal differentiation without atypia

Proliferative cystic lesion


Proliferative trichilemmal lesion

Cyst with proliferative areas

Proliferative area with trichilemmal differentiation

Videos

Proliferating pilar tumor overview by Dr. Gardner

Sample pathology report
  • Skin, scalp, biopsy:
    • Proliferating pilar tumor (see comment)
    • Comment: Examination reveals a well circumscribed nodule composed of lobules of proliferative squamous epithelium showing trichilemmal keratinization with an adjacent typical pilar cyst. No significant cytologic atypia or atypical mitoses are observed. The lesion appears to be excised in total; however, clinical correlation and follow up are recommended.
Differential diagnosis
  • Pilar (trichilemmal) cyst:
    • Well circumscribed cyst lined by stratified squamous epithelium exhibiting trichilemmal keratinization
  • Squamous cell carcinoma:
    • Malignancy of epidermal keratinocytes exhibiting invasion and variable degrees of atypia
    • Key differentiators are absence of areas of trichilemmal keratinization and a lack of staining with markers of outer root sheath differentiation
  • Malignant proliferating pilar tumor:
    • Features of a proliferating pilar tumor but with invasion into the surrounding tissue along with atypical mitoses, marked nuclear atypia and geographic necrosis
Board review style question #1
Proliferating pilar tumors are thought to arise from pilar cysts, potentially incited by trauma to a pre-existing pilar cyst. What is a histologic hallmark of a proliferating pilar tumor?

  1. Cyst with eosinophilic cuticle and associated sebaceous glands
  2. Cystic lining with granular layer and keratinization
  3. Lobules of squamous epithelium arising within a cyst with trichilemmal keratinization
  4. Proliferation of hair matrical cells and ghost cells
Board review style answer #1
C. Lobules of squamous epithelium arising within a cyst with trichilemmal keratinization

Comment Here

Reference: Proliferating pilar tumor
Board review style question #2

The following lesion is found on the scalp of an elderly woman. What is the diagnosis?

  1. Pilomatricoma
  2. Proliferating pilar cyst
  3. Squamous cell carcinoma
  4. Steatocystoma
Board review style answer #2
B. Proliferating pilar cyst

Comment Here

Reference: Proliferating pilar tumor

Pseudocarcinomatous hyperplasia (pending)
[Pending]

Reactive angioendotheliomatosis (pending)
[Pending]

Rosai-Dorfman disease (RDD)

Sézary syndrome

Sclerotic fibroma
Definition / general
  • Solitary lesions are known as circumscribed storiform collagenoma
  • Some cases may represent folliculitis
  • May be associated with Cowden disease
Case reports
Microscopic (histologic) description
  • Bland, well circumscribed hypocellular lesion with focal heavy collagen deposition
Microscopic (histologic) images

Case #190

H&E and CD34

Positive stains

Sebaceoma
Definition / general
  • Benign sebaceous neoplasm composed of basaloid cells and mature sebocytes
  • Associated with Muir-Torre syndrome (MTS) but can also be an isolated (sporadic) tumor
Terminology
  • Sebaceoma, as a term, is used for tumors with > 50% basaloid cells and sebaceous adenoma is used for tumors with < 50% basaloid cells
  • Sebaceous epithelioma, as a term, is confusing and is best not used
    • Sebaceous epithelioma could refer to a low grade form of sebaceous carcinoma, a basal cell carcinoma with sebaceous differentiation or sebaceous proliferations of uncertain potential and thus is not a useful term
ICD coding
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
Epidemiology
  • F:M = 4:1
  • Wide age range but the majority of patients are in the sixth to ninth decades
Sites
  • Primarily occurs on the face and scalp
  • Occurrence on non head and neck regions should prompt consideration of Muir-Torre syndrome
Clinical features
  • Yellow to orange or flesh colored papule, nodule or tumor

  • Muir-Torre syndrome (MTS) is considered a phenotypic variant of hereditary nonpolyposis colorectal carcinoma syndrome (HNPCC, Lynch syndrome) and is caused by germline mutations in one allele of the DNA mismatch repair (MMR) genes, most commonly MLH1, MSH2, MSH6 and PMS2
    • MTS is associated with sebaceous neoplasia (most commonly sebaceous adenoma), colorectal, GU and other visceral adenocarcinomas
    • MSH2 is the gene most commonly mutated in MTS
    • If there is concern for MTS (personal or family history of colorectal, GU or breast cancer; or an older patient with numerous sebaceous neoplasms, especially not on the head and neck), further testing for MMR IHC or microsatellite instability can be performed
    • IHC cannot differentiate between loss of MLH1 expression caused by a germline mutation versus a somatic hypermethylation - some germline missense mutations may be erroneously interpreted as normal by IHC, because they may result in an antigenically intact but nonfunctional protein
    • Microsatellite instability analysis (MSI): performed on formalin fixed tissue, is more sensitive at detecting patients with germline MMR defects than IHC
      • Results are either MSI-H (high degree of MSI) or MSI-L (low degree of MSI)
      • Germline mutation analysis: blood leukocyte genomic sequencing is preferred to identify germline mutations in the MLH1, MSH2, MSH6 and PMS2 genes - if there are no germline mutations, but the tumor shows MMR deficiency, there may be involvement of other MMR proteins, somatic mutations or hypermethylation of the promoter region
Prognostic factors
  • Sebaceomas can be sporadic, not associated with MTS
  • Loss of nuclear staining for MLH1, MSH2, MSH6 or PMS2 suggests microsatellite instability (MSI) and supports a diagnosis of Muir-Torre syndrome
  • Mutations in MLH1 and MSH2 have the most severe effect, producing a high frequency MSI phenotype
    • When MSH2 is deficient, MSH6 usually is as well, because protein products of MSH2 and MSH6 form a heterodimer MMR recognition factor
    • MSH6 is unstable without MSH2 and is quickly degraded
Treatment
  • Once the diagnosis is established, no further treatment is needed
  • However, complete excision should be considered if the tumor is only partially biopsied or there is concern for basal cell carcinoma with sebaceous differentiation or sebaceous carcinoma
Clinical images

Contributed by Oriol Corral-Magaña, M.D. and Luis Javier del Pozo, M.D.
Missing Image

Adenoma

Microscopic (histologic) description
  • Dermal nodule with variable epidermal involvement consisting of basaloid cells and mature sebocytes
  • Greater than 50% of the tumor is made up of basaloid cells
  • Variants of sebaceoma have been described with carcinoid-like, reticulated, cribriform, rippled and Verocay body-like features
Microscopic (histologic) images

Contributed by Jerad Gardner, M.D. and Jennifer Kaley, M.D.
Missing Image

Sebaceoma

Missing Image Missing Image

Sebaceoma


Missing Image

Sebaceoma

Missing Image

Ripple pattern sebaceoma

Positive stains
Negative stains
Videos

Muir-Torre Syndrome

Differential diagnosis
  • Basal cell carcinoma with sebaceous differentiation:
    • Predominantly shows peripheral palisading and cleft formation with incidental sebaceous differentiation
    • BerEP4 staining is positive in basal cell carcinoma and negative in sebaceoma
  • Sebaceous adenoma:
    • Tumor with < 50% basaloid cells, as opposed to a sebaceoma which has > 50% basaloid cells
  • Sebaceous carcinoma:
    • Has more nuclear pleomorphism, nucleolar prominence and mitotic activity than sebaceoma, infiltrative growth pattern
    • Trichoblastoma with sebaceous differentiation:
      • Look for focal hair differentiation and papillary mesenchymal bodies
Board review style question #1
    What is the most common skin tumor associated with Muir-Torre syndrome?

  1. Basal cell carcinoma
  2. Sebaceoma
  3. Sebaceous adenoma
  4. Sebaceous carcinoma
Board review style answer #1
C. Sebaceous adenoma

Comment Here

Reference: Sebaceoma
Board review style question #2
    What is the most common mutation seen in Muir-Torre syndrome?

  1. MLH1
  2. MSH2
  3. MSH6
  4. PMS2
Board review style answer #2
B. MSH2

Comment Here

Reference: Sebaceoma

Sebaceoma
Definition / general
Essential features
  • Benign adnexal neoplasm that clinically presents as a solitary, yellow-tan papule / nodule
  • Histologic sections show well circumscribed cellular lobules composed of predominant immature basaloid cells (> 50% of the lesion) with admixed mature sebocytes
  • Associated with Muir-Torre syndrome
Terminology
  • Sebaceous epithelioma, as a term, is confusing and is best not used
    • Sebaceous epithelioma could refer to a low grade form of sebaceous carcinoma, a basal cell carcinoma with sebaceous differentiation or sebaceous proliferations of uncertain potential and thus is not a useful term
ICD coding
  • ICD-O: 8410/0 - sebaceoma
  • ICD-11: 2F22 & XH0QL4 - benign neoplasms of epidermal appendages & sebaceoma
Epidemiology
Sites
Pathophysiology
  • Sebaceomas commonly have mutation of RAS family genes (HRAS and KRAS), TP53, CDKN2A, EGFR and CTNNB1 when examined by next generation sequencing
  • Abnormalities of TP53 were most frequently found (Pathology 2016;48:454)
Etiology
Diagrams / tables
Not applicable
Clinical features
  • Yellow-orange or flesh colored papule, nodule or tumor (Am J Dermatopathol 1984;6:7, Am J Dermatopathol 2002;24:294)
  • Usually a single lesion but can be multifocal in association with Muir-Torre syndrome (Am J Dermatopathol 2000;22:155)
  • Muir-Torre syndrome is considered a phenotypic variant of hereditary nonpolyposis colorectal carcinoma syndrome (HNPCC, Lynch syndrome) and is caused by germline mutations in one allele of the DNA mismatch repair (MMR) genes, most commonly MLH1, MSH2, MSH6 and PMS2
    • Muir-Torre syndrome is associated with sebaceous neoplasia (most commonly sebaceous adenoma), colorectal, genitourinary (GU) and other visceral adenocarcinomas
    • Defective MMR gene products have been observed in association with sebaceoma, including MLH1, MSH2, MSH6 and PMS2; a subset of these are related to Muir-Torre syndrome (J Cutan Pathol 2009;36:613, Arch Pathol Lab Med 2014;138:1685, J Am Acad Dermatol 2016;74:558, Am J Dermatopathol 2017;39:239, Mod Pathol 2011;24:1004, Histopathology 2010;56:133, Am J Dermatopathol 2021;43:174)
    • MSH2 is the gene most commonly mutated in Muir-Torre syndrome
    • If there is concern for Muir-Torre syndrome (personal or family history of colorectal, GU or breast cancer; an older patient with numerous sebaceous neoplasms, especially not on the head and neck), further testing for MMR IHC or microsatellite instability can be performed
    • IHC cannot differentiate between loss of MLH1 expression caused by a germline mutation versus a somatic hypermethylation; some germline missense mutations may be erroneously interpreted as normal by IHC because they may result in an antigenically intact but nonfunctional protein
    • Microsatellite instability (MSI) analysis, performed on formalin fixed tissue, is more sensitive at detecting patients with germline MMR defects than IHC
      • Results are either MSI-H (high degree of MSI) or MSI-L (low degree of MSI)
      • Germline mutation analysis: blood leukocyte genomic sequencing is preferred to identify germline mutations in the MLH1, MSH2, MSH6 and PMS2 genes; if there are no germline mutations but the tumor shows MMR deficiency, there may be involvement of other MMR proteins, somatic mutations or hypermethylation of the promoter region
  • Head and neck sebaceomas tend to be MMR stable as compared to sebaceomas outside the head and neck regions, which tend to be MMR deficient (Am J Surg Pathol 2008;32:936, Am J Dermatopathol 2021;43:174)
  • Other mutations have been identified in sebaceoma with next generation sequencing studies, including TP53 (most common), RAS family genes (HRAS and KRAS), CDKN2A, EGFR and CTNNB1 (Pathology 2016;48:454)
  • Sebaceoma can arise in a pre-existing sebaceous nevus (Pathology 2016;48:454)
Diagnosis
  • Solitary, yellow-tan papules / nodule (Am J Dermatopathol 1984;6:7)
  • Well circumscribed cellular lobules comprised mostly of small monomorphic immature but cytologically benign basaloid cells admixed with fewer mature sebocytes (< 50%), haphazardly distributed throughout the tumor (Histopathology 2010;56:133)
Laboratory
Not applicable
Radiology description
Not applicable
Radiology images
Not applicable
Prognostic factors
Case reports
Treatment
  • Once the diagnosis is established, no further treatment is needed if the biopsy margins are negative
  • However, complete excision should be considered if the tumor is only partially biopsied or there is concern for basal cell carcinoma with sebaceous differentiation or sebaceous carcinoma
Clinical images

Contributed by Oriol Corral-Magaña, M.D. and Luis Javier del Pozo, M.D.
Missing Image

Cutaneous nodule

Gross description
Not applicable
Gross images
Not applicable
Frozen section description
Not applicable
Frozen section images
Not applicable
Microscopic (histologic) description
Microscopic (histologic) images

Contributed by Jerad Gardner, M.D., Jennifer Kaley, M.D., Yazan Alhalaseh, M.D. and Dinesh Pradhan, M.D.
Dermal based sebaceous proliferation

Dermal based sebaceous proliferation

Sebaceous proliferation with ducts

Sebaceous proliferation with ducts

Admixed multivacuolated sebocytes

Admixed multivacuolated sebocytes

Nodular dermal basaloid tumor

Nodular dermal basaloid tumor

Basaloid tumor with cystic spaces

Basaloid tumor with cystic spaces

Cysts with eosinophilic lining

Cysts with eosinophilic lining


Multiple epidermal attachments

Multiple epidermal attachments

Basaloid proliferation with sebocytes

Basaloid proliferation with sebocytes

Predominance of basaloid cells

Predominance of basaloid cells

Rippled pattern basaloid proliferation

Rippled pattern basaloid proliferation

Rippled pattern

Rippled pattern

Few sebocytes and ducts

Few sebocytes and ducts


Dermal based basaloid tumor

Dermal based basaloid tumor

Admixed multivacuolated sebocytes

Admixed multivacuolated sebocytes

Cystic features and mucin

Cystic features and mucin

Nodular sebaceous proliferation

Nodular sebaceous proliferation

Predominance of basaloid cells

Predominance of basaloid cells

Sebocytes with scalloped nuclei

Sebocytes with scalloped nuclei


Multiple epidermal attachments

Multiple epidermal attachments

Bland appearing mature sebocytes

Bland appearing mature sebocytes

Few mitotic figures

Few mitotic figures

Well circumscribed lobule

Well circumscribed lobule

Irregular shaped dermal nodules

Irregular shaped dermal nodules

Cystic structures and sebocytes

Cystic structures and sebocytes

Virtual slides

Contributed by Yazan Alhalaseh, M.D. and Dinesh Pradhan, M.D.
Sebaceoma with cystic features

Sebaceoma with cystic features

Cytology description
Not applicable
Cytology images
Not applicable
Immunofluorescence description
Not applicable
Immunofluorescence images
Not applicable
Negative stains
Electron microscopy description
Not applicable
Molecular / cytogenetics description
Not applicable
Molecular / cytogenetics images
Not applicable
Videos

Muir-Torre syndrome

Sample pathology report
  • Skin, scalp nodule; biopsy:
    • Sebaceoma
Differential diagnosis
  • Basal cell carcinoma with sebaceous differentiation:
    • Predominantly shows peripheral palisading, mucinous stroma and retraction artifact with incidental sebaceous differentiation
    • BerEP4 staining is positive in basal cell carcinoma and negative in sebaceoma (Histopathology 2007;51:80)
    • EMA and D2-40 are negative in BCC
  • Sebaceous adenoma:
    • Preserved lobular architecture with distinct and regular maturation (similar to the normal sebaceous gland)
    • Tumor with < 50% basaloid cells, as opposed to a sebaceoma, which has > 50% basaloid cells
  • Sebaceous carcinoma:
    • Has more nuclear pleomorphism, nucleolar prominence, mitotic activity and infiltrative growth pattern, as compared to sebaceoma
    • Diffuse p53 expression and high Ki67 proliferation rate (N Am J Med Sci 2015;7:275)
  • Trichoblastoma with sebaceous differentiation:
    • Look for focal hair differentiation and papillary mesenchymal bodies
Board review style question #1
Which immunohistochemical stain favors a diagnosis of basal cell carcinoma (BCC) with sebaceous differentiation over sebaceoma?

  1. EMA
  2. D2-40
  3. BerEP4
  4. Factor XIIIa
Board review style answer #1
C. BerEP4. BerEP4 is the only choice that is positive in BCC with sebaceous differentiation. Answers A, B and D are incorrect because EMA, D2-40 and factor XIII are negative in BCC with sebaceous differentiation and positive in sebaceoma.

Comment Here

Reference: Sebaceoma
Board review style question #2
Which immunohistochemical stain is helpful in distinguishing sebaceoma from sebaceous carcinoma?

  1. p53
  2. Ki67
  3. Adipophilin
  4. EMA
  5. p53 and Ki67
Board review style answer #2
E. p53 and Ki67. Diffuse p53 expression and a high Ki67 support a diagnosis of sebaceous carcinoma whereas low p53 expression and low Ki67 favor a diagnosis of sebaceoma.

Comment Here

Reference: Sebaceoma
Board review style question #3

A 60 year old man presents with an erythematous nodule on the scalp. An excisional biopsy is shown above. What is your diagnosis?

  1. Basal cell carcinoma
  2. Sebaceoma
  3. Sebaceous adenoma
  4. Sebaceous carcinoma
Board review style question #3
B. Sebaceoma. The histologic sections show well circumscribed cellular lobules composed of predominant immature basaloid cells (> 50% of the lesion) with admixed mature sebocytes. Answer C is incorrect because sebaceous adenoma has > 50% mature sebocytes and < 50 basaloid components. Answer D is incorrect because the tumor cells in sebaceous carcinoma lack atypia, necrosis and have low mitotic activity.

Comment Here

Reference: Sebaceoma

Sebaceous adenoma
Definition / general
  • Benign neoplasm composed of mature sebaceous lobules with the expansion of germinative basaloid cell layers at the periphery
  • Associated with Muir-Torre syndrome, especially when it arises outside the head and neck region (Histopathology 2010;56:133)
  • Unrelated to adenoma sebaceum (facial angiofibromas of tuberous sclerosis)
Essential features
  • Well circumscribed, multilobulated tumor
  • Increased number of basaloid germinative cells of more than 2 layers
  • Shows distinct maturation, meaning that the basaloid germinative cells are usually at the periphery and the sebaceous cells are located centrally
  • Associated with Muir-Torre syndrome, a variant of Lynch syndrome with mutations in the DNA mismatch repair (MMR) genes
ICD coding
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
Epidemiology
Sites
  • Mostly in the head and neck, particularly on the face and scalp
Pathophysiology
  • Subset of these lesions has inactivating mutations in LEF1 affecting the Wnt / β catenin pathway, which is important in sebaceous differentiation (Nat Med 2006;12:395)
Clinical features
  • Clinically presents as a yellow or pink-tan papule or nodule
  • Clinically can be mistaken for basal cell carcinoma
  • Muir-Torre syndrome (MTS) is a clinical variant of Lynch syndrome, which is defined as at least 1 sebaceous neoplasm (sebaceous adenoma, sebaceoma and sebaceous carcinoma) or keratoacanthoma and at least 1 Lynch syndrome related internal cancer
  • MTS is caused by germline variants in the DNA mismatch repair (MMR) genes encoding for MSH2 and MLH1, accounting for most of the cases
  • Isolated mutations in MSH6 and PMS2 account for a small minority of cases (J Cutan Pathol 2017;44:931)
  • MUTYH associated polyposis (MAP) is an autosomal recessive disorder associated with colorectal polyps (adenomas) and adenocarcinomas that can also be associated with cutaneous sebaceous neoplasms, closely imitating MTS (Am J Dermatopathol 2016;38:915)
  • Overall quality of evidence in support of MMR IHC reflex testing on MTS associated cutaneous neoplasms is weak to moderate
  • Some pathologists do targeted testing specially if patient has multiple sebaceous neoplasms, outside the head and neck or history of colorectal, breast or urothelial cancer (J Cutan Pathol 2017;44:931)
Diagnosis
  • Diagnosis is made using clinical examination and biopsy
Prognostic factors
  • Solitary tumors are treated by complete surgical removal with a 100% cure rate
  • Incomplete removal has occasionally resulted in local recurrence (Pathology 2017;49:688)
Case reports
  • 57 year old man with a history of colon cancer diagnosed at age 32 and multiple sebaceous adenomas (Cureus 2021;13:e14582)
  • 60 year old man, with a history of kidney transplantation 11 years prior, presented with new lesion on his left lower abdomen, which was sebaceous adenoma (JAAD Case Rep 2019;5:818)
  • 68 year old man presented to the dermatology clinic with a solitary enlarging, bleeding lesion on his right central zygoma (Dermatol Arch 2019;3:77)
Treatment
  • Because sebaceous adenomas are benign, treatment for most individuals is conservative management, although bothersome lesions can be removed for patient comfort
  • Consider complete excision also if the lesion is partially biopsied or of clinical concern for carcinoma with sebaceous differentiation (Pathology 2017;49:688)
Clinical images

Contributed by Michael Inskip, M.B.Ch.B. and Mark R. Wick, M.D.
Yellow lobules visualized on dermoscopy

Yellow lobules visualized on dermoscopy

Ulcerated papule on the face

Ulcerated papule on the face

Gross description
  • Well circumscribed superficial dermal nodule with sharp demarcation from the underlying tissue
Microscopic (histologic) description
  • Well circumscribed, nodular growth of lobules consist of admixture of basaloid cells and mature sebocytes
  • Some lobules may communicate directly with the surface epithelium
  • Basaloid cells are usually located at the periphery of lobules and sebaceous cells with intracytoplasmic lipid vacuoles, which are usually located at the center of lobules
  • Basaloid cells are composed of expanded germinative layer, with more than the normal 2 cell layers seen in mature sebaceous glands or sebaceous hyperplasia but still less than 50% of the tumor volume (> 50% is seen in sebaceoma / sebaceous epithelioma)
  • Increased mitotic activity is sometimes seen in the basaloid cell component (Surg Pathol Clin 2017;10:367)
Microscopic (histologic) images

Contributed by Ata Moshiri, M.D., M.P.H. and Joel Pinczewski, M.D., Ph.D.
Small tumor at scan

Small tumor at scan

Peripherally basaloid, centrally clear lobules

Peripherally basaloid, centrally clear lobules

Intracytoplasmic lipid droplets

Intracytoplasmic lipid droplets

Larger tumor example

Larger tumor example

Positive stains
Videos

Sebaceous adenoma: 5 minute pathology pearls by Dr. Jerad Gardner

Sample pathology report
  • Skin, right cheek, shave biopsy:
    • Sebaceous adenoma
Differential diagnosis
  • Sebaceous hyperplasia:
    • Hyperplastic sebaceous lobules
    • Located slightly higher in the dermis than normal
    • Individual lobules are increased in number but are not significantly different in size compared to normal
    • Basaloid layer is not expanded
  • Sebaceoma / sebaceous epithelioma:
    • Tumor consists of a random admixture of basaloid cells and mature sebocytes with loss of regular maturation that is seen in sebaceous adenoma
    • No nuclear pleomorphism and mitotic activity is generally sparse; however, it can sometimes be prominent
    • No peripheral palisading or cleft formation
  • Sebaceous carcinoma:
    • Significant nuclear pleomorphism, nucleolar prominence and conspicuous mitotic activity
    • Although in well differentiated variants, the tumor may have a smooth regular margin
    • Less well differentiated examples typically show an infiltrating border
  • Basal cell carcinoma with sebaceous differentiation:
    • Features of basal cell carcinoma with proliferation of palisading basaloid cells, retraction artifact and loose mucinous stroma
    • Within these nodules are foci of variable numbers of mature sebocytes
    • EMA- and BerEP4+ (Can J Ophthalmol 2014;49:326)
  • Trichoblastoma with sebaceous differentiation:
    • Papillary mesenchymal bodies are often present
    • Peripheral nuclear palisading and stromal induction are also generally present
  • Balloon cell melanoma:
  • Other lesions with clear cell differentiation (e.g., metastatic renal clear cell carcinoma)
Additional references
Board review style question #1

Multiple sebaceous adenomas or sebaceous carcinoma outside the head and neck region can be associated with which of the following syndromes?

  1. Birt-Hogg-Dubé (BHD) syndrome
  2. Brooke-Spiegler syndrome
  3. Cowden syndrome
  4. Gorlin syndrome
  5. Muir-Torre syndrome
Board review style answer #1
E. Muir-Torre syndrome (MTS) is a clinical variant of Lynch syndrome, which is defined as at least 1 sebaceous neoplasm (sebaceous adenoma, sebaceoma and sebaceous carcinoma) or keratoacanthoma and at least 1 Lynch syndrome related internal cancer. Additionally, among the spectrum sebaceous neoplasms, sebaceous adenoma is the one most associated with Muir-Torre syndrome.

Comment Here

Reference: Sebaceous adenoma
Board review style question #2
Mutations in which of the following genes are associated with Muir-Torre syndrome?

  1. CYLD
  2. Folliculin
  3. MLH1
  4. PTCH
  5. PTEN
Board review style answer #2
C. MLH1. Muir-Torre syndrome (MTS) is caused by germline variants in the DNA mismatch repair (MMR) genes encoding for MSH2 and MLH1, which account for most of the cases. Isolated mutations in MSH6 and PMS2 account for a small minority of cases (J Cutan Pathol 2017;44:931).

Comment Here

Reference: Sebaceous adenoma

Sebaceous carcinoma
Definition / general
Essential features
  • Periocular versus extraocular
  • Atypical sebocytes can be well, moderately or poorly differentiated
  • May rarely occur in association with Muir-Torre syndrome, an autosomal dominant syndrome characterized by a sebaceous neoplasm (adenoma, sebaceoma or carcinoma) and occasionally keratoacanthoma associated with a visceral malignancy (Dermatol Surg 2015;41:1)
  • Aggressive tumor with 5 year survival rate of 92.7%
  • Treatment is primarily surgical excision
Terminology
ICD coding
  • ICD-10: C44.1392 - sebaceous cell carcinoma of skin of eyelid, including canthus
Epidemiology
Sites
Pathophysiology
  • Unknown
Etiology
  • Radiation
  • Immunosuppression
  • Muir-Torre syndrome: autosomal dominant syndrome with mutation in one or more of the mismatch repair genes (MLH1, MSH2, MSH6 PMS2) and associated microsatellite instability
  • Production of nitrosamines (food or medication)
  • Retinoblastoma (Onco Targets Ther 2018;11:3713)
Clinical features
Diagnosis
Radiology description
Prognostic factors
  • 5 year relative survival rate: 92.7% (J Am Acad Dermatol 2016;75:1210)
  • Overall prognosis for localized disease after complete excision is good
  • Most significant predictor of reduced survival is the presence of metastatic disease at the time of diagnosis
  • Other factors: multicentricity, pagetoid spread, perineural vascular and lymphatic invasion, poorly differentiated cytology, periocular location, primary site on the ear or lip, tumor size (> 10 mm), black race (Curr Treat Options Oncol 2017;18:47, Dermatol Surg 2015;41:1)
  • Sebaceous carcinoma may metastasize in 2.4% of the cases; the majority of metastases occur within the first 2 years after initial treatment (Head Neck 2012;34:1765)
Case reports
Treatment
  • Wide local excision or Mohs micrographic surgery are the best initial treatment options
  • Sentinel lymph node biopsy for periorbital sebaceous carcinoma > 10 mm in diameter
  • Radiation therapy and systemic chemotherapy are only used for patients who are poor surgical candidates or those with recurrent or metastatic disease
  • Immunohistochemistry to evaluate for microsatellite instability (MSH2, MSH6, MLH1, PMS2) should be offered on sebaceous carcinoma specimens, especially extraocular ones, to screen for Muir-Torre syndrome
  • Evidence of orbital invasion on imaging necessitates exenteration (Dermatol Surg 2015;41:1)
Clinical images

Contributed by Maria Del Valle Estopinal, M.D.
Periocular sebaceous carcinoma Periocular sebaceous carcinoma

Periocular sebaceous carcinoma



Images hosted on other servers:

Extraocular sebaceous carcinoma

Muir- Torre syndrome

Gross description
  • Papule, nodule or cystic lesion
Frozen section description
  • Lipid and fat staining with frozen sections have been used in the past
Microscopic (histologic) description
  • Architecture: usually sheets or lobules separated by fibrovascular stroma
    • Dermal based tumor with focal connection to the epidermis or follicular epithelium
    • Can be rounded nodular aggregates or angulated infiltrative aggregates
    • Can appear cystic with central comedo type necrosis
    • Less commonly, can have a broad superficial intraepidermal pattern
  • Cytology: classified as poorly, moderately or well differentiated
    • Well differentiated: increased proportion of mature appearing sebocytes (multivacuolated cells) with nuclear indentation relative to basaloid undifferentiated cells
      • Mild pleomorphism, minimal mitoses and necrosis
    • Moderately to poorly differentiated: higher proportion of atypical basaloid (undifferentiated) cells with minimal differentiation toward multivacuolated cells
      • Prominent pleomorphism and atypia, frequent mitoses and necrosis
  • Pagetoid (or intraepidermal sebaceous carcinoma, sebaceous carcinoma in situ) tumor growth is much more commonly observed in periocular than in extraocular locations, where it is rare
  • Can exhibit squamous metaplasia or focal apocrine differentiation
  • References: Dermatol Surg 2015;41:1, Curr Treat Options Oncol 2017;18:47, Int J Surg Pathol 2019;27:432, J Korean Med Sci 2017;32:1351
Microscopic (histologic) images

Contributed by Bonnie Lee, M.D. and Maria Del Valle Estopinal, M.D.
Well differentiated

Well differentiated

Multivacuolated cells

Multivacuolated cells

Poorly differentiated

Poorly differentiated

Atypical basaloid cells

Atypical basaloid cells

Intraepidermal sebaceous carcinoma

Intraepidermal sebaceous carcinoma


Atypical basaloid cells within the epidermis

Atypical basaloid cells within the epidermis

Poorly differentiated with cystic appearance

Poorly differentiated with cystic appearance

Extensive central (comedo) necrosis

Extensive central (comedo) necrosis

Infiltrative sebaceous carcinoma

Infiltrative sebaceous carcinoma

Infiltrating into skeletal muscle

Infiltrating into skeletal muscle


Androgen receptor

Androgen receptor

Androgen receptor nuclear staining

Androgen receptor nuclear staining

Adipophilin - membranous vesicular staining

Adipophilin - membranous vesicular staining

Adipophilin – granular staining

Adipophilin – granular staining

Positive stains
Negative stains
Sample pathology report
  • Skin, right elbow, shave biopsy:
    • Well differentiated sebaceous neoplasm with severe nuclear atypia, consistent with sebaceous carcinoma (G1)
    • Tumor measures at least 5.5 mm in greatest dimension
    • Margins cannot be evaluated in the plane of sections examined
    • No definite lymphovascular or perineural invasion identified
    • American Joint Committee on Cancer pathologic staging (8th edition): pT1 pNx (Amin: AJJC Cancer Staging Manual, 8th edition, 2017)
  • Left lower eyelid, wedge excision:
    • Recurrent, sebaceous cell carcinoma in situ, moderately differentiated involving the skin of the left lower eyelid
    • Tumor size: 0.7 mm
    • Lymphovascular space invasion: not identified
    • Perineural invasion: not identified
    • Mitotic rate: 5 mitoses per square mm
    • Surgical margins: nasal, temporal, inferior and deep margins are negative for sebaceous carcinoma
    • Microsatellite instability markers for sebaceous carcinoma: positive (retained) nuclear staining for MLH1, PMS2, MSH2 and MSH6
    • Pathologic staging: pTIs
    • IHC stains: androgen receptor - positive in tumor cells; adipophilin - positive in tumor cells; BerEP4: negative in tumor cells
Differential diagnosis
  • Basal cell carcinoma with sebaceous differentiation:
    • Small basaloid cells with peripheral palisading, surrounded by a fibromyxoid stroma, with focal differentiation toward mature benign appearing multivacuolated sebocytes
    • IHC: BerEP4+, EMA-, adipophilin is negative or granular pattern in basaloid cells, AR-
  • Clear cell squamous cell carcinoma:
    • Clear cells are not as multivacuolated as sebocytes
    • IHC: AR-, BerEP4-, adipophilin may be negative or granular pattern
  • Balloon cell melanoma:
    • Atypical melanocytes with clear cytoplasm, easily distinguished with melanocytic markers
    • IHC: SOX10+ and S100+
  • Metastatic clear cell carcinoma (renal):
    • Cells with more uniform clearing and prominent capillary vessels
    • Characteristically positive for CD10+, CAIX + and PAX8, adipophilin is negative or granular rather than vacuolated pattern
  • Clear cell sarcoma:
    • Nested to fascicular architecture with epithelioid to plump spindle cells with vesicular nuclei and macronucleoli separated by thin fibrous septa; scattered multinucleated giant cells and pigment
    • IHC: SOX10+ and S100+
Additional references
Board review style question #1

A 75 year old woman had an excisional biopsy for a rapidly growing mass in her upper eyelid. Histologic details are shown in the image above. Which of the following is true regarding this entity?

  1. Pagetoid spread is more common in periocular location
  2. Radiation therapy is the first line treatment
  3. Surgical excision is not helpful
  4. This tumor is S100 positive
Board review style answer #1
A. Pagetoid spread is more common in periocular location. The pagetoid pattern of sebaceous carcinomas is seen much more in the periocular area. This tumor is characteristically S100 negative. First line treatment is surgical excision. Radiation therapy is helpful in metastatic disease and in poor surgical candidates.

Comment Here

Reference: Sebaceous carcinoma
Board review style question #2
Which immunostain is negative in sebaceous carcinoma?

  1. Adipophilin
  2. Androgen receptor
  3. CEA
  4. EMA
Board review style answer #2
C. CEA. Sebaceous carcinoma stains positive for EMA, adipophilin, androgen receptor. It is usually negative for CEA.

Comment Here

Reference: Sebaceous carcinoma

Sebaceous hyperplasia
Definition / general
  • Most common sebaceous gland lesion
  • Usually elderly patients on nose or cheeks
Gross description
  • Umbilicated yellowish papules
Microscopic (histologic) description
  • Expansion of normal lobular sebaceous gland architecture without thickening of peripheral germinative layer of seboblasts
Microscopic (histologic) images

Contributed by Angel Fernandez-Flores, M.D., Ph.D.

Sebaceous hyperplasia



Images hosted on other servers:

Sebaceous hyperplasia

Differential diagnosis

Seborrheic keratosis
Definition / general
  • Common, benign keratinocyte proliferation of middle aged and elderly
Essential features
  • Benign
  • Clinical: waxy, brown slow growing papule
  • Histologic:
    • Proliferation of basaloid keratinocytes without atypia
    • Acanthosis and hyperkeratosis most often with horn pseudocysts
Terminology
  • Senile wart
  • Seborrheic wart
ICD coding
  • ICD-10: L82 - seborrheic keratosis
Epidemiology
Sites
Pathophysiology
  • Immature benign keratinocyte proliferation
  • Multiple somatic mutations (Oncotarget 2017;8:36639)
    • Most common: FGFR3, PIK3CA and HRAS
  • Amyloid precursor protein (APP) (Acta Derm Venereol 2018;98:594)
    • Expression increases in sun exposed areas and with age
    • May contribute to seborrheic keratosis formation
Etiology
  • Aging
  • Chronic UV light exposure
Clinical features
  • Single or multiple
  • Papules or plaques with a stuck on appearance (well demarcated edges)
  • Brown-black or gray in color
  • Waxy or greasy with cerebriform surface
  • Millimeters, up to a centimeter
    • Larger have been reported (rare)
  • Slow growing
  • Irritated / inflamed seborrheic keratosis (World J Nucl Med 2021;20:309)
    • Can be irregular and ulcerated (mimics carcinoma)
  • Dermatosis papulosa nigra (StatPearls: Dermatosis Papulosa Nigra [Accessed 3 May 2022])
    • Multiple seborrheic keratosis on the face (usually cheeks)
    • Present in adolescents
    • More common those of Asian and African descent
  • Leser-Trélat sign: (Cleve Clin J Med 2017;84:918)
    • Sudden appearance of multiple seborrheic keratoses, rapid increase in size, pruritic
    • Paraneoplastic phenomenon typically associated with gastrointestinal adenocarcinoma
Diagnosis
  • Clinical:
    • Dermoscopy: small keratin filled cysts, fissures, ridges, small vessels with perivascular halo (F1000Res 2019;8:1520)
  • Biopsy often performed on irritated lesions or those with rapid growth (Ann Dermatol 2016;28:152)
  • Histologic findings confirm diagnosis
Prognostic factors
  • Benign
  • Leser-Trélat sign may indicate underlying malignancy
Case reports
Treatment
Clinical images

Images hosted on other servers:
Seborrheic keratosis papules

Papules

Various images

Sharply circumscribed

Leser-Trélat sign

Leser-Trélat sign

Microscopic (histologic) description
  • General (shared) features:
    • Intraepidermal, well demarcated edges with a flat base
      • String sign: can draw a horizontal line along the base of the lesion
    • Basaloid keratinocyte proliferation without dysplasia
    • Hyperkeratotic with horn pseudocyst formation (intralesional cysts of loose keratin)
    • Multiple variants (no clinical or prognostic significance)
      • Often overlapping features
  • Acanthotic type:
  • Keratotic (papillomatous) type (Indian J Sex Transm Dis AIDS 2017;38:176):
    • Marked hyperorthokeratosis and papillomatosis
    • Can form a cutaneous horn
  • Reticulated (adenoid) type:
    • Thin, anastomosing strands of basaloid cells
    • Small horn cysts
    • May have increased pigment
  • Clonal type (Dermatol Pract Concept 2015;5:5, Pan Afr Med J 2019;34:54):
    • Pale basaloid keratinocytes in nests (Borst-Jadassohn phenomenon)
    • Horn pseudocysts may be absent
  • Irritated type (Arch Plast Surg 2017;44:570, Dermatol Online J 2019;25:13030):
    • Squamous metaplasia and whorled squamous eddies
    • Reactive squamous atypia
    • Scattered keratinocyte apoptosis and dyskeratosis
    • Spongiosis
    • Scale crust and parakeratosis in the stratum corneum
    • Lichenoid infiltrate in the superficial dermis (variable)
  • Pigmented:
    • Increased melanin pigmentation in the keratinocytes
    • Increased melanophages, mostly in the basal layer
  • Macular:
    • Minimal / mild acanthosis
    • Absent horn pseudocysts
    • Usually basal pigmentation increased
Microscopic (histologic) images

Contributed by Sara Shalin, M.D., Ph.D. and Caroline I.M. Underwood, M.D.
Acanthotic (regular) Acanthotic (regular)

Acanthotic (regular)

Hyperkeratosis

Hyperkeratosis

Papillomatosis without atypia

Papillomatosis without atypia

Reticulated pattern

Reticulated pattern


Adenoid with increased pigment

Adenoid with increased pigment

Clonal nests

Clonal nests

Pale keratinocytes

Pale keratinocytes

Clonal nests and horn pseudocyts

Clonal nests and horn pseudocyts


Irritated Irritated

Irritated

Increased pigmentation

Increased pigmentation

Minimal acanthosis

Minimal acanthosis

Negative stains
Videos

Seborrheic keratosis overview

Seborrheic keratosis and variants

Sample pathology report
  • Left chest, shave of skin:
    • Seborrheic keratosis
Differential diagnosis
  • Melanocytic nevus:
    • Nests of melanocytes in dermis or at dermal epidermal junction
    • Also present in younger patients
  • Solar lentigo:
    • Similar to reticulated seborrheic keratosis
    • Lentigo has pigmented, nonanastomotic, bulbous rete and no horn cysts
  • Malignant melanoma:
    • Malignant melanocytes with invasion into the dermis
  • Pigmented basal cell carcinoma:
    • Clefting, peripheral palisading, mucin, apoptosis and mitosis
  • Condyloma acuminatum:
    • Koilocytes
    • Dysplastic changes
    • Clinical findings (genital areas) and HPV positivity helpful in distinguishing
  • Verruca vulgaris:
    • Hypergranulosis, tiers of parakeratosis, dilated papillary blood vessels, intracorneal hemorrhage
    • Clinical features (present on hands and feet) and HPV positivity helpful in distinguishing
  • Actinic keratosis:
    • Atypia of basal keratinocytes
  • Squamous cell carcinoma:
    • Full thickness squamous atypia
    • Increased, often atypical mitoses
  • Melanoacanthoma:
    • Numerous dendritic melanocytes
    • Regarded by some as a variant of seborrheic keratosis
  • Clear cell acanthoma:
    • Clear keratinocytes (glycogenated)
    • No horn cysts
  • Achrochordon:
    • Usually in flexural areas
    • More polypoid clinically
    • Histologically normal skin lacking adnexal structures
  • Inverted follicular keratosis:
    • May have filiform growth
    • Endophytic growth
    • BCL2 upregulation in dendritic cells
Board review style question #1

Which feature helps distinguish the above lesion from squamous cell carcinoma?

  1. Acanathosis and papillomatosis
  2. Atypical mitotic figures
  3. Lack of atypia
  4. Molecular testing for FGFR3
Board review style answer #1
C. Lack of atypia. Seborrheic keratosis is a benign keratinocyte proliferation that lacks atypia and dysplasia. Acanthosis and papillomatosis (A) are features of seborrheic keratosis but can also be seen in squamous cell carcinoma. Atypical mitotic figures (B) are a feature of squamous cell carcinoma. Many seborrheic keratoses have mutations in FGFR3 (D) but it is not diagnostic or ubiquitous.

Comment Here

Reference: Seborrheic keratosis
Board review style question #2
Sudden eruption of seborrheic keratosis on the trunk should prompt which clinical response?

  1. Evaluation for underlying colonic carcinoma
  2. Full body skin exam for atypical nevi
  3. Immediate treatment with cryotherapy
  4. No further workup is necessary
Board review style answer #2
A. Evaluation for underlying colonic carcinoma. Sudden eruption of multiple seborrheic keratoses, the Leser-Trélat sign, is a paraneoplastic phenomenon. It is associated with underlying malignancy, most often colonic adenocarcinoma and should prompt a clinical workup to evaluate (D). The Leser-Trélat sign is not associated with increased risk for atypical nevi or melanoma (B). While seborrheic keratoses can be treated with cryotherapy (C), they are benign and treatment is not necessary.

Comment Here

Reference: Seborrheic keratosis

Squamous cell carcinoma
Definition / general
  • Cutaneous squamous cell carcinoma is a malignancy of epidermal keratinocytes that displays variable degrees of differentiation and cytological features
Essential features
  • Most patients have a favorable outcome after surgical resection
  • Only a subset of patients carry a higher risk of local recurrence, distant metastasis and mortality
  • Identifying and reporting the high risk features is important
Terminology
  • Cutaneous squamous cell carcinoma (cSCC)
  • Squamous cell carcinoma (SCC)
  • Squamous cell carcinoma in situ (SCCIS)
  • Bowen disease (BD)
ICD coding
  • ICD-O: 8070/3 - squamous cell carcinoma, NOS
Epidemiology
Sites
  • Most often in sun exposed areas
  • Scalp, ear, lip, nose, eyelid are high risk anatomic sites
Pathophysiology
  • Cutaneous squamous cell carcinoma appears to develop through a multistep process
  • UV radiation, mutations involving genes (such as TP53, CDKN2A, NOTCH1 and NOTCH2, EGFR and TERT) and molecular pathways (RAS / RAF / MEK / ERK and PI3K / AKT / mTOR) have been shown to play an important role in the pathogenesis (J Eur Acad Dermatol Venereol 2020;34:932)
Etiology
  • Ultraviolet light radiation and other forms of radiation
  • Chronic immunosuppressions
  • Actinic keratosis (precursor lesion), albinism (lack of pigmentation in skin), arsenic
  • Burn scars
  • Chronic ulcers
  • Chronic inflammation
  • Sinus tract
  • Human papillomavirus infection
  • Tars / oils
  • Xeroderma pigmentosa (J Am Acad Dermatol 2018;78:237)
Clinical features
  • Thin squamous cell carcinoma: erythematous scaly thin papule or plaque
  • Thicker tumors typically present as erythematous plaque, nodule, ulcer (Eur J Cancer 2015;51:1989)
Diagnosis
  • Characteristic gross features are suggestive of the diagnosis
  • Definitive diagnosis is made by shave, punch or excisional biopsies
Prognostic factors
Staging
  • Based on lesion size, depth of invasion, differentiation and perineural invasion
  • Helps to identify cutaneous squamous cell carcinomas with worse prognosis
  • 3 major staging methods (J Am Acad Dermatol 2019;80:106, JAMA Dermatol 2018;154:428)
    • AJCC, seventh edition of the American Joint Committee on Cancer (this staging system is not included in the eighth edition)
      • pT1: Tumor diameter < 2 cm, with < 2 high risk factors
      • pT2: Tumor diameter ≥ 2 cm or with ≥ 2 high risk factorsa
      • pT3: Tumor with invasion of maxilla, mandibular, orbit or temporal bone
      • pT4: Tumor with invasion of skeleton (axial or appendicular) or perineural invasion of skull base
      • aHigh risk factors: tumor thickness > 2 mm, Clark level IV / V, poor or undifferentiated, perineural invasion (PNI), location at ear or lip
    • AJCC, eighth edition of the American Joint Committee on Cancer for cutaneous squamous cell carcinoma of the head and neck
      • pT1: Tumor diameter ≤ 2 cm
      • pT2: Tumor diameter ≥ 2 cm and < 4 cm
      • pT3: Tumor with diameter ≥ 4 cm or with one of the high risk featuresb
      • pT4a: Tumor with gross cortical bone / marrow invasion of maxilla, mandibular orbit or temporal bone
      • pT4b: Tumor with skull base invasion or skull base foramen involvement
      • bHigh risk features: perineural invasion (of a nerve lying beneath the dermis or ≥ 0.1 mm in caliber or presenting with clinical or radiographic involvement of named nerves without skull base invasion or transgression), deep invasion (involvement beyond the subcutaneous fat or > 6 mm) and minor bone erosion
    • Brigham and Women's Hospital (BWH) classification
      • pT1: 0 high risk factorsc
      • pT2a: 1 high risk factor
      • pT2b: 2 - 3 high risk factors
      • pT3: ≥ 4 high risk factors or bone invasion
      • cHigh risk factors: tumor diameter ≥ 2 cm, poorly differentiated, perineural invasion ≥ 0.1 mm or tumor invasion beyond fat
Case reports
Treatment
Clinical images

Images hosted on other servers:

Scalp mass

Exophytic mass

Inoperable tumor

Nasal cavity mass

Gross description
Gross images

Images hosted on other servers:

Resected tumor and mandible

Microscopic (histologic) description
  • Carcinoma of keratinocytes that infiltrates the dermis
  • An associated precursor lesion (actinic keratosis / keratinocytic dysplasia / squamous cell carcinoma in situ) is often present
  • Spectrum of histologic features; all share downward growth below level of adjacent or overlying epidermis
  • Grading based on degree of differentiation and keratinization (J Am Acad Dermatol 2018;78:237)
    • Well differentiated: easily recognizable squamous epithelium, abundant keratinization, intercellular bridges apparent, minimal pleomorphism, mitotic figures basally located
    • Moderately differentiated: focal keratinization; features between well and poorly differentiated
    • Poorly differentiated: no / minimal keratinization, marked nuclear atypia, may be difficult to establish squamous differentiation
    • Undifferentiated: no keratinization, immunohistochemistry is usually necessary to confirm the diagnosis and to exclude melanoma or sarcoma
  • Histologic patterns (Hematol Oncol Clin North Am 2019;33:1)
    • Low risk histologic variants
      • Keratoacanthoma: well differentiated, crateriform appearance
      • Verrucous carcinoma: verruciform surface, blunt endophytic growth, minimal atypia
      • Clear cell squamous cell carcinoma: > 25% cells with cytoplasmic clearing (glycogen accumulation or hydropic degeneration)
    • High risk histologic variants
      • Acantholytic: squamous cell carcinoma with acantholysis, pseudoglandular (CEA negative) (J Cutan Pathol 2022;49:133, J Clin Pathol 2006;59:1206, Dermatol Surg 2011;37:353)
        • Acantholysis is the loss of cell to cell connections between keratinocytes, resulting in loss of intercellular cohesion
        • Acantholytic cutaneous squamous cell carcinomas have been classically considered as a high risk variant of CSCC; however, some recent studies show that acantholytic squamous cell carcinomas does not confer more aggressiveness
      • Invasive Bowen disease: invasive islands of squamous and basaloid cells with overlying classic Bowen disease
      • Spindle cell squamous cell carcinoma:
        • Uncommon variant in which keratinocytes infiltrate dermis as single cells with elongated nuclei, not as cohesive nests or islands and there are no / minimal signs of keratinization of conventional squamous cell carcinoma (Am J Dermatopathol 2008;30:228, J Skin Cancer 2011;2011:210813)
        • Commonly associated with a good prognosis, less often associated with a poor prognosis following exposure to ionizing radiation, trauma or burn
      • Desmoplastic squamous cell carcinoma: poorly differentiated, pleomorphic spindle cells with a dense stromal response
      • Adenosquamous carcinoma: mixed squamous and glandular differentiation (CEA positive)
    • Uncommon variants
      • Lymphoepitheliomatous
      • Pseudovascular squamous cell carcinoma
      • Squamous cell carcinoma with sarcomatoid differentiation
      • Squamous cell carcinoma with osteoclast-like cells
Microscopic (histologic) images

Contributed by Shaofeng Yan, M.D., Ph.D. and Semir Vranić, M.D., Ph.D.

Invasive SCC

Well differentiated SCC

Moderately differentiated SCC

Poorly differentiated SCC

Keratoacanthoma type SCC


Acantholytic SCC

Bowenoid SCC

SCC with sarcomatoid differentiation

SCC with sarcomatoid differentiation: CK5


Perineural invasion

Perineural invasion: S100 / CK5 double stain

Acantholytic type

Cytology images

Images hosted on other servers:

Papanicolaou and Romanowsky

Positive stains
Sample pathology report
  • Skin, left temple, shave biopsy:
    • Invasive squamous cell carcinoma, well differentiated, present at the peripheral and deep specimen edges
  • Skin, left temple, excision:
    • Invasive squamous cell carcinoma, poorly differentiated, present at the peripheral specimen margin (see synoptic report)
Differential diagnosis
  • Inflammatory dermatoses:
    • Hypertrophic lupus erythematosus, lichen planus:
      • Both can mimic due to marked squamous hyperplasia particularly if clinical information is not available
      • Absence of infiltrating squamous epithelium, presence of multiple lesions and features of lupus erythematosus (such as vacuolar interface lymphocytic infiltrate, necrotic keratinocytes, increase of dermal mucin, superficial and deep perivascular and periadnexal lymphocytic infiltrate) or lichen planus (such as wedge shaped hypergranulosis, band-like lymphocytic infiltrate with colloid bodies) are key differentiators
  • Miscellaneous keratoses:
    • Proliferative actinic keratosis:
      • Epithelial dysplasia variable from mild basal layer changes to carcinoma in situ and often associated with solar elastosis and parakeratosis
      • Budding of atypical epithelium into the papillary dermis
      • Distinction from early invasive squamous cell carcinoma is somewhat artificial, the presence of single or groups of atypical keratinocytes detached from the main lesion or associated with a stromal reaction supports invasive squamous cell carcinoma
    • Inverted follicular keratosis, clonal seborrheic keratosis:
      • Intradermal or inverted type of seborrheic keratosis is known as inverted follicular keratosis and is characterized by intradermal whorls of maturing squamous epithelium, so called squamous eddies
      • Sometimes, intraepithelial nesting gives rise to the intraepidermal epitheliomatous (Borst-Jadassohn) nodular appearance, surrounded by normal basaloid cells; this is also called clonal seborrheic keratosis
  • Other malignant tumors:
    • Basal cell carcinoma:
      • Frequently originates from the overlying and consists of small basaloid cells with peripheral palisading, darkly staining nuclei and minimal cytoplasm
      • Intercellular bridges are not as evident
      • Mitoses and apoptosis are commonly present
    • Adnexal carcinoma:
      • Eccrine porocarcinoma may show bowenoid features
      • Microcystic adnexal carcinoma may mimic keratinizing squamous cell carcinoma, especially in superficial biopsies in which ductal structures are not obvious
        • Identification of ductal differentiation by EMA or CEA is helpful
    • Malignant melanoma, sarcoma, lymphoma:
      • Adequate sampling to detect an epithelial origin or a junctional component is important
      • Cytokeratin, SOX10, CD45 or desmin will usually enable distinction from melanoma, lymphoma, sarcoma (or leiomyosarcoma)
      • p63 or p40 are usually negative in atypical fibroxanthoma or undifferentiated pleomorphic sarcoma
  • Pseudoepitheliomatous hyperplasia:
    • After trauma, surgery, infection; may be associated with granular cell tumor
    • Can be seen in association with chronic healing wounds, chronic irritation and infection
    • Benign and reactive epidermal acanthosis showing irregular and often endophytic growth pattern
    • Prominent acanthotic downgrowths and keratinocytes with bland nuclear features containing abundant cytoplasm
Additional references
Board review style question #1
Which subtype of cutaneous squamous cell carcinoma is shown in the image below?



  1. Acantholytic type
  2. Adenosquamous type
  3. Desmoplastic type
  4. Keratoacanthoma type
Board review style answer #1
A. Acantholytic type

Comment Here

Reference: Squamous cell carcinoma
Board review style question #2
Which of the following is a good prognostic factor for cutaneous squamous cell carcinoma?

  1. Desmoplastic type squamous cell carcinoma
  2. Keratoacanthoma type squamous cell carcinoma
  3. Tumor invades beyond subcutaneous fat
  4. Tumor size > 2 cm
Board review style answer #2
B. Keratoacanthoma type squamous cell carcinoma

Comment Here

Reference: Squamous cell carcinoma

Squamous cell carcinoma in situ / Bowen disease
Definition / general
  • Usually on skin NOT exposed to sunlight, such as trunk
  • Called erythroplasia (of Queyrat) if at glans penis, vulva, oral cavity
  • Actinic keratosis with only a single layer of atypical keratinocytes may be invasive but Bowen disease seldom is
  • Mucosal variants in glans penis (penile intraepithelial neoplasia or "PIN") and vulva (vulvar intraepithelial neoplasia or "VIN") are associated with more aggressive behavior
Case reports
Gross description
  • Slightly raised, large scaly erythematous plaque with irregular border
  • Usually single patch or verrucous growth
Microscopic (histologic) description
  • By definition requires full thickness keratinocyte atypia, although may be surrounded by normal keratinocytes
  • Architectural and cellular atypia, apoptotic cells, individual cell dyskeratosis
  • Markedly altered maturation but usually still some surface keratinization and intercellular bridges present
  • Marked nuclear atypia, including nuclear hyperchromasia and multinucleation
  • Numerous mitotic figures, atypical mitotic figures
  • Also cytoplasmic vacuoles; rarely pagetoid cells or ground glass cytoplasm
  • May extend into eccrine sweat glands (not considered invasive disease)
  • Variable melanin, variable lymphocytic infiltrate
  • May have hemangiomatous vascular proliferation, amyloid globules, adnexal differentiation
Microscopic (histologic) images

Contributed by Amy Lynn, M.D., Andrey Bychkov, M.D., Ph.D. and Jijgee Munkhdelger, M.D., Ph.D.

Various images

Full thickness atypia

Hyperkeratosis and acanthosis

Pleomorphism, multinucleation

High power



Images hosted on other servers:

Various images

Differential diagnosis

Staging-Merkel cell carcinoma
Definition / general
  • All primary cutaneous neuroendocrine carcinomas are covered by this staging system
  • Metastatic extracutaneous neuroendocrine carcinomas are not covered
Essential features
  • AJCC 7th Edition staging was sunset on December 31, 2017; as of January 1, 2018, use of the 8th Edition is mandatory
ICD coding
  • ICD-10: C4A - Merkel cell carcinoma
  • ICD-10: C7B.1 - Metastatic Merkel cell carcinoma or nodal presentation without known primary
Primary tumor (pT)
  • pTX: primary tumor cannot be assessed (e.g. curetted)
  • pT0: no evidence of primary tumor
  • pTis: in situ primary tumor
  • pT1: maximum clinical tumor diameter ≤ 2 cm
  • pT2: maximum clinical tumor diameter > 2 cm but ≤ 5 cm
  • pT3: maximum clinical tumor diameter > 5 cm
  • pT4: primary tumor invades fascia, muscle, cartilage or bone

Notes:
  • Tumor diameter: measured as the largest diameter of the primary tumor; this is a clinical measurement
  • Gross or microscopic measurement should only be used when the clinical measurement is unavailable since tumor shrinkage during formalin fixation will result in underestimation of diameter
Regional lymph nodes (pN)
  • pNX: cannot be assessed (staging procedure not performed or previously removed)
  • pN0: no regional metastasis detected on pathological evaluation
  • pN1a(sn): microscopic lymph node metastasis detected by sentinel node biopsy, clinically occult
  • pN1a: microscopic regional lymph node metastasis following dissection, clinically occult
  • pN1b: microscopic confirmation of clinically detected regional lymph node metastasis
  • pN2: in transit metastasis without lymph node metastasis
  • pN3: in transit metastasis with lymph node metastases

Notes:
  • Regional lymph nodes / sentinel lymph nodes: sentinel node(s) receive direct lymphatic drainage from the primary tumor site
  • Clinically occult: nodal metastasis detectable only by microscopic evaluation
  • Clinically detected: lymph node enlarged or abnormal by physical or radiologic examination
  • Isolated tumor cells: counted as metastases whether identified on H&E or by IHC
  • In transit metastasis: intralymphatic tumor deposits discontinuous from the primary lesion and located either between the primary lesion and the draining regional lymph nodes or distal to the primary lesion
Distant metastasis (pM)
  • pM0: no evidence of distant metastasis by clinical or radiological examination
  • pM1a: microscopic confirmation of metastases to skin, distant subcutaneous tissue or distant lymph nodes
  • pM1b: microscopic confirmation of metastases to lung
  • pM1c: microscopic confirmation of metastases to all other distant sites
AJCC pathological prognostic stage groups (pTNM)
    Stage group 0: Tis N0 M0
    Stage group I: T1 N0 M0
    Stage group IIA: T2 - T3 N0 M0
    Stage group IIB: T4 N0 M0
    Stage group IIIA: T1 - T4 N1a(sn) or N1a M0
    T0 N1b M0
    Stage group IIIB: T1 - T4 N1b - N3 M0
    Stage group IV: T0 - 4 N0 - N3 M1

Notes:
  • Unknown primary tumor: patients presenting with metastatic Merkel cell carcinoma to a lymph node in the absence of a primary tumor have a significantly more favorable prognosis than patients with a primary tumor and a clinically detected nodal metastasis
  • Survival rates are similar to those of patients with a primary tumor and occult nodal metastases detected by sentinel lymph node biopsy (J Am Acad Dermatol 2013;68:433, Am J Surg 2013;206:752, J Am Acad Dermatol 2012;67:395)
Registry data collection variables
  • Largest tumor diameter (clinical measurement in millimeters)
  • Regional node status (clinically detected, pathologically detected or neither)
  • Unknown primary status (yes or no)
  • Tumor thickness (reported in millimeters)
  • Excision margin status (tumor base transected or not transected)
  • Profound immunosuppression (no conditions, HIV / AIDS, solid organ transplant recipient, CLL / SLL, other non-Hodgkin lymphoma, multiple conditions or condition NOS)
  • Lymphovascular invasion (present or absent)
  • Merkel cell polyomavirus positive staining by IHC (yes, no or not applicable)
  • p63 positive staining by IHC (yes or no)
  • Tumor infiltrating lymphocytes in primary tumor (not present, present nonbrisk or brisk)
  • Growth pattern of primary tumor (circumscribed, nodular or infiltrative)
  • Extranodal extension in regional lymph nodes (yes or no)
  • Tumor nest size in regional lymph nodes (greatest dimension of largest aggregate in millimeters)
  • Isolated tumor cells in regional lymph node (yes or no)
  • Eyelid tumor (no, yes upper lid, yes lower lid or yes involving both lids)
  • Eyelid tumor involving eyelid margin, defined as the juncture of eyelid skin and tarsal plate at the lash line (no, yes non full thickness or yes full thickness)

Notes:
  • Tumor thickness: measure from the surface of the epidermal granular layer to the point of maximum tumor thickness at a right angle to adjacent epidermis
  • Tumor infiltrating lymphocytes: considered brisk when the inflammation diffusely infiltrates the tumor or is present around the entire base of the tumor
Histologic grade (G)
  • Not used in Merkel cell carcinoma staging
Histopathologic type
  • Not used in Merkel cell carcinoma staging
Board review style question #1
    Which of the following Merkel cell carcinoma is associated with the worst prognosis?

  1. Primary tumor involves dermis and there are clinically detected regional lymph node metastases
  2. Primary tumor involves fat and there are no regional lymph node metastases
  3. Primary tumor involves muscle and there are no regional lymph node metastases
  4. Metastatic Merkel cell carcinoma is present in a lymph node but no primary tumor is identified
Board review style answer #1
A. The presence of nodal metastases upstages the carcinoma regardless of primary tumor attributes. Patients with clinically detected nodal metastases in the absence of an identifiable primary Merkel cell carcinoma (Stage IIIA) fare significantly better than patients with a known primary tumor and clinically detected regional node metastases (Stage IIIB).

Comment Here

Reference: Pathologic TNM staging of Merkel cell carcinoma (AJCC 8th edition)
Board review style question #2
In the AJCC 8th edition, the most important parameters determining the pT category for Merkel cell carcinoma is which of the following?

  1. Lymphovascular invasion and p16 expression
  2. Maximum clinical tumor diameter and extent of anatomic invasion
  3. Tumor infiltrating lymphocytes and necrosis
  4. Tumor thickness and mitotic rate
  5. Ulceration and regression
Board review style answer #2
B. Maximum clinical tumor diameter and extent of anatomic invasion

pT categories for primary cutaneous Merkel cell carcinoma (AJCC 8th edition) are as follows:
pTis: in situ primary tumor
pT1: maximum clinical tumor diameter ≤ 2 cm
pT2: maximum clinical tumor diameter > 2 cm but ≤ 5 cm
pT3: maximum clinical tumor diameter > 5 cm
pT4: primary tumor invades fascia, muscle, cartilage or bone

Comment Here

Reference: Pathologic TNM staging of Merkel cell carcinoma (AJCC 8th edition)

Staging-nonmelanocytic carcinoma
Definition / general
  • Staging of melanomas is described in Skin-Melanocytic tumors chapter
  • Staging of related eye tumors is described in the Eye chapter for Conjunctiva, Eyelid, Lacrimal gland
  • Includes spindle cell variant of squamous cell carcinoma and adnexal carcinomas
  • Applies to clinical and pathologic staging

TNM descriptors
  • Required only if applicable
    • m (multiple)
    • r (recurrent)
    • y (posttreatment)

Primary tumor (T)
  • TX: Primary tumor cannot be assessed
  • T0: No evidence of primary tumor
  • Tis: Carcinoma in situ
  • T1: Tumor 2 cm or less in greatest dimension with less than 2 high risk features
  • T2: Tumor more than 2 cm in greatest dimension with or without one additional high risk features, or any size with two or more high risk features.
  • T3: Tumor with invasion of maxilla, mandible, orbit, or temporal bone.
  • T4: Tumor invades skeleton (axial or appendicular) or perineural invasion of skull base

Regional lymph nodes (N)
  • NX: Regional lymph nodes cannot be assessed
  • N0: No regional lymph node metastases
  • N1: Metastasis in a single ipsilateral lymph node, 3 cm or less in greatest dimension
  • N2: Metastasis in a single ipsilateral lymph node, more than 3 cm but not more than 6 cm in greatest dimension; or in multiple ipsilateral lymph nodes, none more than 6 cm in greatest dimension; or in bilateral or contralateral lymph nodes, none more than 6 cm in greatest dimension
  • N2a: Metastasis in a single ipsilateral lymph node, more than 3 cm but not more than 6 cm in greatest dimension
  • N2b: Metastasis in multiple ipsilateral lymph nodes, none more than 6 cm in greatest dimension
  • N2c: Metastasis in bilateral or contralateral lymph nodes, none more than 6 cm in greatest dimension
  • N3: Metastasis in a lymph node, more than 6 cm in greatest dimension

Distant metastasis (M)
  • Not applicable
  • M0: No distant metastasis
  • M1: Distant metastasis, specifiy sites if known

Stage grouping
  • Stage 0: Tis N0 M0
  • Stage I: T1 N0 M0
  • Stage II: T2 N0 M0
  • Stage III: T3 N0 M0 or T1-3 N1 M0
  • Stage IV: T1-3 N2 M0 or any T N3 M0 or T4 any N M0 or any T any N M1

Notes
  • Patients with primary clinical squamous cell carcinoma or other cutaneous carcinomas with no evidence (clinical, radiologic or pathologic) of regional or distant metastases are divided into two stages:
    • Stage I for tumors measuring 2 cm or less in size
    • Stage II for those that are greater than 2 cm in size
  • If there is clinical concern for extension of tumor into bone and radiologic evaluation is negative, these data may be included to support the Stage I vs. II designation
  • Tumors that are 2 cm or less in size can be upstaged to Stage II if they contain two or more high-risk features
  • Stage III patients are those with:
    • Clinical, histologic or radiologic evidence of one solitary node measuring 3 cm or less in size or
    • Tumor extension into bone: maxilla, mandible, orbit or temporal bone
  • Stage IV patients are those with:
    • Tumor with direct or perineural invasion of skull base or axial skeleton
    • 2 or more involved lymph nodes or
    • Single or multiple involved lymph nodes measuring > 3 cm in size or
    • Distant metastasis
Additional references

Steatocystoma
Definition / general
  • Benign cysts with squamous epithelial lining and associated sebaceous glands
  • Can present as single or multiple flesh colored papules
Essential features
  • Benign, sebaceous gland associated cysts, with a characteristic hyaline cuticle and jagged squamous epithelial lining
  • Can occur as sporadic lesions or in an autosomal dominant inheritance pattern
  • Keratin 17 (KRT17) is the most common mutation
Terminology
  • Steatocystoma simplex (single lesion clinically)
  • Steatocystoma multiplex (multiple lesions clinically)
ICD coding
  • ICD-10: L72.2 - steatocystoma multiplex
Epidemiology
  • No gender predominance
Sites
  • Face and neck, chest, arms, axillae
Etiology
  • Sporadic (simplex and multiplex) (J Dermatol 2002;29:152)
  • Autosomal dominant mutation in keratin 17 (multiplex)
Clinical features
Diagnosis
  • Generally a clinical diagnosis aided by histopathology
  • May be incidentally found on imaging
Radiology description
  • Not required for diagnosis
  • Well circumscribed, subepidermal fat signals, generally found incidentally on ultrasound, mammogram or noncontrast magnetic resonance imaging (MRI) (Breast J 2021;27:389, Cureus 2022;14:e27756)
Prognostic factors
Case reports
Clinical images

Images hosted on other servers:
Skin colored papules

Skin colored papules

Single yellow hued papule

Single yellow hued papule

Multiple papules on cheek

Multiple papules on cheek

Microscopic (histologic) description
Microscopic (histologic) images

Contributed by Bethany R. Rohr, M.D. and Claire Mazahery, M.D., Ph.D.
sebaceous glands in lining

Sebaceous glands in lining

sawtooth hyaline cuticle

Sawtooth hyaline cuticle

characteristic lining, debris, gland

Characteristic lining, debris, gland

sebaceous glands in wall

Sebaceous glands in wall

multiple sebaceous glands

Multiple sebaceous glands

sebaceous glands abutting lumen

Sebaceous glands abutting lumen

Virtual slides

Images hosted on other servers:
Skin, steatocystoma

Skin, steatocystoma

Positive stains
Videos

Steatocystoma: 5 minute pathology pearls by Dr. Jerad Gardner

Sample pathology report
  • Skin, left proximal forearm, shave biopsy:
    • Steatocystoma (see comment)
    • Comment: In the dermis, there is a cyst with a stratified squamous epithelial lining lacking a granular layer. There is a sawtoothed pattern cuticle and scattered sebaceous glands in the cyst lining.
Differential diagnosis
Board review style question #1

A 30 year old woman presented with a flesh colored papule on the arm. A biopsy was performed. What is the predominant inheritance pattern of the familial form of this entity?

  1. Autosomal dominant mutations in keratin 17
  2. Autosomal dominant mutations in keratin 18
  3. Autosomal recessive mutations in keratin 17
  4. Sporadic lesions only
Board review style answer #1
A. Autosomal dominant mutations in keratin 17. The solitary lesion in the question above is steatocystoma simplex. Steatocystoma multiplex can be seen as a sporadic or an autosomal dominant disease presenting with multiple steatocystomas. The heritable form is thought to be due to mutations in keratin 17. Answer D is incorrect because although the patient in the question stem has a single lesion (steatocystoma simplex, which is sporadic), steatocystoma multiplex has identical lesions on histology. Answer C is incorrect because steatocystoma multiplex is an autosomal dominant (not recessive) mutation in keratin 17. Answer B is incorrect because the mutated gene is keratin 17, not keratin 18.

Comment Here

Reference: Steatocystoma
Board review style question #2
A 40 year old man presents with a longstanding history of multiple asymptomatic, firm, flesh colored papules across the chest and upper back. He notes several family members with similar skin lesions. A close physical examination revealed thickened nails that he says have been a source of embarrassment since childhood. What adnexal structure is associated with his skin lesions?

  1. Apocrine glands
  2. Eccrine glands
  3. Follicular structures
  4. Sebaceous glands
Board review style answer #2
D. Sebaceous glands. The patient's nail changes present since early life are suggestive of pachyonychia congenita, which can co-occur with familial steatocystoma multiplex (keratin 17 mutations present in both diseases), accounting for the skin findings in this patient and his family members. The cysts of steatocystoma multiplex are associated with sebaceous glands. Answers A and B are incorrect because these may be seen in hidrocystoma. Answer C is incorrect because these are found in follicular based cysts (such as pilar cyst, vellus cyst or epidermal inclusion cyst).

Comment Here

Reference: Steatocystoma

Subcutaneous panniculitis-like

Subungual exostosis

Superficial acral fibromyxoma

Superficial angiomyxoma
Microscopic (histologic) images

Contributed by Mark R. Wick, M.D.

Supernumerary digit
Definition / general
  • Acral neuroma, also called rudimentary polydactyly
  • On radial side of fifth digit
Microscopic (histologic) description
  • Haphazard nerves with displaced Meissner bodies
Microscopic (histologic) images

Contributed by Andrey Bychkov, M.D., Ph.D.

Nerve bundles


Sweat gland carcinoma
Definition / general
  • Uncommon
  • Usually adults
  • Difficult to diagnose
  • May be life threatening

Low grade:
  • Microcystic carcinoma, adenoid cystic carcinoma, mucinous carcinoma, extramammary Paget’s disease, mucoepidermoid carcinoma

Intermediate grade:
  • Ductal adenocarcinoma, aggressive digital papillary adenocarcinoma, acrospirocarcinoma

High grade:
  • Porocarcinoma, clear cell acrospirocarcinoma

Unknown grade:
  • Signet ring cell carcinoma, papillary syringadenocarcinoma
  • Also includes adenoid cystic carcinoma, apocrine carcinoma, malignant acrospiroma, malignant chondroid syringoma (malignant mixed tumor), malignant dermal cylindroma, malignant eccrine poroma (most common), malignant myoepithelioma, malignant syringoma (syringoid eccrine carcinoma), mucinous syringometaplasia
  • Benign sweat gland tumors can also undergo malignant transformation to high grade carcinoma
Microscopic (histologic) description
  • May resemble breast carcinoma, renal cell carcinoma, basal cell carcinoma
Negative stains
Differential diagnosis

Synovial cysts

Syringocystadenoma papilliferum
Definition / general
Essential features
  • Benign adnexal tumor, most commonly occurring in the head and neck in early childhood (J Dermatol 2004;31:939)
  • Most are solitary papules but variable clinical presentation
  • Macroscopic: pink, hairless plaque or nodule
  • Microscopic: cystic invaginations of the infundibular epithelium extending into the dermis with a double cell layer of inner columnar and outer cuboidal cells
Terminology
Epidemiology
Sites
Pathophysiology
Etiology
  • Often observed in association with other benign adnexal neoplasms, such as nevus sebaceus (8 - 19% of patients), apocrine nevus, tubular apocrine adenoma, apocrine hidrocystoma, apocrine cystadenoma and clear cell syringoma (Pathologica 2006;98:178)
Clinical features
Diagnosis
  • Skin biopsy
Case reports
Treatment
Clinical images

Images hosted on other servers:

Posterior cranium, exophytic mass

Rose colored papule, left flank

Erythematous plaque, central crustation

Papillomatous exophytic, temporal scalp

Gross description
Microscopic (histologic) description
Microscopic (histologic) images

Contributed by Kiran Motaparthi, M.D.

Glandular proliferation

Cystic proliferation

Glands with epidermal connection

Glands with double layer


Glands forming papillae

Cystic and papillary appearance glands

Papillary architecture

Double layer epithelium

Positive stains
Molecular / cytogenetics description
Sample pathology report
  • Scalp, biopsy:
    • Syringocystadenoma papilliferum (see comment)
    • Comment: Several cystic invaginations arise from a papillomatous epidermis. These invaginations demonstrate papillae lined by 2 rows of cuboidal to columnar epithelial cells, with oval nuclei and a pale eosinophilic cytoplasm. The deep dermis contains tubular glands with apocrine decapitation secretion. The stroma contains a dense mononuclear infiltrate, which is comprised predominantly of plasma cells.
Differential diagnosis
Board review style question #1

A 38 year old man presents with a verrucous nodule on the scalp, present for the past 12 years. Representative histopathology is shown in the above image. Which of the following statements is correct?

  1. More common in adulthood
  2. Represents a malignant adnexal tumor
  3. Subset of tumors have loss of heterozygosity for PTCH or p16
  4. Usually does not communicate with surface epithelium
Board review style answer #1
C. Subset of tumors have loss of heterozygosity for PTCH or p16

Comment Here

Reference: Syringocystadenoma papilliferum
Board review style question #2

An 8 year old girl presents with a slowly growing, 3 cm, erythematous nodule with smooth surface on the left labium majus, present since birth. A biopsy is performed (see above image). Which of the following is most associated with this tumor?

  1. Basal cell carcinoma
  2. Merkel cell carcinoma
  3. Nevus sebaceus
  4. Squamous cell carcinoma
Board review style answer #2
C. Nevus sebaceus

Comment Here

Reference: Syringocystadenoma papilliferum

Syringoma
Definition / general
  • Benign adnexal neoplasm of sweat gland (eccrine) origin
  • Most common clinical presentation on lower eyelids of women
  • Appears to derive from sweat duct ridge
  • Malignant counterpart can be termed syringomatous carcinoma / sweat gland carcinoma
Essential features
  • Often multiple 1 - 4 mm, firm papules
  • Well circumscribed dermal lesion with comma shaped ductules lined by basaloid cells and with sclerotic stroma
ICD coding
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
Epidemiology
Sites
Pathophysiology
Etiology
Clinical features
Diagnosis
  • On face, in particular, can be clinically suspected
  • Biopsy required for confirmation
Prognostic factors
  • Benign, nonprogressive lesions but can be cosmetically bothersome if multiple
  • Recurrence, scarring and dyspigmentation after treatment
Case reports
Treatment
Clinical images

Images hosted on other servers:

Disseminated brownish papules

Eruptive syringoma:
multiple flat papules
on thighs

Eyelid syringoma

Gross description
  • 1 - 4 mm, firm, skin colored papule with or without eruption
Gross images
Microscopic (histologic) description
  • Well circumscribed proliferation with 2 components:
    • Epithelial cells forming ductules, nests, cysts and cords
      • Cells are basaloid, cuboidal and double layered (in ductules) with an eosinophilic cuticle
      • Ducts are often described as comma or tadpole shaped or in a paisley pattern (Hong Kong Med J 2018;24:200)
      • May have cell clearing glycogen (clear cell syringoma), more common in diabetics (J Am Acad Dermatol 1987;16:310)
      • Some small cysts may have squamous lining
      • Variable luminal proteinaceous debris / keratin debris
    • Stromal fibrosis / sclerosis
  • Usually in superficial reticular dermis, rarely deep dermal extension
  • No cytologic atypia
  • Very rare mitoses
  • Reference: Calonje: McKee's Pathology of the Skin, 5th Edition, 2019
Microscopic (histologic) images

Contributed by Aadil Ahmed, M.D., Sara C. Shalin, M.D., Ph.D. and Nicole Riddle, M.D.

Well circumscribed syringoma

Comma shaped ductules and nests

Basaloid proliferation

Paisley or tadpole shaped pattern

Clear cell change


Clear cell syringoma

Conventional and clear cell components

Incidental syringoma

Missing Image

Superficial reticular dermis

Missing Image

Basaloidesque cells

Cytology description
Positive stains
Electron microscopy description
  • Eccrine origin
Molecular / cytogenetics description
Sample pathology report
  • Skin, eyelid, biopsy:
    • Syringoma
    • Microscopic description: Histologic sections show skin with an unremarkable epidermis. Within the superficial dermis, a circumscribed proliferation of multiple ductular structures lined by 2 layers of epithelial cells is present coursing within densely fibrotic stroma. There is minimal cytologic atypia and mitoses are not abundant.
Differential diagnosis
Board review style question #1

Which of these characteristics is typical of this well circumscribed, dermal lesion?

  1. Associated with Turner syndrome
  2. May recur after excision
  3. Often multiple 1 - 4 mm, firm, flesh colored papules
  4. Painful lesions
  5. Strongly SOX10 immunoreactive
Board review style answer #1
C. Often multiple 1 - 4 mm, firm, flesh colored papules. The image shows a syringoma. These lesions are often multiple 1 - 4 mm, firm, flesh colored papules. They are typically asymptomatic but may present with pruritus. Various cell components are immunoreactive for CEA, EMA and CK5 but not for SOX10. They have benign behavior and excision is adequate. They are associated with Ehlers-Danlos, Marfan and Down syndromes.

Comment Here

Reference: Syringoma
Board review style question #2
Clear cell change seen in syringomas is due to

  1. Clearing artifact
  2. Globulin
  3. Glycogen
  4. Lipids
  5. Mucin
Board review style answer #2
C. Glycogen. The ductal cells in syringoma can infrequently exhibit clear cell change in focal areas that is due to accumulation of glycogen. Rarely, all cells are glycogen rich, termed clear cell variant and commonly associated with diabetes mellitus.

Comment Here

Reference: Syringoma

Systemic EBV+ of childhood

Telangiectases (pending)
[Pending]

Trichilemmal (pilar) type
Definition / general
  • Trichilemmal cyst, also known as a pilar cyst, is a keratin filled cyst that originates from the outer hair root sheath that is commonly located on the scalp
Essential features
  • 90% present on scalp
  • Circumscribed simple cyst with stratified squamous epithelium lining, which lacks a granular cell layer and contains dense eosinophilic keratin
  • Proliferating trichilemmal cyst and other neoplasms, such as Merkel cell carcinoma, can develop from or within a benign trichilemmal cyst
Terminology
  • Also called pilar cyst, isthmus catagen cyst or a wen
ICD coding
Epidemiology
Sites
  • 90% on scalp
  • Scrotum
  • Rarely on pulp of finger
Pathophysiology
  • Origin unknown but may arise from external root sheath as a genetically determined structural aberration
  • Mostly autosomal dominant inheritance (J Invest Dermatol 2019;139:2075)
  • Hair follicle's outer root sheath is recapitulated at the level of the follicular isthmus in the cyst wall
Clinical features
  • Most commonly found on the scalp (90%) and scrotum
  • When present as multiple lesions, autosomal dominance is common
  • Asymptomatic, firm, mobile, dermal or subcutaneous nodules measuring 0.5 to 5 cm in diameter
  • No central punctum is present
  • Typically, encapsulated cyst and uncomplicated lesions are easily shelled out at surgery
  • Acute inflammation is usually nonbacterial, may be associated with cyst rupture
  • Cholesterol clefts in up to 90%
  • Calcification in 25%, independent of patient age or size of cyst (J Ultrasound Med 2019;38:91)
Diagnosis
  • Clinical impression or excision
Prognostic factors
  • Benign but may be locally aggressive
  • Rarely malignant but may result in metastasis
  • Rarely other neoplasms, such as Merkel cell carcinoma, colonize or arise in trichilemmal cyst (An Bras Dermatol 2019;94:452)
  • Proliferating trichilemmal cysts can develop from a benign trichilemmal cyst; growth may be provoked by an unknown trigger, such as trauma, irritation or inflammation (Arch Craniofac Surg 2017;18:50)
Case reports
Treatment
  • No treatment necessary for asymptomatic lesions
  • Incision and drainage under local anesthesia
  • Enucleation of the cyst
  • Incision followed by expression of contents and removal of cyst wall
  • Surgical excision if clinically indicated
  • Reference: Int J Trichology 2013;5:115
Clinical images

Contributed by David Rosenfeld, M.D. and Ada Agidi, M.D.
Pilar cyst

Pilar cyst

Gross description
Microscopic (histologic) description
  • Well circumscribed subcutaneous or dermal simple cyst, lined by stratified squamous epithelium that has a palisaded outer layer and contains dense laminated eosinophilic keratin
  • Granular layer is absent
  • Calcification in up to 25%
  • Granulomatous response due to rupture
  • Sebaceous or apocrine glands may be seen (Requena: Cutaneous Adnexal Neoplasms, 1st Edition, 2017)
  • Hidrocystoma-like lining
Microscopic (histologic) images

Contributed by Aaron Muhlbauer, M.D. and Jodi Speiser, M.D.
Pilar cyst Pilar cyst

Pilar cyst

Virtual slides

Images hosted on other servers:

Pilar cyst

Molecular / cytogenetics description
Sample pathology report
  • Scalp, biopsy:
    • Trichilemmal (pilar) cyst (see comment)
    • Comment: The sections show a dermal cyst lined by stratified squamous epithelium with a palisaded outer layer, lack of granular layer and containing dense laminated eosinophilic keratin.
Differential diagnosis
  • Infundibular cyst:
    • Most common sites are face, neck and trunk
    • Central punctum is present
    • Origin is epithelium of hair follicle infundibulum
    • Cyst wall is delicate and prone to rupture
    • On histology, the granular cell layer is present
    • Contains laminated keratin unlike pilar cyst with dense, eosinophilic keratin material in the cyst lumen and no granular layer
  • Proliferating pilar cyst:
    • Can grow as large as 25 cm, may cause pressure necrosis (destruction) on underlying tissues, ulceration and foul smelling discharge
    • Well defined lobular proliferation of squamous cystic islands centered in the dermis
    • The lining epithelium is a stratified squamous epithelium exhibiting trichilemmal keratinization
    • Well circumscribed with foci of necrosis and dyskeratosis
    • Increased typical mitotic figures in basilar epithelium along with mild cytologic atypia
    • Squamous eddies are common
  • Hidrocystoma:
    • Uni or multi locular cyst in dermis
    • Consist of two or more layers of columnar-cuboidal epithelium with an outer layer of myoepithelial cells
  • Malignant pilar tumor:
    • Infiltrative border
    • Marked cytologic atypia with numerous atypical mitotic figures
    • Perineural or vascular invasion
    • Geographic necrosis
Additional references
Board review style question #1

A 68 year old man presents with a dermal nodule on his scalp. Which of the following statements is correct?

  1. Histology shows marked cytologic atypia with numerous atypical mitotic figures
  2. Histology shows simple cyst with squamous epithelium lining, lack of a granular cell layer and containing dense keratin
  3. Histology shows thin walled clear cystic spaces in dermis
  4. Histology shows well defined lobular proliferation of squamous cystic islands centered in the dermis
Board review style answer #1
B. Histology shows simple cyst with squamous epithelium lining, lack of a granular cell layer and containing dense keratin

Comment Here

Reference: Trichilemmal (pilar) cyst
Board review style question #2
A 50 year old woman presents with an encapsulated cyst on her scalp. The gross findings include a dermal cyst with a thick cyst wall filled with solid, homogenous material. Which of the following is the most likely neoplasm to arise within this cyst?

  1. Basal cell carcinoma
  2. Malignant proliferating pilar cyst
  3. Merkel cell carcinoma
  4. Proliferating pilar cyst
  5. Squamous cell carcinoma
Board review style answer #2
D. Proliferating pilar cyst

Comment Here

Reference: Trichilemmal (pilar) cyst

Trichilemmoma
Essential features
Terminology
  • Also called tricholemmoma
ICD coding
  • ICD-10: D23 - other benign neoplasms of skin
Sites
Pathophysiology
Etiology
Clinical features
  • Clinically nondescript wart-like or smooth dome shaped appearance
Diagnosis
  • Diagnosis is rarely made by clinical examination, more often made by pathologist via tissue examination revealing characteristic histological morphology
Case reports
Treatment
  • Not required
  • Curettage or shave excision
  • Excision
  • Laser
Clinical images

Images hosted on other servers:

Trichilemmoma

Central ulceration

Microscopic (histologic) description
  • Squamoproliferative lesion in continuity with the epidermis
  • Hyperkeratosis and stromal clefting are often associated
  • Pale eosinophilic or clear cells
  • Rounded lobular profile
  • Peripheral palisade of cuboidal or columnar cells
  • Clear cells (characteristic but uncommon)
  • Reverse polarity of peripheral cells (characteristic but uncommon)
  • Distinct basement membrane resembling the hair outer root sheath zone below the level of the follicular isthmus (Arch Dermatol 1962;86:430, Am J Dermatopathol 2018;40:561, Current Diagnostic Pathology 2007;13:273)
  • Basaloid cell predominance may be seen mimicking basal cell carcinoma
  • Central desmoplasia, squamous morules and mucin pools may be seen (uncommon)
Microscopic (histologic) images

Contributed by Nicholas Turnbull, M.B.Ch.B. and Richard A. Carr, M.B.Ch.B.

Bulbous basaloid tumor

Desmoplasia

Area composed of clear cells


Squamous morules and mucinous pools

BerEP4

EMA

CD34

Positive stains
Sample pathology report
  • Skin, right side of nose, punch biopsy:
    • Trichilemmoma (see comment)
    • Comment: This is a benign tumor. Multiple trichilemmomas may be associated with Cowden syndrome. Clinical correlation advised or specialist dermatology assessment should be considered if clinically indicated.
    • Microscopic description: A folliculocentric tumor with bulbous profile and stromal clefting is seen in the dermis with connection to the epidermis. The tumor is composed of basaloid cells and cells with clear cytoplasm. Peripheral palisading and a thickened basement membrane is noted. The overlying epidermis demonstrates hyperkeratosis. No dysplasia is seen.
Differential diagnosis
  • Inverted follicular keratosis:
    • Characterized by small basaloid cells and tight squamous eddies
  • Viral wart (verruca):
    • May be indistinguishable but typically demonstrates peripheral in turning of the rete ridges, marked hyperkeratosis, with columns of parakeratosis overlying the papillomatous projections
    • Blood is often seen with the tips of parakeratosis
  • Basal cell carcinoma:
    • May have similar silhouette, in particular to the basaloid variant trichilemmoma
    • Artifactual clefting with mucin within the space, in contrast to the stromal clefting of a benign follicular tumor, is seen
    • Mitotic activity is usually obvious
    • BerEP4 is strong and diffuse (unless superficial or eroded)
    • CD34 is negative
  • Squamous cell carcinoma:
    • In particular the follicular variant may be confused with trichilemmoma
    • Has cellular pleomorphism, brisk and abnormal mitotic activity and invasive borders
    • Lack CD34 expression
  • Trichilemmal carcinoma:
    • Considered the malignant counterpart of trichilemmoma
    • It is very rare
    • Distinction is made by cellular pleomorphism
    • Brisk and abnormal mitotic activity
    • Usually lack CD34 expression
Board review style question #1
Which of the following is true of trichilemmomas?

  1. Are a cutaneous marker of internal malignancy
  2. Are found predominantly on the trunk and acral sites
  3. Are typically present in children
  4. Multiple trichilemmomas are pathognomonic of Cowden syndrome
Board review style answer #1
D. Multiple trichilemmomas are pathognomonic of Cowden syndrome

Comment Here

Reference: Trichilemmoma
Board review style question #2
A 30 year old man had a punch biopsy of a forehead papule which was possible basal cell carcinoma based on appearance.



With respect to the pictographs above

  1. Any focal BerEP4 stain positivity confirms this as a basaloid trichilemmoma
  2. CD34 staining may be very focal and require careful sectioning to demonstrate
  3. Mucin and atypia distinguish this lesion as a follicular squamous cell carcinoma
  4. Presence of clear cells can distinguish basaloid trichilemmoma from basal cell carcinoma
Board review style answer #2
B. This is a trichilemmoma. CD34 staining may be very focal and require careful sectioning to demonstrate.

Comment Here

Reference: Trichilemmoma

Trichoepithelioma / trichoblastoma
Definition / general
  • Trichoepitheliomas and trichoblastomas are benign adnexal tumors, which may show morphologic overlap and recapitulate features of germinative hair bulb epithelium and associated mesenchymal stroma
Essential features
  • Trichoepitheliomas and trichoblastomas are benign adnexal tumors which may display some features that mimic basal cell carcinoma
  • Multiple trichoepitheliomas can be associated with Brooke-Spiegler syndrome and multiple familial trichoepithelioma (CYLD mutations)
  • Trichoblastomas are not known to be familial but are the most common tumor type found in association with nevus sebaceus
Terminology
  • Previous names not widely used: trichogenic tumors, giant solitary trichoepithelioma, trichogerminoma
ICD coding
  • ICD-10: D23.9 - Other benign neoplasm of skin, unspecified
Epidemiology
  • Adults
  • Sporadic trichoepithelioma:
    • Rare
    • Incidence unknown
  • Trichoblastoma:
    • Rare
    • F = M
  • Brooke-Spiegler syndrome / multiple familial trichoepitheliomas:
    • Multiple trichoepitheliomas may present in adolescence / early adulthood
    • F > M
    • CYLD mutations
  • Reference: Cureus 2020;12:e8272
Sites
  • Trichoepithelioma (solitary):
    • Face
    • Rarely scalp, trunk, extremities, genital area
  • Trichoepithelioma (multiple / familial):
    • Symmetrical distribution over central face
    • Rarely other sites as above
    • Rarely dermatomal
  • Trichoblastoma:
    • Scalp, head and neck
    • Occasionally trunk, extremities, genital area
Pathophysiology
  • CYLD mutations give rise to inherited forms of trichoepithelioma
    • Loss of function of CYLD, a deubiquitinating enzyme, results in constitutive NFκB signaling (Nature 2003;424:801)
  • Molecular pathogenesis of sporadic forms of trichoepithelioma unknown
  • Although there may be morphologic overlap, trichoblastomas lack PTCH mutations, distinguishing them molecularly from basal cell carcinomas (Hum Pathol 2007;38:1496)
Clinical features
Diagnosis
Prognostic factors
  • Benign tumors; good prognosis
Case reports
Treatment
  • Surgical excision
Clinical images

Images hosted on other servers:

Multiple unilateral face nodules

Multiple central facial nodules

Microscopic (histologic) description
  • Trichoepithelioma (Am J Dermatopathol 2011;33:251):
    • Usually superficial dermal tumors
    • Superficial nests of basaloid cells with keratin horn cysts
    • Can show leaf-like or frond-like architectural pattern
    • Fibrous cellular stroma closely associated with the epithelial components
    • May have papillary mesenchymal bodies and calcifications
    • May have epidermal connection
    • Ulceration rare
    • Can be basaloid cell predominant with few horn cysts making distinction from trichoblastoma or basal cell carcinoma more difficult
  • Trichoblastoma:
    • Well circumscribed, predominantly dermal tumor nodule which may extend to subcutis
    • Predominantly basaloid epithelial cells in nests with peripheral palisading
    • May have keratin cysts
    • Mitoses and apoptosis can be evident but cellular pleomorphism is minimal
    • Prominent cellular stromal component with papillary mesenchymal body formation
    • Clefting occurs between the epithelial stromal tumor mass and surrounding dermis rather than between epithelial and stromal components
    • May show cribriform, rippled or solid patterns
    • May contain dendritic melanocytes and appear pigmented
    • Typically retains CK20 positive Merkel cells (Arch Pathol Lab Med 2017;141:1490)
Microscopic (histologic) images

Contributed by Eleanor Russell-Goldman, M.D., Ph.D.

Superficial dermal tumor

Prominent keratin cysts

Dermal basaloid tumor

Bland cytology

Papillary mesenchymal body

Positive stains
  • CK20 positive Merkel cell retention is more common in benign follicular tumors versus basal cell carcinoma, although is not specific (Arch Pathol Lab Med 2017;141:1490)
    • This staining pattern should be interpreted with caution as CK20 positive Merkel cells may be only focally represented and may not be apparent without multiple sections
Sample pathology report
  • Skin, right cheek, shave biopsy:
    • Trichoepithelioma / trichoblastoma (see comment)
    • Comment: If partially sampled and a basal cell carcinoma cannot be excluded, recommend complete excision.
Differential diagnosis
  • Basal cell carcinoma
    • Nodular basal cell carcinoma:
      • Clefting between epithelial stromal components
      • Cellular pleomorphism
      • Mucin
      • Frequent epidermal connection
    • Infundibulocystic basal cell carcinoma:
      • Basaloid nests with prominent follicular differentiation
      • May have clefting between epithelial stromal components
      • Less well developed stroma
Board review style question #1

Papillary mesenchymal bodies are a feature of which of the following skin tumors, shown in the image?

  1. Basal cell carcinoma
  2. Microcystic adnexal carcinoma
  3. Spiradenoma
  4. Trichoepithelioma / trichoblastoma
Board review style answer #1
D. Trichoepithelioma / trichoblastoma. Trichoepithelioma is shown.

Comment Here

Reference: Trichoepithelioma / trichoblastoma

Trichofolliculoma
Definition / general
  • Benign adnexal hamartomatous follicular tumor
  • Histologically shows multiple follicles in various stages spreading from a central cystic follicle
Essential features
  • Solitary papule in adults in the head and neck region
  • Central dilated primary follicle with secondary follicles budding from the primary follicle
  • Can show a spectrum of morphology depending on the hair follicle cycle
ICD coding
  • ICD-O: 839 - 842 - adnexal and skin appendage neoplasms
  • ICD-10: D23.9 - other benign neoplasm of skin, unspecified
Epidemiology
Sites
Pathophysiology
  • Repeated development of hair follicles with disordered hair cycle; defective sonic hedgehog polarization (J Dermatol 2017;44:1050, Am J Dermatopathol 2009;31:248)
  • Distorted ability to control the size of hair follicles
  • Trichofolliculoma with sebaceous differentiation: follicular and sebaceous components have independent cycles
  • Primary follicle: (J Dermatol 2017;44:1050)
    • Primary infundibular cystic structure which shows a thin wall with radiating secondary follicles
  • Secondary follicles:
    • Follicles radiating from the primary follicle
    • Most follicles are in anagen phase
  • Tertiary follicles:
    • Regression of secondary follicles to tertiary follicles
    • Shift of anagen hair to catagen phase
    • Variation in size of hair from vellus hair to thick terminal hair
  • Quaternary follicles:
    • Regression of tertiary follicles to quaternary follicles
Clinical features
Diagnosis
  • Clinical: central primary follicle with multiple tufts of vellus hair (Am J Pathol 1976;85:479)
  • If the hair is plucked, trichofolliculoma can be clinically misdiagnosed as basal cell carcinoma, molluscum contagiosum, keratoacanthoma, milium, trichoepithelioma, syringoma or sebaceous hyperplasia (J Eur Acad Dermatol Venereol 2017;31:e123)
  • Dermoscopy: shows troll hair sign - tight plumes of white and thin hairs, similar to children's troll dolls (Australas J Dermatol 2021;62:90)
  • Biopsy and histological examination
Prognostic factors
Case reports
Treatment
Clinical images

Contributed by Sepideh Nikki Asadbeigi, M.D.
Trichofolliculoma

Trichofolliculoma



Images hosted on other servers:

Papule with white vellus hair

Microscopic (histologic) description
  • Dilated central cystic follicle with surrounding multiple fully formed vellus or terminal follicles
  • The central cystic follicle shows connection / opening to epidermis
  • Early lesion: a mildly dilated infundibulum and radiating secondary curved vellus follicles; cystic dilatation may be absent
  • Late lesion: thin walled primary infundibular cystic structure and radiating vellus or terminal follicles that are mostly in the anagen phase
  • Trichofolliculoma demonstrates outer root sheath differentiation
  • Central follicle shows stratified squamous cell epithelium with a granular layer with dilation or cystic changes and contains keratinous material and may have vellus hairs
  • Primary follicle has keratinized stratified epithelium with keratohyaline granules
  • Branched follicles may show varying degree of maturation, including rudimentary structures or epithelial cords and anagen, catagen or telogen hair in older lesions
  • Secondary follicles are small with many epithelial strands and abortive pilar formation
  • Sebaceous differentiation may be present
  • Trichofolliculoma is usually surrounded by well developed connective tissue, which is frequently cellular
  • Each follicle is surrounded by an individual perifollicular sheath
  • Late stage can show a solid pattern as the regressing secondary follicles and developing tertiary follicles coalesce
  • Sebaceous trichofolliculoma variant: sebaceous gland attached to radiating follicles (J Cutan Pathol 1980;7:394)
Microscopic (histologic) images

Contributed by Sepideh Nikki Asadbeigi, M.D.
Primary and secondary follicles

Primary and secondary follicles

Secondary follicles

Secondary follicles

Primary cystic follicle

Primary cystic follicle

Dilated hair follicle

Dilated hair follicle

Secondary hair follicle

Secondary hair follicle

Virtual slides

Images hosted on other servers:
Trichofolliculoma with sebaceous differentiation

Trichofolliculoma with sebaceous differentiation

Positive stains
Molecular / cytogenetics description
Videos

Trichofolliculoma clinical and pathology by Dr. Michael Lee

Trichofolliculoma histopathology by Dr. Jerad Gardner

Sample pathology report
  • Forehead skin, shave biopsy:
    • Trichofolliculoma (see comment)
    • Comment: Atypia is not identified.
    • Microscopic description: Sections demonstrate a centrally dilated follicular unit with multiple radiating follicular units. At the center of the cystic structure there is keratotic debris and immature hair shafts (trichoids). Surrounding the structure is a mantle of well organized connective tissue. Atypical features were not noted.
Differential diagnosis
  • Trichoadenoma:
    • Multiple multilayered squamous epithelial islands with a central cystic cavity containing keratinous material
    • Does not contain hair shafts
  • Solitary trichoepithelioma:
    • Islands of basaloid cells are present, which is not a feature of trichofolliculoma
  • Fibrofolliculoma:
    • Both show a large central follicle with multiple arising epithelial attachments
    • Fibrofolliculoma only has strands of follicular epithelium and does not contain any hair shafts
  • Dilated pore of Winer:
    • Widened follicular infundibulum
    • Unlike trichofolliculoma, it does not show the radiating secondary follicles
  • Hair follicle nevus:
    • Both lesions show multiple hair follicles with vellus hairs but trichofolliculoma has a central cystic component
  • Follicular infundibulum cyst:
    • Does not contain the secondary or tertiary follicular structures
  • Pilar sheath acanthoma:
    • Less florid pattern than trichofolliculoma
    • Dilated central follicle with cystic features and radiating acanthotic epithelium
    • Unlike trichofolliculoma, pilar sheath acanthoma does not have hair shafts in the peripheral buds
  • Folliculosebaceous cystic hamartoma:
    • Folliculosebaceous structures with surrounding stroma with various mesenchymal elements
    • Sebaceous component is the more prominent component
    • Although it has been suggested that this is a late stage of sebaceous trichofolliculoma, reports of congenital folliculosebaceous cystic hamartoma suggest that this idea is incorrect (Am J Dermatopathol 2008;30:500, J Cutan Pathol 2008;35:843)
Additional references
Board review style question #1

A 50 year old Caucasian woman presents with a solitary flesh colored papule with a central depression. What is the diagnosis?

  1. Dilated pore of Winer
  2. Pilar sheath acanthoma
  3. Trichoepithelioma
  4. Trichofolliculoma
Board review style answer #1
D. Trichofolliculoma

Comment Here

Reference: Trichofolliculoma
Board review style question #2
The biopsy of a skin colored papule on the face of a 34 year old man shows a central dilated follicle with smaller radiating follicular units containing hair shafts. What is the most accurate diagnosis?

  1. Hair follicle nevus
  2. Pilar sheath acanthoma
  3. Trichoadenoma
  4. Trichofolliculoma
Board review style answer #2
D. Trichofolliculoma

Comment Here

Reference: Trichofolliculoma

Tufted angioma
Definition / general
  • Benign, acquired vascular tumor most often arising in infancy or early childhood
  • Rare; approximately 200 cases reported in literature to date
Essential features
  • Tufts of capillaries infiltrating the dermis and subcutaneous adipose tissue in a “cannonball” or lobular pattern
  • Considered to be on the same neoplastic spectrum as kaposiform hemangioendothelioma; some consider it to be the same entity
Terminology
  • Synonyms: Tufted angioma, Nakagawa’s angioblastoma, progressive capillary hemangioma, tufted hemangioma
Epidemiology
  • Most commonly affects children and young adults with no gender predilection
  • Rare cases have been reported in adults
  • May be seen in association with Kasabach-Merritt syndrome
  • Some cases have been associated with liver transplantation, pregnancy, healed herpes zoster sites, or vaccination sites
Sites
  • Commonly found on neck, shoulders and trunk
  • Cranial and facial lesions are uncommon
Clinical features
  • Erythematous, poorly defined mottled macules and plaques typically ranging from 2 - 5 cm
  • Usually solitary
  • May have overlying hypertrichosis
  • Slowly growing, spreading lesion
  • Spontaneous regression rarely occurs; most lesions persist
Case reports
Treatment
  • Surgical excision for small lesions; recurrences are common
  • Low dose aspirin
  • High dose steroids (intralesional or systemic)
  • Pulsed dye laser
  • Chemotherapy(vincristine)
Clinical images

Contributed by Mark R. Wick, M.D.


Images hosted on other servers:

Subcutaneous
nodule

Microscopic (histologic) description
  • Multiple, scattered lobules of small capillary type vessels with small oval to spindle shaped cells throughout the dermis and subcutaneous tissue imparting a “cannonball” or glomerular appearance
  • May have variable mitoses without nuclear atypia
  • Hemosiderin may be present; in contrast to pyogenic granuloma, inflammation is typically absent
  • In contrast to Kaposi sarcoma, no slit-like vascular spaces and no plasma cells
  • In contrast to kaposiform hemangioendothelioma, confined to skin and less infiltrative
Microscopic (histologic) images

Contributed by Manuel Valdebran, M.D. and Phil LeBoit, M.D.



Contributed by Joel Tjarks, M.D.



Images hosted on other servers:

Cannon ball distribution

Positive stains
  • CD31, CD34
  • D2-40 highlights surrounding dilated dermal lymphatics
Negative stains
Differential diagnosis
Changed name (dropped Acquired) per Dr. Ho, by Nat, 10Mar22

Tumor of the follicular infundibulum (pending)

Vellus hair cyst
Definition / general
  • Originally reported in children and young adults
  • No gender or racial predilection
Epidemiology
  • Occasionally associated with renal failure or various genodermatoses, including pachyonychia congenita, anhidrotic ectodermal dysplasia, hidrotic ectodermal dysplasia
  • Rarely associated with Lowe syndrome, an oculocerebrorenal syndrome characterized by Fanconi-type renal failure, intellectual disabilities, ocular abnormalities
  • Cases may be inherited (autosomal dominant), which may be manifest at birth, and is more likely to show cysts on extensor aspects of limbs
Sites
  • Small, multiple cysts over chest wall and extremities
Pathophysiology
  • Eruptive vellus hair cysts probably develop due to occlusion of the infundibulum of vellus hairs with resultant cystic dilatation and retention of keratinous debris and vellus hairs
  • The primary cause of the obstruction is unknown
  • They may also represent follicular hamartomas
Clinical features
  • Patients present with numerous asymptomatic, discrete, soft, flesh-colored or reddish-brown papules, 1 - 5 mm across, particularly over the parasternal area, although the distribution may be quite widespread
Case reports
Treatment
  • Local surgery
Clinical images

Images hosted on other servers:
Yellowish, dome shaped papules

Yellowish, dome shaped papules

Hyperpigmented follicular papules

Hyperpigmented follicular papules

Microscopic (histologic) description
  • Mid-dermal cyst containing laminated keratin and many vellus hairs
  • Epithelial lining consists of several layers of squamous epithelium, often with a granular cell layer
Microscopic (histologic) images

Images hosted on other servers:
Acanthotic epidermis

Acanthotic epidermis

Dermal cyst

Dermal cyst

Laminated keratinous material

Laminated keratinous material

Differential diagnosis

Venous lake (pending)
[Pending]

Verrucous carcinoma
Definition / general
  • Verrucous carcinoma is a subtype of squamous cell carcinoma that affects skin and mucosa and has characteristic slow growth and bland histological features; suspect if a longstanding verrucous lesion or cyst is unresponsive to routine treatment (Int J Surg Pathol 2017;25:438)
Essential features
  • Low grade subtype of squamous cell carcinoma with good prognosis when affecting skin
  • Association with human papillomavirus (HPV) DNA is rarely reported in mucosal and genital areas; may represent incidental colonization
  • Association with alcohol consumption, smoking, areca nut chewing and oral microbiota has been reported in oral verrucous carcinoma
  • Can arise in association with inflammatory and neoplastic conditions
  • Diagnosis can be delayed due to bland histopathological features and mimicking reactive processes
  • Caution should be exercised when making diagnosis in superficial or fragmented biopsies
Terminology
  • Ackerman tumor, oral florid papillomatosis, papillomatosis cutis carcinoides
ICD coding
  • ICD-10: C44 - subtype of squamous cell carcinoma
Epidemiology
  • M > F
  • Older individuals (average 50 - 70 years old); 75% of patients > 60 years old (Cancer 2001;92:110)
Sites
  • Skin (e.g., wrists, fingers, nail bed, sole of the foot, ear, nose eyelid, scalp, buttocks, anorectal region, penis, vulva, shoulder, axilla, abdominal wall and lip) and oral cavity
  • Oral cavity: also called Ackerman tumor, oral florid papillomatosis; see esophagus, larynx, oral cavity
Pathophysiology
  • Carcinogens interfere with DNA replication and cause DNA single strand breaks and mutations that facilitate tumor growth (Int J Oncol 2016;49:59)
Etiology
Clinical features
  • Unicentric (in the skin), hyperkeratotic warty growths that are longstanding, progressive and may or may not be locally destructive
Diagnosis
  • Longstanding and slow growing exophytic hyperkeratotic tumor at typical anatomical sites, such as foot and oral cavity
  • Imaging evidence of deep endophytic growth with or without invasion to the bone
  • Evidence of well differentiated squamous proliferation on a shave or punch biopsy in an appropriate clinical and imaging context
  • Definitive diagnosis requires a deep incisional sample or excision of tumor
  • Can be mistaken for benign squamous lesions on superficial biopsies (J Am Acad Dermatol 1995;32:1)
Prognostic factors
  • Recurrence rate high if incompletely excised
  • Recurrent tumor can be more aggressive than original, with bone and cartilage invasion and destruction
  • Metastases are rarely documented, most often from oral and mucosal lesions (In Vivo 2007;21:909, Cancer 2001;92:110)
  • Enlargement of draining lymph nodes is common and usually represents reactive hyperplasia secondary to inflammatory reaction to the tumor (Head Neck Surg 1982;5:29)
  • Progression to aggressive squamous cell carcinoma after radiation or chemotherapy
Case reports
Treatment
  • Surgical excision is the most common mode of treatment (J Dermatolog Treat 2021 Apr 14 [Epub ahead of print])
  • Other modes: cryosurgery, carbon dioxide laser, chemotherapy, intralesional or iontophoretic methods, photodynamic therapy, systemic retinoid therapy, radiotherapy
Clinical images

Images hosted on other servers:
Finger tumors Finger tumors

Finger tumors

Vulvar tumor

Vulvar tumor

Heel tumor

Heel tumor

Gross description
  • Fungating, verrucous or polypoid lesions sometimes with ulceration
  • May invade and destroy underlying bone
Gross images

Contributed by Amanda Ireland, M.B.B.S.
Dorsal view

Dorsal view

Palmar view

Palmar view

Lateral view

Lateral view

Microscopic (histologic) description
  • Histologically similar regardless of anatomical site
  • Well differentiated squamous proliferation with exophytic and endophytic components
  • Exophytic component with papillomatosis and massive hyperkeratosis, often with expanded granular layer with parakeratosis
  • Endophytic component with blunt projections of well differentiated squamous epithelium with deep bulbous processes and a pushing margin
  • Constituent cells; large polygonal squamous cells with abundant pink cytoplasm and enlarged nuclei with minimal nuclear atypia (apart from several layers at the interface of bulbous tips)
  • Often inflamed edematous stroma filled with lymphocytic cells immediately adjacent to the advancing edge
  • Tissue between bulbous processes is greatly diminished with reduced vascularity
  • Deeper biopsy shows broad bands of epidermal proliferation with parakeratotic centers; bases of proliferation are large and bulbous and invade deep dermis in a pushing manner (Calonje: McKee's Pathology of the Skin, 5th Edition, 2019)
Microscopic (histologic) images

Contributed by Amanda Ireland, M.B.B.S.
Exophytic / endophytic squamous proliferation

Exophytic / endophytic squamous proliferation

Superficial aspect

Superficial aspect

Deep aspect

Deep aspect

Pushing margins

Pushing margins

Tumor stromal interface

Tumor stromal interface

Virtual slides

Images hosted on other servers:
Verrucous carcinoma, skin Verrucous carcinoma, skin

Verrucous carcinoma, skin

Verrucous carcinoma in wart

Verrucous carcinoma in wart

Verrucous carcinoma, vulva

Verrucous carcinoma, vulva

Molecular / cytogenetics description
Sample pathology report
  • Skin, excision:
    • Verrucous carcinoma, clear of the margins (see comment)
    • Comment: Microscopic examination reveals a well differentiated squamous proliferation characterized by an acanthotic squamous epithelium with exophytic papillary processes with overlying hyperkeratosis and parakeratosis. There is also an associated endophytic component composed of deeply penetrating bulbous processes with pushing borders. The keratinocytes are well differentiated with abundant glassy cytoplasm and enlarged nuclei. Mitoses are rare and there is no tumor necrosis. The superficial dermis shows a band-like lymphocytic inflammation. There is no lymphovascular or perineural invasion. The tumor is well clear of the resection margins.
Differential diagnosis
  • Giant condyloma / Buschke-Löwenstein tumor:
    • Histologically identical but detection of low risk HPV (6 and 11) by PCR indicates a giant condyloma (Histopathology 2017;70:938)
  • Verrucae (warts):
    • Distinction difficult in superficial biopsies
    • Warts often have well formed lateral collarets, a significantly expanded granular layer with coarse basophilic keratohyalin granules and no deeply extending bulbous processes
  • Pseudoepitheliomatous epidermal hyperplasia (PEH):
    • Distinction is difficult in superficial biopsies
    • PEH is more superficial, extends horizontally with at most a focal, limited deep, bulbous growth
  • Verrucous psoriasis:
    • Regular acanthosis and Munro microabscesses
    • If there is considerable traumatization and pseudoepitheliomatous hyperplasia, clinical correlation and history of psoriasis is key (JAAD Case Rep 2019;5:723)
  • Fungal / protozoal / mycobacterial infection with extensive pseudoepitheliomatous hyperplasia:
    • Look for organisms in the dermis with or without special stains
  • Keratoacanthoma:
    • Often more endophytic growth with a central cavity / crater filled with keratin
    • May contain intraepidermal neutrophilic microabscesses
    • May show signs of regression / atrophy, surface undulation and stromal fibrosis and inflammation
    • Usually does not extend beyond eccrine glands
  • Squamous cell carcinoma (conventional and nonverrucous subtypes):
    • Depending on differentiation, often shows variable degrees of cytologic atypia, mitotic activity and infiltrative growth, at least focally
    • Distinction from a well differentiated conventional SCC is difficult; however, the latter is usually less exophytic
  • Carcinoma cuniculatum / epithelioma cuniculatum:
    • Well differentiated squamous carcinoma that presents on the plantar foot
    • No significant verrucous or exophytic growth
    • Surface is flat with infiltrative growth of sinus tracts (J Cutan Pathol 2012;39:1083)

Differential diagnosis of verruciform tumors (adapted from figure 12.215 on page 534 of Calonje: McKee's Pathology of the Skin, 5th Edition, 2019)
Papillae Viral cytoplasmic effect Lower border HPV status
Verrucous carcinoma Thin and elongated - usually without fibrovascular core Absent Bulbous Often negative
Giant condyloma Fibrovascular cores Focal, mainly superficial Bulbous Low risk HPV
Warty carcinoma Various shapes with thick cores Diffuse Irregular High risk HPV
Papillary carcinoma Complex Absent Irregular Negative
Board review style question #1

Which statement is applicable to the tumor depicted in the image shown above?

  1. Enlargement of draining lymph nodes commonly occurs
  2. Initial rapid growth often followed by regression
  3. Poorly differentiated and aggressive squamous neoplasm with bulbous processes
  4. Recurrence with local destructive growth is common following a complete surgical excision
Board review style answer #1
A. Enlargement of draining lymph nodes commonly occurs

Comment Here

Reference: Verrucous carcinoma
Board review style question #2
Which of the following options best describes the histological features of verrucous carcinoma?

  1. Exophytic and endophytic growth of bland appearing squamous epithelium with deep pushing borders
  2. Exophytic verrucous growth of squamous epithelium commonly with koilocytosis change
  3. Exophytic verrucous growth of squamous epithelium with high mitotic activity and necrosis
  4. Well differentiated squamous neoplasm with mainly endophytic infiltrative growth
Board review style answer #2
A. Exophytic and endophytic growth of bland appearing squamous epithelium with deep pushing borders

Comment Here

Reference: Verrucous carcinoma

WHO classification
Definition / general
  • WHO classification of nonmelanocytic skin tumors
Major updates
  • Epidermal tumors
    • Keratoacanthoma (KA) is kept separate from squamous cell carcinoma (SCC), while recognizing it may be a well differentiated SCC with self resolving tendencies
    • Merkel cell carcinoma is now classified as a neuroendocrine carcinoma of the skin
  • Appendageal tumors
    • Genetic tumor syndromes associated with skin malignancies chapter discusses adnexal tumor syndromes in depth
    • New molecular aberrations in nonsyndromic appendageal tumors added
    • Cribriform carcinoma renamed cribriform tumor
    • Oncogenic HPV42 present in digital papillary adenocarcinoma
    • Addition of (provisional) entity: primary adnexal NUT carcinoma added
  • Tumors of nail unit added
  • Hematolymphoid tumors
    • New insights into the molecular pathophysiology and response to targeted therapy
    • Updates for histiocytic / dendritic cell neoplasms (ALK and BRAF mutation drivers)
    • Cutaneous peripheral T cell lymphomas listed separately
    • Cutaneous peripheral T cell lymphoma, NOS is applied to cases not falling within a specific category
    • Section on primary immunodeficiency associated lymphoproliferative disorders added
    • Reinclusion of reactive lymphoid hyperplasia (last covered in 3rd edition)
  • Soft tissue tumors
    • Introduction of 4 new entities
      • CRTC1::TRIM11 cutaneous tumor
      • Superficial CD34+ fibroblastic tumor (PRDM10 rearranged soft tissue tumor)
      • EWSR1::SMAD3 rearranged fibroblastic tumor
      • NTRK rearranged spindle cell neoplasm
    • Atypical intradermal smooth muscle neoplasm preferred terminology for dermal leiomyosarcoma
    • Reclassification of epithelioid fibrous histiocytoma in family of tumors of uncertain differentiation
  • Skin metastasis
    • Known tumors metastasizing to skin
    • Carcinoma, unknown primary
WHO (2022)
    Keratinocytic / epidermal tumors
    ICD-O
    ICD-11
    • Carcinoma precursors and benign simulants
    • Epidermal carcinomas
      • Basal cell carcinomas
        8090/3
        2C32.Z
        • Superficial basal cell carcinoma
          8091/3
          2C32.2
        • Nodular basal cell carcinoma
          8097/3
          2C32.0
        • Micronodular basal cell carcinoma
          8097/3
          2C32.Y & XH4GJ2
        • Infiltrating basal cell carcinoma
          8092/3
          2C32.Y & XH5VK4
        • Sclerosing / morphoeic basal cell carcinoma
          8092/3
          2C32.1
        • Basosquamous carcinoma
          8094/3
          2C32.Y & XH4C18
        • Basal cell carcinoma with sarcomatoid differentiation
          8092/3
          2C32.Y & XH1JH6
        • Basal cell carcinoma with adnexal differentiation
          8090/3
          2C32.Y & XH6S67
        • Fibroepithelial basal cell carcinoma
          8093/3
          2C32.Y & XH45F3
      • Keratoacanthomas
        • Keratoacanthoma
          8071/3
          2C31.1
          • Giant keratoacanthoma
          • Keratoacanthoma centrifugum marginatum
          • Multiple keratoacanthoma-like proliferations and eruptive squamous atypia
          • Keratoacanthoma in Muir-Torre syndrome
          • Multiple self healing epitheliomas of Ferguson-Smith
          • Generalized eruptive keratoacanthoma of Grzybowski
      • Squamous cell carcinomas
        8070/3
        2C31.Z
        • Verrucous squamous cell carcinoma
          8051/3
          2C31.0
        • Acantholytic squamous cell carcinoma
          8075/3
          2C31.Z & XH7LH0
        • Lymphoepithelial carcinoma
          8082/3
          2C31.Z & XH1E40
        • Clear cell squamous cell carcinoma
          8084/3
          2C31.Z & XH9DC1
        • Spindle cell squamous cell carcinoma
          8074/3
          2C31.Z & XH6D80
        • Squamous cell carcinoma with sarcomatoid differentiation
          8074/3
          2C31.Z & XH6D80
    • Neuroendocrine neoplasms
      • Neuroendocrine carcinomas
        • Merkel cell carcinoma
          8247/3
          2C34 & XH81N8
          • Merkel cell polyomavirus positive versus negative
          • Pure versus combined (most commonly with squamous cell carcinoma)










    Metastases to skin
    ICD-O
    ICD-11


    Genetic tumor syndromes associated with skin malignancies
    ICD-11
  • Familial melanoma
  • BAP1 tumor predisposition syndrome
  • Xeroderma pigmentosum
    LD27.1
  • Nevoid basal cell carcinoma syndrome (Gorlin syndrome)
    LD2D.4
  • Carney complex
  • Muir-Torre syndrome
  • Brooke-Spiegler and related syndromes
    2F22
Microscopic (histologic) images

Contributed by Jonathan D. Ho, M.B.B.S., D.Sc.
Keratoacanthoma

Keratoacanthoma

Bowen disease (squamous cell carcinoma in situ)

Bowen disease (squamous cell carcinoma in situ)

Merkel cell carcinoma

Merkel cell carcinoma

Syringocystadenoma papilliferum

Syringocystadenoma
papilliferum

Eccrine syringofibroadenoma

Eccrine
syringofibroadenoma

Ductule formation in syringofibroadenoma

Ductule formation in
syringofibroadenoma


Microcystic adnexal carcinoma

Microcystic adnexal carcinoma

Trichilemmoma

Trichilemmoma

Malignant proliferating trichilemmal tumor

Malignant proliferating trichilemmal tumor

Eccrine porocarcinoma

Eccrine porocarcinoma

Eccrine porocarcinoma

Eccrine porocarcinoma

Eccrine porocarcinoma

Eccrine porocarcinoma


Onychomatricoma

Onychomatricoma

Onychomatricoma keratinization

Onychomatricoma
keratinization

Mycosis fungoides

Mycosis fungoides

Primary cutaneous anaplastic large cell lymphoma

Primary cutaneous anaplastic large cell lymphoma

CD30 positivity in ALCL

CD30 positivity in ALCL

Adult maculopapular cutaneous mastocytosis

Adult maculopapular cutaneous mastocytosis


Angiolipoma

Angiolipoma

Dermatofibrosarcoma protuberans

Dermatofibrosarcoma
protuberans

Cutaneous angiosarcoma

Cutaneous angiosarcoma

Schwannoma

Schwannoma

Kaposi sarcoma

Kaposi sarcoma

HHV8 in Kaposi sarcoma

HHV8 in Kaposi sarcoma

Board review style question #1

Which of the following statements is true regarding Merkel cell carcinoma?

  1. Characterized by a coarse chromatin pattern
  2. Considered a tumor of neural origin
  3. Has an indolent course
  4. May be associated with polyomavirus infection
  5. Typically expresses CK7
Board review style answer #1
D. May be associated with polyomavirus infection. Merkel cell polyomavirus may be seen in up to 80% of tumors in some populations. Answer A is incorrect because Merkel cell carcinoma is characterized by finely dispersed salt and pepper chromatin. Answer B is incorrect because Merkel cell carcinoma is classified as a cutaneous neuroendocrine tumor. Answer C is incorrect because Merkel cell carcinoma has an aggressive course with metastatic potential. Answer E is incorrect because Merkel cell carcinoma is typically CK20+ and CK7-. Only very rare cases of CK7+ have been described.

Comment Here

Reference: Skin nonmelanocytic tumor - WHO classification

Warty dyskeratoma
Definition / general
  • Also called isolated follicular keratosis
  • Small maculopapular lesion of sun-exposed skin of head / neck
  • Either follicular counterpart of actinic keratosis or a follicular neoplasm
  • Not related to Darier’s disease
Case reports
Clinical images

Images hosted on other servers:
Warty dyskeratoma

Warty dyskeratoma

Gross description
  • Solitary, well-defined nodule
Microscopic (histologic) description
  • Cup-shaped keratin-filled invagination
  • Acanthotic epidermis with acantholytic dyskeratotic cells
  • Suprabasilar clefting with villi projecting into clefts
Microscopic (histologic) images

Contributed by Mowafak Hamodat, M.B.Ch.B., M.Sc.


Warty dyskeratoma Warty dyskeratoma Warty dyskeratoma Warty dyskeratoma

Warty dyskeratoma


White sponge nevus
Definition / general
  • Not a melanocytic entity; benign keratinization defect leading to white plaques on mucosa
  • Primarily affects nonkeratinized stratified squamous epithelium
  • First described by Cannon in 1935 (Arch Dermatol Syphilol 1935;31:365)
Terminology
  • Synonyms: familial white folded mucosal dysplasia, leukoderma exfoliativum mucosae oris, hereditary leukokeratosis
Epidemiology
Sites
  • Buccal, gingival, esophageal, rectal or genital mucosal surfaces
Clinical features
  • White to gray, diffuse, painless, spongy and folded plaques on buccal mucosa
Case reports
Treatment
  • No standard treatment; although reports of tetracycline, chlorhexidine
  • Progression usually stops at puberty
  • No malignant transformation
Clinical images

Images hosted on other servers:

White plaques on buccal mucosa

White plaques on tongue

Microscopic (histologic) description
  • Parakeratosis, acanthosis with formation of large blunt rete ridges and spongiosis
  • Extensive vacuolation of suprabasal keratinocytes
  • Dyskeratotic cells exhibit dense peri and paranuclear eosinophilic condensations, which correspond to tonofilament aggregates
  • Abundant Odland bodies (keratinosome, membrane bound granule in upper stratum spinosum) within keratinocytes but few are present in the intercellular spaces
Microscopic (histologic) images

Images hosted on other servers:

Parakeratosis

Differential diagnosis

Xanthoma
Definition / general
  • Nonneoplastic
  • Often periarticular; also trunk or extremities of males
  • Associated with hyperlipidemia (primary or secondary to diabetes, hypothyroidism, myeloma, lymphoma, leukemia, obstructive liver disease)

Eruptive xanthoma:
  • Abrupt onset of crops of yellow papules with erythematous halos on extremities, which wax and wane with triglyceride and cholesterol levels

Plane xanthoma:
  • Linear yellow lesions in skinfolds, including palmar creases
  • Associated with primary biliary cirrhosis

Tuberous / tendinous xanthoma:
  • Yellow nodules on Achilles tendon and extensor tendons of fingers

Verruciform xanthoma:
  • Papillomatous, verruca-like change of overlying epidermis

Xanthelasma:
  • Soft yellow papules and plaques in eyelid
  • Some cases lack lipid abnormalities
Gross description
  • Nodules
Microscopic (histologic) description
  • Fat laden histiocytes in dermis or subcutis
  • Also in tendons, synovium and bone
Microscopic (histologic) images

Contributed by Hillary Rose Elwood, M.D., Andrey Bychkov, M.D., Ph.D. and Jijgee Munkhdelger, M.D., Ph.D.

Eruptive xanthoma: dermal foamy histiocytes with admixed neutrophils and extravascular lipid


Mild chronic inflammation

Foam cells

Cholesterol cleft

Abundant foam cells

Lipid laden macrophages

Back to top
Recent Skin nonmelanocytic tumor Pathology books

Bhawan: 2022

Biswas: 2017

Bolognia: 2017

Brenn: 2017

Brenn: 2021

Busam: 2015

Calonje: 2019

Carter: 2015

Cerroni: 2020

Dadzie: 2016

Elder: 2022

Elder: 2020

Elston: 2018

Gardner: 2019

Gru: 2023

IARC: 2018

Johnston: 2023

Patterson: 2020

Plaza: 2016

Rapini: 2021

Subtil: 2019



Find related Pathology books: dermatopathology, hematopathology, IHC
Image 01 Image 02