Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Abstracts
Book Review
Case Report
Case Series
CMAS‡ - Pancreas - EUS-FNA Cytopathology (PSC guidelines) S1:1 of 5
CMAS‡ - Pancreas - EUS-FNA Cytopathology (PSC guidelines) S1:3 of 5
CMAS‡ - Pancreas - EUS-FNA Cytopathology (PSC guidelines) S1:4 of 5
CMAS‡ - Pancreas -Sampling Techniques for Cytopathology (PSC guidelines) S1:2 of 5
CMAS‡ - Pancreas- EUS-FNA Cytopathology (PSC guidelines) S1:5 of 5
Commentary
CytoJournal Monograph Related Review Series
CytoJournal Monograph Related Review Series (CMAS), Editorial
CytoJournal Monograph Related Review Series: Editorial
Cytojournal Quiz Case
Editorial
Erratum
Letter to Editor
Letter to the Editor
Letters to Editor
Methodology
Methodology Article
Methodology Articles
Original Article
Pap Smear Collection and Preparation: Key Points
Quiz Case
Research
Research Article
Review
Review Article
Systematic Review and Meta Analysis
View Point

Add Your Heading Text Here


Fine-needle aspiration of basaloid scalp lesion: Potential diagnostic pitfall

QUIZ DESCRIPTION

Gradually, enlarging 4 × 4 cm, well-circumscribed, mobile scalp nodule over the left parietal area in a 38-year-old man, noted after hitting a towel rack 7 months ago. Papanicolaou stained fine-needle aspiration smears stained showed groups of basaloid cells and eosinophilic shadows (ghost cells) with focal foreign body giant cells [Figure 1].

(a) The aspirate showed scattered groups of basaloi d cells with cohesive clumps of keratin debris. (b) ×60 the basaloid cells had scant cytoplasm with indistinct cell borders and hyperchromatic nuclei. (c) ×20 Tight clumps of keratinized debris with red arrow indicatingthe area undergoing zooming Tight clumps of keratinized debris with some identifiable ghost cells (yellow arrows in d). (d) 100×: ghost cells identified by yellow arrows. The tightly cohesive groups of anucleate keratinized cells did not spread well as compared to other squamous epithelial lesions and so the details of individual cells as ghost cells in cytology smears were difficult to evaluate and photograph (Pap stained direct smear. a: ×10; b: ×60; c: ×20; d: ×100 Oil with light increased).

MORPHOLOGY QUIZ

1. What is the most likely diagnosis?

Pilomatricoma

Melanoma

Merkle cell carcinoma

Squamous cell carcinoma

Proliferating trichilemmal tumor

Answer: a

EXPLANATION

Option a: Pilomatricoma showed cellular aspirates comprised of micro-fragments containing groups of basaloid cells without peripheral palisading. The basaloid cells showed high nuclear: cytoplasmic ratio, evenly dispersed chromatin with a few cells showing prominent nucleoli. Clumps of refractile keratin and multinucleated giant cells were also seen in the background. These tumors can have mitosis and, hence, could be misinterpreted as carcinomas. Even thought, it may be difficult to evaluate in fine-needle aspiration (FNA) aspirate, ghost cells (a.k.a. eosinophilic shadow cells), which have no nuclear staining, are clues to correct diagnosis.

Option b: Melanoma may present as asymmetrical and gradually enlarging lesion with irregular borders. However, this lesion is symmetrical with well-defined borders and uniform tan color. Desmoplastic melanoma is usually present on the head-and-neck region and can appear as erythematous, pink or pale nodules or plaque.[1] Histologically, melanoma can mimic features of other tumors such as neuroendocrine tumors, poorly differentiated carcinoma, and lymphoma.[2] A typical case of melanoma shows high nuclear to cytoplasmic ratio with a prominent cherry red nucleoli and hyperchromatic nuclei with irregular nuclear membrane.[2] Immunohistochemistry is widely used in melanoma to differentiate it from other tumors. S-100 is commonly used and has good sensitivity.[2] Other markers have good specificity such as Melan-A, tyrosinase, or HMB45.[2]

Option c: Merkel cell carcinoma (MCC) often appears as asymptomatic red, pink, or blue papules or nodules and lacks distinctive clinical features. Histopathologically, it is marked by round nuclei with finely granular chromatin, inconspicuous nucleoli, scant cytoplasm, and nuclear crush artifacts, along with multiple mitotic figures and apoptotic bodies resembling small cell carcinoma.[3] MCC shows high sensitivity to cytokeratin 20 and neurofilaments, whereas negative staining for thyroid transcription factor 1 and cytokeratin 7 helps distinguish it from small cell carcinoma.[3]

Option d: Squamous cell carcinoma (SCC) usually presents as a red scaly plaque. SCC can be classified with various degrees of differentiation from well to poorly differentiated. SCC can be classified into many subtypes and histopathologic features include atypical squamous cells, keratin pearls, and sometimes hyperkeratosis.[4] Well-differentiated tumors exhibit interconnecting follicular infundibular type squamous epithelium.[4] SCC is characterized by atypical squamous cells, whereas pilomatricoma consists of basaloid cells and shadow cells. In addition, immunohistochemistry is not typically used except in the cases of poorly differentiated SCC.[4] Cutaneous SCC stains positive with p63, p40, MUC1 (epithelial membrane antigen), CK5/6, MNF116, and high-molecular weight 34 E12. BerEp4 should be negative in SCC.[5]

Option e: Proliferating trichilemmal tumor is a tumor originating from the outer root sheath of a hair follicle.[6] The fine-needle aspirates are usually cellular and show syncitial sheets of squamous epithelial cells, amorphous material, keratin, and clusters of small cells with hyperchromatic nuclei mostly basaloid cells.[6] This lesion does not show ghost cells, calcification, cholesterol clefts, and foreign body giant cells.[6]

FOLLOW-UP OF THE CASE

The lesion showed [Figure 2a-d] two types of cells: Basaloid cells and shadow cells. Basaloid cells are present along the periphery in variable proportion. They are small, monotonous with scant cytoplasm with indistinct cellular borders and evenly distributed chromatin. Basaloid cells merge gradually or abruptly with so-called ghost or shadow cells. Ghost cells have abundant pink cytoplasm and open space at their center. Focally, the periphery of the lesion showed foreign body giant cell reaction.

(a-d) Clumps of keratinized debris with some identifiable ghost cells (yellow arrows). The tightly cohesive groups of keratinized cells did not spread well as compared to other squamous epithelial lesions, making the details of individual cells as ghost cells in cytology smears difficult to appreciate and photograph. (Pap stained direct smear. a: ×10; b: ×60; c: ×100 Oil, Focus 1 to highlight the overall structure; d: ×100 Oil, Focus 2 to provide enhanced detail with increased light.

The excisional biopsy showed an intradermal, fairly well-circumscribed lesion measuring 4 × 4 × 4 cm. Microscopic images are shown in Figure 3.

Hematoxylin and Eosin (HE) stained sections of resection (a: ×4; b-d: ×20 ). (a) Islands of basaloid cells at periphery with center showing shadow cells. (b) Basaloid cells with shadow cells. (c and d) Giant cells surrounding an area with keratinized ghost cells.

ADDITIONAL QUIZ QUESTIONS

2. How does the expression of beta-catenin and p63 in pilomatricoma differ in intensity and distribution from other hair matrix tumors?

Pilomatricoma exhibits nuclear beta-catenin and nuclear p63

Pilomatricoma exhibits membranous beta-catenin and cytoplasmic p63

Pilomatricoma exhibits nuclear beta-catenin and absent p63

Pilomatricoma exhibits absent both beta-catenin and p63

Answer: a

EXPLANATION

a) Pilomatricoma exhibits nuclear beta-catenin and nuclear p63 Explanation: The expression and distribution of certain proteins, such as beta-catenin and p63, can be used to differentiate pilomatricomas from other types of hair matrix tumors. Beta-catenin was positive in the basaloid cell population, showing both cytoplasmic and nuclear staining.[7,8] Nuclear localization is particularly characteristic of pilomatricoma and can help distinguish it from other skin tumors, where beta-catenin is usually restricted to the membrane.[8] p63 is a marker that is often used to identify squamous epithelium and squamous differentiation.[8] In pilomatricoma, p63 typically shows strong positivity in the basaloid cells surrounding the ghost cells, reflecting the squamous nature of the tumor.[7]

3. What is the recommended treatment approach for pilomatricoma?

Surgical excision with clear margins

Observation as the pilomatricoma will spontaneously regress

Steroid injection

Radiation therapy

Topical chemotherapy

Answer: a

a) Surgical excision with clear margins. Because it is a benign tumor, complete removal with clear margins is usually curative and prevents recurrence.[9]

BRIEF REVIEW OF TOPIC

Pilomatricoma (also known as pilomatrixoma and calcifying epithelioma of Malherbe) is a benign tumor of hair follicle matrix that most frequently develops with bimodal age distribution of first and sixth decade.[9] Pilomatricoma usually presents as a firm nodule with the overlying skin appearing normal but can present with blue-red discoloration.[9] The tumor can occasionally have keratotic appearance, imitating squamous cell carcinoma, or can present with telangiectasia resembling basal cell carcinoma (BCC).[9] Although malignancy is rare, pilomatricoma can progress into pilomatrix carcinoma.[10] Pilomatricoma is commonly seen in the head-and-neck area.[10] Involvement of palms and soles or genitalia has not been reported.[10] Surgical excision with clear margins is the recommended treatment.[10] Following total resection, recurrence is uncommon.[10]

The histological appearance of a pilomatricoma is solid and nests of basaloid cells. This is different from other types of hair matrix tumors which typically have peripheral palisading. The presence of ghost cells, which are tiny, pale cells that seem “ghostly” under the microscope, is a crucial diagnostic clue of pilomatricomas. They arise when the cytoplasm of maturing cells is replaced by keratin, a protein found in the epidermis, or outermost layer of the skin.[11] Beta-catenin in pilomatricoma often shows both nuclear and cytoplasmic accumulation and p63 typically demonstrates strong nuclear positivity in the basaloid cells surrounding the ghost cells.[7,8] The nuclear distribution of both beta-catenin and p63 is a distinguishing feature from other hair matrix tumors, where they may show a predominantly membranous pattern of beta-catenin and variable P63.

Pilomatricomas are commonly misdiagnosed and are rarely considered as part of the differential diagnosis due to multiple overlapping cytologic features as shown in Figure 4. Clinical differential diagnosis includes but is not limited to BCC which lacks shadow cells and frequently has a mucinous stroma. BCC has many subtypes and are characterized as high-risk and low-risk histologic subtypes. Nodular BCC is the most common and falls under low-risk subtypes.[12] Nodular BCC can appear as a pearly, translucent papule or nodule with telangiectasia and histopathology usually show basaloid keratinocytes with peripheral palisading and clefting.[12,13] Micronodular, infiltrative, sclerosing, and morpheaform are considered higher-risk subtypes and more likely to recur than low-risk histologic subtypes which include superficial and nodular BCC.[13]

Pilomatricoma fine-needle aspiration alg orithm. SCC: Squamos cell carcinoma The figure was made using the software PowerPoint for Microsoft 365 (2024), Microsoft, USA.

Trichilemmal cysts (TCs) are benign cystic lesions originating from follicular isthmus epithelium and can present similar to pilomatricoma with slow-growing, asymptomatic, and firm nodules in the scalp.[14] However, histologically TC found to have squamoid cells with abundant eosinophilic cytoplasm, compared to basaloid small cells in pilomatricoma.

Epidermal inclusion cyst was considered on the differential and usually occurs on the scalp, neck, back, and extremities.[15] However, it is less likely given absence of central punctum on examination. Lipoma is a benign adipose tumors and can present similar to pilomatricoma but less likely since FNA did not visualize adipose tissue cells.[16]

MCCs do not have distinctive clinical features and can present as asymptomatic red or pink or blue papules or nodules.[3] Histopathologic features include round nuclei with finely granular chromatin, inconspicuous nucleoli, scant cytoplasm, showing molding, and crush artifact.[3] The high nucleus: cytoplasm ratio gives it a blue or basaloid appearance (3). In addition, immunostains have good sensitivity and can further confirm the diagnosis of MCC which includes cytokeratin 20, low-molecular-weight cytokeratins (e.g., CAM5.2), and neurofilaments.[3] Electron microscopy shows paranuclear and/or cytoplasmic bundles of tonofilaments (keratin intermediate filaments), cytoplasmic dense-core granules, and desmosomal attachments.[3] FNA of MCC typicall show small, round, or oval cells with round-to-oval nuclei and scant cytoplasm. Nuclei have fine chromatin and small, inconspicuous nucleoli. Occasional nuclear moldingcan be observed.[17]MCC is less lik ely because histopathology lacks shadow cells. Table 1[3,4,13-18] shows the differential diagnosis for pilomatricoma.

Differential diagnosis.

Diagnosis Description Physical examination Histology Fine-needle aspiration
TC Slow-growing, asymptomatic or mildly painful. Usually present in the scalp. Freely mobile, and firm, skin-colored nodules. TC Can be erythematous after trauma. Cyst lumen contains homogeneous eosinophilic and keratinous materials. Cystic structure lined by stratified squamous epithelium without a granular cell layer.[14] Anucleate and nucleated squamous cells without atypia, basaloid cells without peripheral palisading but with abrupt keratinization and keratinous debris.[14]
EIC Asymptomatic, slowly enlarging, usually occur on the scalp, skin of head and neck, back and extremities.[15] Firm, round, and subcutaneous nodule with central punctum.[15] EIC is lined by a stratified squamous epithelium with a granular layer and contains lamellated keratin flakes.[18] numerous anucleate squames alongside nucleated benign squamous cells. With secondary infection, aspirates turn turbid, showing inflammatory cells like neutrophils and histiocytes.[15]
Lipoma Asymptomatic. Skin-colored subcutaneous nodule. Lobules of mature adipocytes separated by thin fibrous septa. Mature adipocytes, which appear as large, uniform, and empty-appearing cells with eccentric nuclei.[16]
BCC Slow-growing and commonly found in sun-exposed areas. Nodular BCC can appear as a pearly, translucent papule or nodule with telangiectasia. Nodular subtype-basaloid keratinocytes with peripheral palisading and clefting.[13] Clusters of basaloid cells, possibly with peripheral palisading and mucinous stroma.
SCC Typically found on sun-exposed areas. Red, scaly plaques or nodules, often with ulceration or crusting. Atypical squamous cells with pleomorphism and mitotic figures, forming nests and infiltrating the dermis. Keratin pearls may be present.[4] Dysplastic squamous cells, singly dispersed or in clusters, with high nucleus-to-cytoplasm ratio and keratin pearls.
MCC Varying presentation. Red or pink or blue papules or nodules Round nuclei with finely granular chromatin, inconspicuous nucleoli, scant cytoplasm, showing molding, and crush artifact.[3] Small, round, or oval cells with round-to-oval nuclei and scant cytoplasm. Nuclei have fine chromatin and small, inconspicuous nucleoli. Occasional nuclear molding can be observed.[17]

TC: Trichilemmal cyst, EIC: Epidermal inclusion cyst, BCC: Basal cell carcinoma, SCC: Squamous cell carcinoma, MCC: Merkel cell carcinoma

SUMMARY

Rare pilomatrixoma can be elusive if not kept in the differential of basaloid neoplasms in fine needle aspiration of subcutaneous head-and-neck lesions. FNA from pilomatrixoma can be cellular with cells showing high nuclear to cytoplasmic ratio, prominent nuclei, and mitotic figures. Clumps of refractile keratin and “ghost cells” are major clues to appropriate diagnosis. Atypical basaloid cells and mitotic figures may lead to a diagnostic pitfall to report this entity as a malignancy. Ghost cells are important for the cytological diagnosis of pilomatricoma.

AVAILABILITY OF DATA AND MATERIALS

All data generated or analyzed during this study are included in this published article. No additional datasets were generated or analyzed during the current study. Therefore, data sharing is not applicable to this article as all necessary information is provided within.

ABBREVIATIONS

FNAC – Fine-needle aspiration cytology.

AUTHOR CONTRIBUTIONS

CB, SA, MYAK and VBS: Made substantial contributions to the conception and design of the work, led the initial drafting of the manuscript, and was primarily responsible for its overall structure and content; CB and SA: Significantly contributed to the study design and data analysis; MYAK: Provided critical insights into the methodology and interpretation of results; VBS: Offered expert guidance on the clinical implications and supervised the project.

All authors critically reviewed and revised the manuscript to ensure its intellectual content and integrity, and have given final approval of the version to be published. They all agree to be accountable for all aspects of the work.


References

Rastrelli M Tropea S Rossi CR Alaibac M Melanoma: Epidemiology, risk factors, pathogenesis, diagnosis and classification In Vivo 201428100511
Ohsie SJ Sarantopoulos GP Cochran AJ Binder SW Immunohistochemical characteristics of melanoma J Cutan Pathol 20083543344 10.1111/j.1600-0560.2007.00891.x
Han SY North JP Canavan T Kim N Yu SS Merkel cell carcinoma Hematol Oncol Clin North Am 201226135174 10.1016/j.hoc.2012.08.007
Waldman A Schmults C Cutaneous squamous cell carcinoma Hematol Oncol Clin North Am 201933112 10.1016/j.hoc.2018.08.001
Murphy G Beer T Cerio R Kao G Nagore E Pulitzer M Keratinocytic/epidermal tumours Elder D Massi D Scolyer R Willemze R editors World Health Organization (WHO) Classification of skin tumours 4th ed 11 International Agency for Research on Cancer (IARC) Argonay, France 20183143 Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9913635/ [Last accessed on 2023 Jan 04]
Abdou AG Cytologic findings of proliferating trichilemmal tumor (PTT) of scalp Diagn Cytopathol 20204818689 10.1002/dc.24322
Ammendola S Bariani E Tsvetkova V Gisondi P Rosina P Girolami I et al Ki67/MART1 and p63/SOX10 dual immunohistochemistry allows a correct interpretation of the melanocytic component in the diagnosis of pigmented pilomatricoma Indian J Dermatol 2021665259 10.4103/ijd.ijd_137_21
Travaglino A Raffone A Russo D Guadagno E Pignatiello S Moretta P et al Does endometrial morular metaplasia represent odontogenic differentiation? Virchows Arch 202147960716 10.1007/s00428-021-03060-2
Julian CG Bowers PW A clinical review of 209 pilomatricomas J Am Acad Dermatol 1998391915 10.1016/S0190-9622(98)70073-8
DeRosa DC Lin-Hurtubise K Pilomatricoma: An unusual dermatologic neoplasm Hawaii J Med Public Health 2012712826
Dyson S Cassarino D Pilomatrixoma and pilomatrical carcinoma ExpertPath Available from: https://app.expertpath.com/document/pilomatrixoma-and-pilomatrical-car-/a75411a1-6792-437f-877d-2ff415c7f97e?searchTerm=pilomatricoma [Last accessed on 2023 Jan 04]
Kim DP Kus KJB Ruiz E Basal cell carcinoma review Hematol Oncol Clin North Am 2019331324 10.1016/j.hoc.2018.09.004
Marzuka AG Book SE Basal cell carcinoma: Pathogenesis, epidemiology, clinical features, diagnosis, histopathology, and management Yale J Biol Med 20158816779
He P Cui LG Wang JR Zhao B Chen W Xu Y Trichilemmal cyst: Clinical and sonographic features J Ultrasound Med 201938916 10.1002/jum.14666
Phukan JP Sinha A Pal S Sinha R Cytological diagnosis of epidermal inclusion cyst of breast: A rare benign lesion J Nat Sci Biol Med 201454602 10.4103/0976-9668.136251
Wakely PE Jr Atypical spindle cell/pleomorphic lipomatous tumour (ASPLT): A report of three FNA cases and comparison with spindle cell/pleomorphic lipoma cytopathology Cytopathology 20233434652 10.1111/cyt.13227
Khairwa A Dey P Bal A Fine-needle aspiration cytology of recurrent Merkel cell carcinoma of eye-brow J Cytol 20143117980 10.4103/0970-9371.145666
Epidermal inclusion cyst Libre Pathology https://librepathology.org/wiki/Epidermal_inclusion_cyst [Last accessed on 2023 May 06]


An unusual neck mass diagnosed by fine needle aspiration: Cytological findings and challenges

QUIZ DESCRIPTION

A 60-year-old woman with a left neck mass was referred for an ultrasound-guided fine needle aspiration (FNA). Ultrasound examination revealed a 2.7 × 2.2 × 1.2 cm, well-circumscribed, solid, hypoechoic lesion with smooth sharp borders in the right neck at level 2A. During the physical examination and interview, the patient reported to have an undiagnosed palate mass. The left neck mass was targeted for FNA. The Diff-Quik stain of the smear showed clusters of papillary-like epithelioid cells with round-to-oval nuclei with wrinkled membranes and extracellular metachromatic stromal fragments. The Papanicolaou stain showed cells with nuclear grooves, small nucleoli, and fine vesicular chromatin. The cells had a moderate amount of delicate cytoplasm, focally intermingled with scant stromal components. The cell block also showed clusters of papillary structures with a cribriform-like architecture [Figure 1a-d].

(a) Hypercellular smears consisting mostly of cohesive cellular aggregates with scattered single cells in the background. Metachromatic stromal fragments are present (Diff-Quik stain, ×10 objective). (b and c) Papillary clusters of neoplastic cells with slightly irregular small to mediumsized nuclei showing pale chromatin and intranuclear grooves (Papanicolaou stain, ×10 and ×60 objectives). (d) Hematoxylin and eosin stain of a cell-block section showing papillary-like structures with lumens consisting of small to medium-sized cells with pale nuclei (×40 objective).

MORPHOLOGY QUIZ

Q1. What is the most likely diagnosis?

Papillary thyroid carcinoma

Salivary gland myoepithelioma

Polymorphous adenocarcinoma (PAC)

Pleomorphic adenoma

Basal cell adenoma.

Answer: C

EXPLANATION

Slightly irregular nuclei with grooves similar to those seen in papillary carcinoma of the thyroid gland are present; however, intranuclear inclusions are absent. The cytological findings combined with clinical information regarding the presence of a mass in the palate are most consistent with metastatic PAC. Pleomorphic adenoma, basal cell adenoma, and myoepithelioma are benign tumors and do not metastasize to the neck lymph nodes.[1]

Q2. Which immunohistochemical panel and findings would be best to confirm the diagnosis of PAC?

Thyroid transcription factor-1 (TTF-1) positive, S100 positive, cytokeratin-7 (CK-7) positive

TTF-1 negative, CK-7 positive, S100 positive

CK-7 negative, paired-box gene 8 (PAX 8) positive, S100 negative

p40 positive, p63 positive, S100 negative.

Answer: B

Immunohistochemistry panel was performed on the cell block which revealed these cells to be positive for CK-7, S100, and Sry-related HMg-Box 10 (SOX10) gene and negative for p40, p63, smooth muscle actin, TTF-1, thyroglobulin, and PAX8 [Figure 2]. Negative TTF-1, thyroglobulin, and PAX8 ruled out metastatic papillary thyroid carcinoma. In the setting of the patient’s palate mass, PAC was considered in this FNA.

(a) Cytokeratin-7 showing cytoplasmic stain (×40 objectives). (b) S100 demonstrates nuclear and cytoplasmic staining of neoplastic cells (×40 objectives). (c) SOX10 shows nuclear staining of the tumor cells (×40 objectives). (d) Thyroid transcription factor-1 shows negative nuclear stain (×40 objectives).

EXPLANATION

PAC is immunoreactive for CKs including CK7, in 100% of cases and S100 protein 97–100%. p63 is positive in 78–100% of cases, whereas p40 is usually negative.[1] SOX10 is positive in 86% of PACs.[2]

There is some degree of overlap in the immunohistochemical profile of secretory carcinoma of the salivary glands and PACs. Secretory carcinomas are positive for CK7, S100, SOX10, vimentin, and mammaglobin, and negative for p63, p40, NR4A3, and discovered on GIST-1.[1] However, the cytomorphology of secretory carcinomas varies in that they are composed of microcystic/solid, tubular, and papillary-cystic structures with luminal secretions. The tumor cells in secretory carcinomas have vacuolated pink cytoplasm and granular chromatin.[1]

Q3. Which one of the following is correct about PAC?

PAC is the most common intraoral salivary gland tumor

PAC commonly metastasizes to the neck lymph nodes

PAC has an overall good prognosis

The most common location of PAC is the parotid gland.

Answer: C.

EXPLANATION

PAC is the second most common intraoral salivary gland carcinoma, which rarely metastasizes to the neck lymph nodes. Pleomorphic adenoma is the most common benign tumor and mucoepidermoid carcinoma is the most common malignant tumor of the minor salivary glands of the oral cavity.[3] The palate is the most common site for PAC and the overall prognosis of PAC is excellent with a 10-year survival rate of 94–99%.[1] The main malignant salivary gland tumors that can metastasize to the neck lymph nodes are mucoepidermoid carcinoma, adenoid cystic carcinoma, acinic cell carcinoma, secretory carcinoma, salivary duct carcinoma, myoepithelial carcinoma, epithelial-myoepithelial carcinoma, hyalinizing clear cell carcinoma, carcinoma ex pleomorphic adenoma, and lymphoepithelial carcinoma.[4]

Q4. What chromosomal abnormality or molecular findings are seen in PAC?

ETS Variant Transcription Factor 6 (ETV6)-Neurotrophic Tyrosine Kinase Receptor Type 3 (NTRK3) fusion

Myelobastosis viral oncogene homolog (MYB) and nuclear factor I/B (NFIB) and MYBL1-NFIB fusions

Mutations in TP53 (55%), Harvey Rat sarcoma virus (HRAS) (23%), Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) (23%)

Protein kinase D (PRKD)1, PRKD2, or PRKD3 fusions.

Answer: D.

EXPLANATION

Most cases of secretory carcinomas show chromosomal translocation, t (12; 15) (p13; q25) resulting in an ETV6-NTRK3 fusion. Adenoid cystic carcinomas have (6;9) or t (8;9) translocations, resulting in MYB-NFIB and MYBL1-NFIB fusions. Mutations in TP53 (55%), HRAS (23%), and PIK3CA (23%) are seen in salivary duct carcinoma. Conventional PAC has predominantly PRKD1 mutation and cribriform subtype has PRKD1,2,3 fusions. Cribriform subtypes are more likely metastasize to the neck lymph nodes.[1,5]

ADDITIONAL DETAILS, FOLLOW-UP

The patient underwent a right maxillectomy with ipsilateral neck dissection 5 months after the FNA procedure, which showed a 5.3 cm high-grade PAC with metastasis to the two lymph nodes. Microscopic examination of the specimen revealed an infiltrative tumor with papillary structures, fibrous connective tissue stroma, and open chromatin nuclei [Figure 3a-c].

(a) Hematoxylin and eosin (H&E) stain shows an infiltrative neoplasm (×4 objective). (b) The tumor had a prominent papillary architecture (H&E, ×10 objective). (c) The nuclei show open chromatin (H&E, ×40 objective).

BRIEF REVIEW OF THE TOPIC

PAC of the minor salivary glands is a rare head-and-neck cancer, which generally has a good prognosis. PACs were previously known as “polymorphous low-grade adenocarcinoma”. The recent World Health Organization classification of head-and-neck tumors has characterized it as PAC.[1,6]

There are two subtypes of this tumor, conventional subtype and cribriform subtype. The conventional subtype shows various patterns, including tubules, trabeculae, and microcysts with a mucoid/myxoid background. The cribriform subtype shows papillary structures and glomeruloid bodies in a fibrous stroma. Their nuclei show open chromatin.[1] Recognizing PAC can be challenging due to diverse architectural patterns.[1]

Due to the intraoral location of PAC and rare occasions of metastasizing to the neck lymph nodes, these tumors are hardly sampled by FNA, and therefore, cytopathologists have less experience with the diverse cytomorphology of PAC.[7]

Overall, cytology smears show uniform moderately sized cells with relatively scant cytoplasm and round-to-oval nuclei, forming papillary, tubular, and irregular solid clusters. Dense globular or fibrillary stroma can be seen. Therefore, PAC can resemble adenoid cystic carcinoma or epithelial-rich pleomorphic adenoma. Occasional nuclear grooves and pale chromatin are also seen in PAC and can be misinterpreted as papillary thyroid carcinoma.[1,7]

In our case, TTF-1 and thyroglobulin stains were performed to rule out metastatic papillary thyroid carcinoma.

PAC is immunoreactive for CKs and S100 protein. Staining for p63 is variable. p40 is typically negative. Other positive immunomarkers include mammaglobin (67–100%), CD117 (60%), carcinoembryonic antigen (54%), glial fibrillary acidic protein (15%), melanoma-specific antigen (13%), and epithelial membrane antigen (12%).[1]

Pleomorphic adenomas are immunoreactive with pleomorphic adenoma gene 1 and human high-mobility group protein A2. Cytologically, most of the time, pleomorphic adenomas show the characteristic fibrillary matrix, mixed with myoepithelial cells which merge into the epithelial component. The chromatin of the tumor cells is dark unlike PAC.[1]

Adenoid cystic carcinomas are basaloid tumors. The smears show cohesive clusters of cells forming sheets or show tubular, complex, or cribriform architecture containing hyaline globules. The tumor cells have hyperchromatic nuclei.[1]

Other differential diagnosis includes cribriform-morular thyroid carcinoma (CMTC) which is now considered a distinct malignant thyroid neoplasm. The smears of CMTCs are hypercellular showing papillary and cribriform clusters with slit-like spaces and swirling of nuclei, representing morulae. Nuclei can show peculiar nuclear clearing with thick membranes, grooves, and pseudoinclusions.[8]

Genetic alterations in PRKD 1/2/3 genes have been described in PACs and may be useful in distinguishing PAC from other salivary gland neoplasms.[5]

SUMMARY

In summary, the cytologic features of PAC overlap with other salivary gland neoplasms and papillary thyroid carcinoma. In our case, the immunohistochemical stains in addition to cytomorphology (pale chromatin and papillary-like structures) with the clinical information helped us reach a diagnosis of FNA.

ABBREVIATIONS

FNA – Fine needle aspiration

PAC – Polymorphous adenocarcinoma.


References

Michal M Leivo I Simpson RH Xu B Poorten VV Wasserman JK Polymorphous adenocarcinoma Skalova A Hyrcza MD Mehrotra R editors WHO classification of tumours: Head and neck tumours 5th ed 9 Lyon, France IARC 2022
Ko YC Varma S Zhu CF Zhu SX Vennam S Poh CF et al Gene expression profiling of head and neck tumors identifies FOXP1 and SOX10 expression as useful for distinguishing ameloblastoma from basaloid salivary gland tumors Am J Surg Pathol 20204466572 10.1097/PAS.0000000000001421 31895100
Waldron CA El-Mofty SK Gnepp DR Tumors of the intraoral minor salivary glands: A demographic and histologic study of 426 cases Oral Surg Oral Med Oral Pathol 19886632333 10.1016/0030-4220(88)90240-X 2845326
Stennert E Kisner D Jungehuelsing M Guntinas-Lichius O Schroder U Eckel HE et al High incidence of lymph node metastasis in major salivary gland cancer Arch Otolaryngol Head Neck Surg 20031297203 10.1001/archotol.129.7.720 12874071
Andreasen S Melchior LC Kiss K Bishop JA Hogdall E Grauslund M et al The PRKD1 E710D hotspot mutation is highly specific in separating polymorphous adenocarcinoma of the palate from adenoid cystic carcinoma and pleomorphic adenoma on FNA Cancer Cytopathol 201812627581 10.1002/cncy.21959 29266837
Vander Poorten V Triantafyllou A Skalova A Stenman G Bishop JA Hauben E et al Polymorphous adenocarcinoma of the salivary glands: Reappraisal and update Eur Arch Otorhinolaryngol 2018275168195 10.1007/s00405-018-4985-5 29761209
Jimenez-Heffernan JA Rodriguez-Garcia AM Gonzalez-Peramato P Lopez-Ferrer P Munoz-Hernandez P,Gordillo CH et al Fine needle aspiration cytology of polymorphous adenocarcinoma of the salivary glands: A report of 11 patients and review of the literature Diagn Cytopathol 202048101320 10.1002/dc.24473 32421929
Hirokawa M Maekawa M Kuma S Miyauchi A Cribriformmorular variant of papillary thyroid carcinoma--cytological and immunocytochemical findings of 18 cases Diagn Cytopathol 2010388906 10.1002/dc.21309 20091902


Challenge in the cytological interpretation of a not-so-typical breast carcinoma

A 52-year-old postmenopausal female presented with swelling in the right breast of size 4x3 cm. Consistency was hard and margins were ill-defined. The nipple was retracted and the skin over the swelling was fixed to it, showing ulceration and puckering. The tumor was fixed to the chest wall. A history of blood-mixed discharge from the ulcer was present. No axillary lymph nodes were palpable. Clinically the stage of the lesion was T4cN0M0, Stage IIIB. Ultrasonography showed a solid cystic lesion in the breast which was reported as BIRADS IV and two subcentimetric lymph nodes were present in the right axilla. Fine-needle aspiration cytology (FNAC) smears showed predominantly sheets and clusters of cells with abundant vacuolated cytoplasm, along with clusters of epithelial cells that showed abundant eosinophilic cytoplasm. The background showed acute and chronic inflammatory cells, occasional giant histiocytes, bare nuclei, and proteinaceous material. Biopsy showed two populations of cells with sharply defined cell borders, one with abundant eosinophilic, periodic acid-schiff (PAS) positive, diastase resistant, granular cytoplasm, and the other with abundant vacuolated cytoplasm. The cells showed marked pleomorphism, vesicular nuclei, prominent nucleoli with brisk mitotic activity, and atypical mitosis. Subsequently modified radical mastectomy specimen confirmed the infiltrative nature of the tumor. The tumor cells were arranged in papillary, micropapillary, acinar and the tubular patterns, and solid sheets. Extensive necrosis, stromal desmoplastic reaction, acute and chronic inflammatory cells, and vascular tumor emboli were also found. No ductal carcinoma in situ (DCIS) component was noted. Nottingham’s histologic score was 9 (Grade III). The skin over the swelling showed dermal infiltration by the tumor. The other resected margins were free of tumor. Eighteen axillary lymph nodes were harvested and two of them showed metastasis. The pathological stage was pT3N1aMx. On immunohistochemical evaluation, estrogen receptor (ER) (Clone: EP1) and progesterone receptor (PR) (Clone: EP2) were negative. Human epidermal growth factor receptor 2 (HER2) (Clone: EP3) showed diffuse strong (3+) membranous positivity in the tumor cells. Ki-67 (Clone: MIB-1) (proliferation index) was 46%. Cytokeratin (CK) 5/6/8/18 (Clone: 5D3/LP34) showed diffuse strong membranous positivity in the tumor cells. Androgen receptor (AR) (Clone: EP120) was positive in the apocrine cells.

QUESTION # 1

What is the most likely diagnosis?

Secretory carcinoma

Oncocytic carcinoma

Apocrine carcinoma

Lipid-rich carcinoma

ANSWER

c. Apocrine carcinoma

EXPLANATION

The patient presented with features of invasive carcinoma of the breast and with skin ulceration. Fine needle Aspiration cytology (FNAC) smears predominantly showed cells with abundant vacuolated cytoplasm [Figure 1]. Biopsy showed two populations of cells with well-defined cytoplasmic borders; one with abundant eosinophilic cytoplasm with PAS-positive diastase-resistant granules, and the other with abundant vacuolated cytoplasm. The cells showed nuclear atypia, brisk mitotic activity, and atypical mitosis [Figure 2]. Two types of cells are seen in apocrine carcinoma. Type A cells have abundant eosinophilic granular cytoplasm, enlarged nuclei and the prominent nucleoli, and type B cells have abundant vacuolated cytoplasm with intracytoplasmic lipids. The type A cells are diastase-resistant and, the PAS-positive.[1-7]

Microphotographs of the cytology smears (May GrunwaldGiemsa [MGG]): (a and b) (MGG, ×200), Fine-needle aspiration cytology (FNAC) smears showing cells with vacuolated cytoplasm, (c) (MGG, ×200) FNAC smear showing benign ductal epithelial cells with apocrine metaplasia along with the foamy cells, (d and e) (MGG, ×400) Fine-needle aspiration cytology (FNAC) smears showing cells with vacuolated cytoplasm, (f) (MGG, ×1000) Cells with well-defined cytoplasmic borders.

Gross and histopathology of the lesion: (a) Gross image of the modified radical mastectomy specimen, (b) Cut section of the tumor, (c) Histological section of the tumor (hematoxylin & eosin [H & E], ×200), and (d) Section showing the cells with apocrine differentiation (H & E, ×400).

The diagnostic interpretation of apocrine carcinoma on cytology smears may be challenging due to its morphologic mimics. FNAC smears show large polygonal cells with abundant granular cytoplasm and sharply defined borders. The nuclei are vesicular, with irregular nuclear bor ders, and show prominent nucleoli.[8] Predominance of type B cells in cytology smears poses difficulty in arriving at the diagnosis. This might be due to sampling error. In 2005, Japaze et al.,[6] proposed the following criteria for diagnosing apocrine carcinoma: (i) 75% of tumor cells exhibiting apocrine features, (ii) large cells with granular eosinophilic cytoplasm, (iii) sharply defined cell borders, (iv) large, round, vesicular, may be the pleomorphic nucleus, and (v) low N: C ratio (≤1:2).[6]

QUESTION # 2

Which of the following immunohistochemical marker is generally negative in apocrine carcinoma of breast?

ER

AR

HER2

GCDFP-15

ANSWER

a. ER

EXPLANATION

Apocrine carcinomas lack ER and PRs, Bcl-2, but have AR and express gross cystic disease fluid protein-15 (GCDFP-15), GATA binding protein 3, CK7, CK8, CK18, CK19, CK20, expression of MUC1 (EMA), and E-Cadherin. HER2/neu may or may not be positive. Basal cytokeratins such as CK5/6, CK14, CK17, and p63 are variably positive. GCDFP is present in the breast cysts and in apocrine cells of mammary glands, salivary glands, sweat glands, Paget disease, etc. HER2/neu is positive in 30–60% of carcinomas with apocrine differentiation. GCDFP-15 and AR are considered the hallmarks of apocrine differentiation [Figure 3]. The expression of GCDFP-15 appears to be reduced in advanced apocrine carcinoma. In oncocytic carcinoma , ER, PR, and anti-mitochondrial antibodies are positive and AR and GCDFP-15 are negative.[1-15] Hence, apocrine carcinomas have two molecular subtypes: Triple-negative and HER2-positive.[10]

Immunohistochemistry of the tumor: (a) Estrogen receptor (ER) is negative in tumor cells (ER and Diaminobenzidine [DAB], ×200), (b) Progesterone receptor (PR) is negative in tumor cells (PR and DAB, ×200), (c) Human epidermal growth factor receptor 2 (HER2)/neu shows diffuse, strong membranous positivity (Grade 3+) (HER2 and DAB, ×200), (d) Ki-67 (Proliferation marker) is positive in 46% of cells (Ki-67 and DAB, ×200), (e) Cytokeratin (CK5/6/8/18): Strong cytoplasmic to membranous positivity in >50% of the cells (CK5/6/8/18 and DAB, ×200), and (f) Androgen receptor is positive in the nuclei of cells with apocrine morphology (androgen receptor and DAB, ×200).

QUESTION # 3

To be designated as “Carcinoma with apocrine differentiation,” _________% of the tumor cells should have distinct apocrine morphology.

50%

25%

75%

90%

ANSWER

d. 90%

EXPLANATION

To be designated as “Carcinoma with apocrine differentiation,” the distinct apocrine morphology should be evident in >90% of the cancer cells. Previously, the cutoff percentage of tumor cells had been defined as 75% by Japaze et al., to be diagnosed as apocrine carcinoma [1-4,9-12]

QUESTION # 4

Is apocrine carcinoma associated with BRCA 1 or 2?

Yes

No

Not applicable

Cannot be commented.

ANSWER

b. No

EXPLANATION

Although loss of PTEN (Phosphatase and tensin homolog) function may indicate familial breast carcinoma, there is no association between apocrine carcinoma and BRCA1 or BRCA2.[5]

QUESTION # 5

Which of the following breast carcinomas has a worse prognosis?

Triple-negative invasive breast carcinoma of no special type

Triple-negative invasive apocrine carcinoma

Luminal A type breast carcinoma

Luminal B type breast carcinoma

ANSWER

a. Triple-negative invasive duct carcinoma of no special type

EXPLANATION

The triple-negative subtype of apocrine carcinoma of the breast has better prognosis than triple-negative invasive breast carcinoma of no special type (NST), as targeted therapy with drugs used in prostate carcinoma such as fluoxymesterone that inhibits androgen signaling, is available.[1-6,11] The other two subtypes carry better prognosis.

BRIEF REVIEW OF THE TOPIC

Invasive carcinoma of breast with apocrine differentiation is a special subtype of breast carcinoma.[16] The age of presentation ranges from 48 to 60 years.[1] It is uncommon and the incidence ranges from 0.3% to 4% of female invasive breast carcinomas. It is seen more commonly in middle-aged women. Its incidence is extremely rare in male breasts. It is an aggressive malignancy. It can be misdiagnosed as invasive duct carcinoma. Lymph node metastasis has been reported in apocrine carcinomas. Malignant adnexal tumors of the skin may arise from apocrine, eccrine, sebaceous, ceruminous, and sweat glands. Normal breast ductal cells undergo metaplasia and transform into apocrine cells. The pattern of growth of apocrine carcinoma is like that of invasive duct carcinoma.[1-6,8-12]

Apocrine carcinoma was first described by Krompecher et al. in 1916. However, the histologic criteria to diagnose the apocrine carcinomas were defined by Japaze et al.[3,6]

The 5th edition of the World Health Organization (WHO) Classification of Breast Tumors recognized “Carcinoma with apocrine differentiation” as a distinct entity. Apocrine differentiation occurs in invasive carcinomas NST, lobular, tubular, medullary, and micropapillary carcinomas, DCIS, and lobular carcinoma in situ.[1,7]

Apocrine carcinoma arises from the milk duct of the breast. Grossly, it presents as a solidified whitish mass. It generally presents with skin ulceration. Nipple discharge may or may not be present. It has been reported in accessory breast also. The growth pattern is like that of invasive duct carcinoma. The cells of apocrine carcinoma have abundant eosinophilic granular cytoplasm, well-defined cytoplasmic borders, apical snouting at places, large round vesicular nuclei, and multiple nucleoli.

Differential diagnoses of carcinoma with apocrine differentiation include malignancies such as secretory carcinoma, oncocytic carcinoma, lipid-rich carcinoma, invasive duct carcinoma, and benign lesions such as granular cell tumor, apocrine metaplasia, and histiocytic proliferation [Figure 4].[8,17-27] Immunohistochemistry can help in cases where morphological interpretation alone cannot aid in differentiating between these cases [Table 1].[1-8,10,17-28]

Algorithm for the cytological approach to cells with vacuolated cytoplasm in breast aspirate. (PAS: periodic acid-schiff, H/O: History of.) Algorithm plotted using Microsoft PowerPoint [Microsoft Office Standard 2016, Version 16.0; Manufacturer: Microsoft Corporation, Origin: Silicon Valley, CA, USA]

Differentiation of breast carcinoma with cells having vacuolated cytoplasm by immunohistochemical evaluation of cell block sections.

Breast carcinoma subtype Cellblock IHC (Immunohistochemistry) profile
Apocrine Ca ER−, PR−, HER2±, AR+, GCDFP+, GATA-3+, Mammaglobin+, CK7+, CK8+, CK18+, CK19+, CK20+, EMA+, E-cadherin+, Bcl-2−
Sebaceous Ca ER±, PR±, HER2+, AR+
Secretory Ca ER−, PR−, HER2−, EMA+, α-Lactalbumin+, S100+, E-cadherin+, CK8+, CK18+, CD117+, α-SMA+, Mammaglobin+, GCDFP+, SOX10+, Pan-TRK
Papillary Ca ER+, PR+, HER2−, CK8+, CK18+ CK5/6−, CK14−
Mucinous Ca ER+, PR+, WT1+, AR±, HER2−

IHC: Immunohistochemistry, ER: Estrogen receptor, PR: Progesterone receptor, GCDFP 15: Gross cystic disease fluid protein 15, GATA-3: GATA-binding protein 3, AR: Androgen receptor, HER2: Human epidermal growth factor receptor 2, CK: Cytokeratin, EMA: Expression of MUC1

Focal apocrine changes can be seen in several breast lesions ranging from benign cysts to invasive carcinomas. Benign breast lesions with apocrine morphology include fibrocystic disease, apocrine cysts, the apocrine adenosis, and apocrine adenoma. Malignant lesions with apocrine morphology include apocrine DCIS and apocrine carcinoma. Hence, diagnosis of apocrine carcinoma can be challenging. Overlapping of cells, nuclear pleomorphism, and high N: C ratio are not usually seen in benign lesions with apocrine differentiation.[5,8]

Cytogenetic analysis of apocrine carcinoma cells reveals gains of 1p, 1q, 2q, 7 and 17, losses of 1p, 12q, 16q, 17q, and 22q.[5]

Electron microscopy of the apocrine carcinoma cells shows abundant cytoplasm with well-defined outlines, membrane-bound electron-dense granules in the cytoplasm, abundant Golgi apparatus, mitochondria with incomplete cristae and perinuclear condensation, and empty vesicles. In oncocytic carcinoma, numerous mitochondria are seen occupying >60% of the cytoplasm and they are seen to be dispersed, in contrast to apocrine carcinoma.[4,8]

In the gene expression studies, these tumors express luminal cytokeratins (AMACR) and lack basal features. Hence, these tumors are called “Luminal Androgen Receptor” (LAR) tumors. A small proportion of cases may show a basal phenotype. AMACR is positive in 97% of invasive carcinomas with apocrine differentiation, the 96% of apocrine DCIS, but only in 22% of carcinomas without apocrine differentiation.[10]

Next-generation sequencing frequently shows loss of PIK3CA/PTEN/AKT and TP53, followed by mutations of KRAS, NRAS, and BRAF.[10,13]

Genetic studies on the AR gene showed the highest CAG repeats in DCIS with apocrine differentiation compared to fibroadenomas and invasive breast carcinomas.[10] Immunohistochemical and molecular features do not necessarily correlate in all cases of carcinomas with apocrine differentiation.[10]

The expression of PD-L1 is low in apocrine carcinoma. They are found to be microsatellite stable.[10] Expression of 5a-reductase is found in 60% of apocrine carcinomas, and it correlates with poor prognosis in terms of invasion of lymphatics, blood vessels, and higher histologic grade. Gamma-glutamyl transferase 1 and tumor-associated glycoprotein-72 are specific markers of apocrine differentiation.[5]

The unavailability of molecular and genetic studies due to financial constraints in the setting of developing countries may pose the challenge in diagnosing apocrine carcinoma.[3]

SUMMARY

Apocrine carcinoma is an aggressive malignancy that tends to ulcerate the skin, and metastasize to lymph nodes. It is of two types – triple-negative and HER2-positive. Even in triple-negative cases, targeted therapy with anti-androgen is available. Hence, making a correct diagnosis of the tumor is necessary. Whenever vacuolated cells are encountered in the cytology smears, especially in an elderly female/male, it is prudent to sample the lesion thoroughly so that making an inappropriate diagnosis can be avoided.

AVAILABILITY OF DATA AND MATERIALS

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

ABBREVIATIONS (In alphabetic order)

AMACR – Alpha-methyl acyl-CoA racemase

GATA3 – GATA binding protein 3

GCDFP 15 – Gross cystic disease fluid protein 15

AR – Androgen receptor

DCIS – Ductal carcinoma in situ

LCIS – Lobular carcinoma in situ

ER – Estrogen receptor

FNAC – Fine-needle aspiration cytology

H & E – Hematoxylin & Eosin

IHC – Immunohistochemistry

LAR – Luminal androgen receptor

MGG – May Grunwald-Giemsa

MRM – Modified radical mastectomy

PR – Progesterone receptor.


References

Jagtap SV Jagtap SS Phalke A Vartak S Billawaria S Apocrine carcinoma of breast-uncommon variant of breast malignancy Clin Pathol Res J 2021514 10.23880/CPRJ-16000135
Goel D Maurya MK Ramakant P Apocrine carcinoma of breast: A rare case report Indian J Pathol Microbiol 2022652213
Ismail S Kherbek H Skef J Zahlouk N Abdulal R Alshehabi Z Triple-negative apocrine carcinoma as a rare cause of a breast lump in a Syrian female: A case report and review of the literature BMC Womens Health 202121396 10.1186/s12905-021-01539-3 34823507
Muthusamy RK Mehta SS Giant apocrine carcinoma of the breast: A case report with review Asian J Oncol 20184214 10.4103/ASJO.ASJO_112_16
Vranic S Schmitt F Sapino A Costa JL Reddy S Castro M et al Apocrine carcinoma of the breast: A comprehensive review Histol Histopathol 2013281393409
Wader JV Jain A Bhosale SJ Chougale PG Kumbhar SS Apocrine carcinoma of breast: A case report with review of the literature Case Rep Pathol 20132013e170918 10.1155/2013/170918 23864975
Allison KH Brogi E Ellis IO Fox SB Lakhani SR Lax SF et al Epithelial tumours of the breast Dilani Lokuhetty Valerie A. White Reiko Watanabe Ian A. Cree WHO classification of tumours: Breast tumours 5th ed 2 France IARC/WHO Committee 20199161
Khandeparkar SG Deshmukh SD Bhayekar PD A rare case of apocrine carcinoma of the breast: Cytopathological and immunohistopathological study J Cytol 201431968 10.4103/0970-9371.138679 25210239
Adiputra PA Sudarsa IW Irawan H Saputra H Malignant adnexal tumor of the skin on breast: A case report of apocrine carcinoma Int J Surg Case Rep 2023108108383 10.1016/j.ijscr.2023.108383 37327766
Vranic S Gatalica Z An update on the molecular and clinical characteristics of apocrine carcinoma of the breast Clin Breast Cancer 202222e57685 10.1016/j.clbc.2021.12.009 35027319
Touimi SH N'chiepo D Mbarki I Elkacemi H Elmajjaoui S Kebdani T et al Apocrine carcinoma of breast in a male patient: Case report Tumori J 2020106123 10.1177/0300891620914132
Fujiwara-Tani R Hashizume J Hanaoka K Kuniyasu H Axillary carcinoma with apocrine differentiation: A case report Clin Diagn Pathol 2018213 10.15761/CDP.1000127
Vranic S Feldman R Gatalica Z Apocrine carcinoma of the breast: A brief update on the molecular features and targetable biomarkers Bosn J Basic Med Sci 201717911 10.17305/bjbms.2016.1811 28027454
Tsuchiya A Rokkaku Y Nihei M Nomizu T Abe R Apocrine carcinoma of the breast-a case report Jpn J Surg 1988187147 10.1007/BF02471535 3246780
Sinn HP Kreipe H A brief overview of the WHO classification of breast tumors, 4th edition, focusing on issues and updates from the 3rd edition Breast Care (Basel) 2013814954 10.1159/000350774 24415964
Tan PH Ellis I Allison K Brogi E Fox SB Lakhani S et al The 2019 World Health Organization classification of tumours of the breast Histopathology 2020771815 10.1111/his.14091 32056259
Singh TH Rathod GB Diagnosis of fat necrosis on FNAC-A case report Int Arch Integr Med 201522369
Jyoti K Preeti B Sandip S Archana P Manjushree G Suresh M Comparison of fine needle aspiration cytology of non-neoplastic lesions of breast with histopathology Sch J Appl Med Sci 2013180413
Nikumbh DB Ningurkar NU Gondane SR Dravid NV Cytological diagnosis of ductal papilloma: An unusual entity in male breast IP Arch Cytol Histopathol Res 201613941
Aggarwal D Soin N Kalita D Pant L Kudesia M Singh S Cytodiagnosis of papillary carcinoma of the breast: Report of a case with histological correlation J Cytol 20143111921 10.4103/0970-9371.138694 25210247
Singla T Singla G Singla S Singh M FNAC diagnosis of crystallizing Galactocele-an unusual presentation Journal of Cytology 20203714950 10.4103/JOC.JOC_18_20 33088035
Javadzadeh B Finley J Williams HJ Fine needle aspiration cytology of mammary duct ectasia: Report of a case with novel cytologic and immunocytochemical findings Acta Cytol 200145102731 10.1159/000328349 11726098
Mendoza P Lacambra M Tan PH Tse GM Fine needle aspiration cytology of the breast: The nonmalignant categories Patholog Res Int 20112011547580 10.4061/2011/547580 21660275
Chikkannaiah P Thangngeo D Guruprasad C Venkataramanappa S Clinicopathological study of mucinous carcinoma of breast with emphasis on cytological features: A study at tertiary care teaching hospital of South India J Lab Physicians 2020126875 10.1055/s-0040-1714935 32792796
Miesbauerová M Tommola S Šteiner P Banecková M Skálová A Kholová I Cytopathological features of secretory carcinoma of salivary glands and ancillary techniques in its diagnostics: Impact of new Milan system for reporting salivary gland cytopathology APMIS 2019127491502 10.1111/apm.12950 30942913
Jung MJ Kim SY Nam SY Roh JL Choi SH Lee JH et al Aspiration cytology of mammary analogue secretory carcinoma of the salivary gland Diagn Cytopathol 20154328793 10.1002/dc.23208 25087572
Jo VY Krane JF Ancillary testing in salivary gland cytology: A practical guide Cancer Cytopathol 2018126 Suppl 862742 10.1002/cncy.22010 30156767
Cimino-Mathews A Novel uses of immunohistochemistry in breast pathology: Interpretation and pitfalls Mod Pathol 2021346277 10.1038/s41379-020-00697-3 33110239


Squash surprise in an elderly female

QUIZ DESCRIPTION

A 57-year-old female presented with nausea, vomiting, and headache over a year. The patient had no specific complaints except for gradual weight loss over the past year. Computed tomography scan (CT) was done. T2-weighted sagittal images show an ill-defined T2 hyperintense cystic space occupying a lesion in the occipital lobe. There is ipsilateral compression of the occipital horn of the ventricle and disproportionate perilesional edema with midline shift [Figure 1a]. A craniotomy was performed on the suspicion of a glioma. Intraoperatively, the tumor was yellowish white non-vascular, and soft surrounded by viscous yellow fluid. The bits of tumor tissue sent for squash diagnosis were yellow, firm, and difficult to squash and spread. Rapid hematoxylin and eosin stain was performed.

(a) Computed tomography (CT) image showing ill defined T2 hyperintense cystic space occupying lesion in the occipital lobe. (b-c) Squash cytology smears showing cohesive clusters of hyperchromatic tumor cells against a thin mucoidy background and foci of calcification(blue arrow). (b, c: Hematoxylin and Eosin H&E, 200x and 40x.).

Definitive sections were sent for histopathology and immunohistochemical staining was done.

MORPHOLOGY QUIZ

What is the diagnosis based on the clinical history, imaging findings, and photomicrographs [Figure 1] provided?

Glioma

Meningioma

Metastatic carcinoma

Lymphoma

Which of the following immunohistochemical panels would be most useful for diagnosis?

CK7, CK20, TTF-1, GATA3, PAX-8, CDX2

CK7, CK20, Calretinin, WT1

IDH1/2, ATRX, P53

GFAP, Vimentin, NeuN

Answers

1. (c) Metastatic carcinoma

2. (a) CK7, CK20, TTF-1, GATA3, PAX-8, and CDX2

EXPLANATION

Squash smear cytology is a rapid and reliable method for intraoperative diagnosis of inflammatory and neoplastic brain lesions.[1]

Squash smears were moderately cellular and showed cohesive clusters and glandular arrangement of tumor cells against a thin mucoid background [Figure 1b-c]. The tumor cells had round to oval hyperchromatic nuclei and a moderate amount of cytoplasm. A focus shows calcification. The findings were suggestive of a non-glial lesion, possibly metastases. Histopathology sections showed well-formed glands and sheets of tumor cells with pools of mucin and psammoma bodies confirming the diagnosis of metastatic adenocarcinoma [Figure 2a-d].

Microphotograph panel (a-d) showing well-formed glands and solid sheets of tumor cells with pools of mucin and focal calcification (hematoxylin and eosin stain) (a: 40x, b: 100x, c: 200x, d: 400x), (e) Tumor sections showing immunopositivity for CK7 200x (f) Tumor sections showing immunopositivity for CK20 [IPOX] 200x.

The most common tumors leading to brain metastases include lung and breast followed by renal tumors. In the present case, the tumor cells showed strong immunopositivity for CK7 and were immunonegative for CK20 [Figure 2e and f]. In a female patient, CK7-positive and CK20-negative adenocarcinomas warrant the search for primary in the lung, breast followed by the uterus (endometrial), pancreas, and gallbladder.

ADDITIONAL QUIZ QUESTIONS

3. Which of the following is the most common intracranial tumor?

Gliomas

Metastases

Meningioma

Lymphoma

4. Which of the following is a differential diagnosis of metastases to the brain?

Abscess

Parasitic infestation

Primary tumor-glioma/ependymoma

All of the above

Answers

3. (b) Metastas es

4. (d) All of the above

EXPLANATION

The differential diagnosis of metastases to the brain includes primary tumors of the brain, parasites, and abscesses.

Squash cytology smears of glial tumors are cellular with specific cytomorphological features of the tumor cells. For instance, the squash smears of low-grade gliomas show sparse cellularity of tumor cells in a fibrillary background in contrast to high-grade gliomas which are characterized by hypercellularity, endothelial proliferation, necrosis, and mitoses. Necrosis is also often noted in cases of brain abscess, characteristically described as liquefactive necrosis. Fungal, tubercular, and parasitic infestation can also show necrotic background.

BRIEF REVIEW OF TOPIC

Metastases to the brain are a known complication in approximately 20% of malignancies.[2] Brain metastases are often detected in CT and magnetic resonance imaging scans and previously known cases of malignancy during routine investigations. In most cases, metastases travel through the hematogenous route from lung tumors, breast carcinomas, renal cell carcinomas, and melanomas by breaching the blood–brain barrier.[3] Metastases are usually noted as intra-axial, intraparenchymal masses with a varied range of radiological differential diagnoses including gliomas, abscesses, and infections. Furthermore, in approximately 15% of cases, a primary tumor is not found.[4] In the index case, in the absence of any specific complaints elsewhere and a mass lesion in the brain, the suspicion of metastases was unlikely on clinical and radiological examination.

These cases are challenging and the surgeon relies on intraoperative diagnosis for immediate surgical management. In such scenarios, squash cytology can be reliably used to differentiate between metastatic from primary central nervous system neoplasms.

The squash smear cytomorphology depends on the type of primary tumor (adenocarcinoma, small cell carcinomas, melanoma, lymphoma, etc.). An algorithmic approach and workup with immunohistochemical markers can be useful for the detection of the primary tumor.

Immunohistochemical panel for morphologically designated adenocarcinoma of unknown primary involves the cytokeratin status (CK7 and CK20). Further organ-specific markers can be tested after analysis of CK7 and CK20 expression.[5]

In the index case, CK7 immunopositivity was found; however, the patient was lost to follow-up and further markers were not done.

Extensive radiological examination and accurate subtyping of the primary tumor enable the pathologist to choose the apt immunohistochemistry marker panel and document the origin of the tumor.

SUMMARY

The awareness of metastatic carcinoma as a differential diagnosis on cytology and identification of cytomorphological features can guide the surgeon and management of the patient.


References

Krishnani N Kumari N Behari S Rana C Gupta P Intraoperative squash cytology: accuracy and impact on immediate surgical management of central nervous system tumors: Impact of squash cytology on immediate surgical management Cytopathology 2012 2330814 10.1111/j.1365-2303.2011.00905.x 21838720
Sacks P Rahman M Epidemiology of brain metastases Neurosurg Clin N Am 2020314818 10.1016/j.nec.2020.06.001 32921345
Tyagi A Wu SY Watabe K Metabolism in the progression and metastasis of brain tumors Cancer Lett 2022539215713 10.1016/j.canlet.2022.215713 35513201
Balestrino R Rudà R Soffietti R Brain metastasis from unknown primary tumour: Moving from old retrospective studies to clinical trials on targeted agents Cancers (Basel) 2020123350 10.3390/cancers12113350 33198246
Varadhachary GR Carcinoma of unknown primary origin Gastrointest Cancer Res 2007122935


Atypical epithelioid cells in pleural effusion as foreign second population: A diagnostic cytopathology dilemma

A 56-year-old female presented with a right pleural effusion. The chest computed tomography (CT) showed a round mass shadow of the right middle and lower lobe of the lung. The pleural fluid showed atypical epithelioid cells [Figure 1].

(a) Pleural effusion smear showed many atypical epithelioid cells as second foreign population (high magnification of hematoxylin eosin (Hematoxylin and Eosin 200×)). (b) The embedded pleural fluid section showed that the tumor cells were scattered or clustered (medium magnification of Hematoxylin and Eosin 200×). (c) The embedded pleural fluid section showed significant atypia, nuclear disorientation and nucleolus (high magnification of Hematoxylin and Eosin 400×).

Q1. What is your interpretation of the pleural effusion smear?

Proliferated mesothelial cells

Mesothelioma

Metastatic adenocarcinoma

Malignant cells, pending immune characterization

ANSWER

The correct cytological interpretation is d.

EXPLANATION

A diagnosis of the pleural effusion cell block is a “malignant tumor.” The pleural effusion smear cells block showed tumor cells epithelioid, with significant atypical, prominent nucleoli and giant tumor cells, and nuclear deviation. In this case, the touch prep shows variably sized loose clusters and single cells with acinar formation. The cells show abundant eosinophilic cytoplasm. The nuclei have moderate-to-severe heteroplasia with occasional prominent nucleoli. At the time, “malignant tumor” was the most appropriate diagnosis. The differential diagnosis would include suspicion for carcinoma and other sarcomas.

ADDITIONAL QUIZ QUESTIONS

Q2. Hematoxylin-eosin(HE) stained sections are prepared from the tissue cores [Figure 2] and show a solid, cellular neoplasm. What is your interpretation?

Non-small cell lung cancer (NSCLC)

Epithelioid Sarcoma

Sarcomatoid carcinoma

Metastatic epithelioid hemangioendothelioma (EHE)

(a) Tumor cells were seen in the fibrous stroma as epithelioid, with the low magnification of Hematoxylin and Eosin, 40×. (b) The tumor cells were arranged in two ways. The glandular and the solid area are patchy, with a medium magnification of Hematoxylin and Eosin, 100×. (c) A small amount of red blood cells can be seen in the vascular lumen. The cells had significant atypia and high magnification of Hematoxylin and Eosin, 200×. (d) The cells in the solid area were epithelioid or short spindle-shaped, with nuclear deviation, with high magnification of Hematoxylin and Eosin, 400×.

ANSWER

The correct cytological interpretation is d.

Immunohistochemistry and molecular results

Immunohistochemical (IHC) staining was performed by the envisioned method. The results of IHC staining were as follows:

Tumor cells did not express CKpan, TTF1, P40, GATA3, CD10, calretinin, smooth muscle actin, or desmin; tumor cells expressed vascular markers such as CAMTA1, CD31, and ERG [Figure 3a-c], and vimentin was positive; the positive index of Ki-67 was approximately 5%. T (1p36) (CAMTA1) gene translocation (+) was detected by fluorescence in situ hybridization (FISH) (Note: red and green separation signals can be seen in > 50% of tumor cells) [Figure 3d].

(a) Immunohistochemical (IHC) CAMTA1 test showed that the tumor nucleus was positive, amplified by the envision method, 100×. (b) IHC CD31 test showed that the tumor nucleus was positive, amplified by the envision method, 100×. (c) IHC ERG test showed that the tumor nucleus was positive, amplified by the envision method, 100×. (d) Fluorescence in situ hybridization (FISH) detection showed that WWTR1-CAMTA1 fusion signal appeared in more than 50% of cells-the FISH method ×1000 (400×).

EXPLANATION FOR ADDITIONAL QUIZ QUESTIONS

Microscopic appearance: Tumor cells in the biopsy tissue were arranged in two ways: Adenoid structural area and solid area. A small amount of red blood cells could be seen.

In the cavity of the adenoid area, epithelioid tumor cells, conspicuous small nucleoli could be seen, and tumor cells had significant atypia. Short spindle cells could be seen in the solid area, some nuclei were large and biased. Moreover, some cells had cytoplasmic vacuoles, and a single red blood cell could be seen in individual cytoplasmic vacuoles.

No clear myxochondroid or no clear myxochondroid or hyaline matrix was found. IHC staining was as follows: Tumor cells did not express CKpan, TTF1, P40, and so on; so, epithelial cancer was not considered. Tumor cells expressed vascular markers such as CD31, and ERG positive; so, EHE specific antibody CAMTA1 was added and positive. T (1p36) (CAMTA1) gene translocation (+) was detected by FISH (Note: Red and green separation signals can be seen in >50% of tumor cells). Hence, we considered it as EHE, angiosarcoma differentiated in some areas with blood lakes.

The final diagnosis was EHE of the lung with angiosarcoma-like components.

DISCUSSION

EHE is a rare low-grade malignant angiogenic tumor that is most common in the lung and liver, always with multiple nodules.

EHE was first proposed by Weiss and Enzinger[1] in 1982 and was easily misdiagnosed morphologically as cancer. Its biological behavior was between epithelioid hemangioma and epithelioid angiosarcoma, which was defined as intermediate tumors. Along with the in-depth understanding of the disease, more studies[2,3] found that the tumor had a relatively high local recurrence rate (10–15%), metastasis rate (20–30%), and mortality (10–20%), which far exceeded the definition of rare recurrence and rare metastasis of intermediate tumors. Therefore, EHE was classified as a low-grade malignant tumor.[4] EHE often occurs in the lung and, or liver, typically as multiple nodules; however, it could occur in any body part, including bone, soft tissue, and heart.[5]

Morphologically, mucinous cartilage-like matrix or hyaline degeneration, the tumor cells were arranged in a cord or nest shape. The tumor cells were relatively uniform in size, round, and oval. The nuclear chromatin was consistent, and the nucleolus was not prominent. The cytoplasm was light eosinophilic, or the nucleus was biased. Moreover, vacuoles could be seen in some cytoplasm containing single or multiple red blood cells, which are called vesicular cells. It suggested the formation of a vascular lumen. Nuclear mitosis and necrosis were rare in classic EHE tumors.

Many studies have reported the morphological characteristics and diagnostic criteria of atypical EHE. The morphological criteria of atypical EHE vary in different studies, including tumor size, necrosis, nuclear grade, and threshold of the mitotic count. Anderson et al. reported a clinicopathological study of 52 cases of thoracic epithelioid malignant vascular tumors.[6] This showed epithelioid vascular tumors involving the chest divided into low-grade EHE, moderate EHE, and highly malignant epithelioid angiosarcoma with 4-year survival rates of 83%, 22%, and 9%, respectively. Studies have shown that the prognosis of moderate EHE was significantly worse than that of low-grade EHEs. In addition, survival analysis also showed that pleural involvement was a poor prognostic indicator. Similarly, the EHE of the pleura previously reported in our research group had a poor prognosis.[7] This patient had pleural involvement and pleural effusion. The examination report showed that the prognosis was poor. Deyrup et al.[2] performed a morphological risk assessment on the biological behavior of 49 cases of soft-tissue EHE. The risk model showed that a maximum tumor diameter >3 cm and mitosis >3/50 HPF were significantly correlated with poor patient prognosis. Therefore, the EHE could be divided into a high-risk group and a low-risk group. Shibayama et al.[3] scored 61 patients with EHE based on tumor size (i30 vs. >30 mm) and histological characteristics (typical vs. atypical). Survival analysis showed that the 5-year overall survival rates of the low (24 cases), medium (28 cases), and high (nine cases) risk groups were 100%, 81.8%, and 16.9%, respectively. Gong et al.[8] discussed the clinicopathological features of EHE, diagnosis, and differential diagnosis of eight cases of atypical EHE. They found that there were high-grade nuclei, active mitosis, solid flake growth mode, and tumor necrosis in the morphology of atypical EHE, and the biological behavior was more invasive. The follow-up results showed that there were six cases of metastasis, of which three cases died, and the prognosis was worse than that of classical EHE. Certain lineage changes in the morphology of EHE are significantly related to the prediction of patients. Therefore, how to accurately grade and guide the clinical treatment of EHEs will be the direction of future research.

EHE tumor cells express the vascular markers CD31, CD34, ERG, and FLI1. Cytokeratin can be expressed in 25% ~ 30% of cases, especially in biopsy specimens, which may be misdiagnosed as poorly differentiated carcinoma.[5] In addition, EHE highly expresses CAMTA1. EHE with vascular lacuna generally does not express CAMTA1 but is TFE3 positive. However, it should be noted that TFE3 immunohistochemistry is not completely specific. Both CAMTA1 and TFE3 were nuclear positive in this case, but CAMTA1 gene translocation was detected.

Atypical EHE and epithelioid angiosarcoma can cross and migrate morphologically. The application of IHC markers and the detection of specific fusion genes are very essential for differential diagnosis. Approximately 90% of EHE can produce t (1; 3) (p36.3; q23-25) ectopically, resulting in the WWTR1-CAMTA1 fusion gene, while about 5% of EHE contains the YAP1-TFE3 gene fusion. Studies have shown that EHE of ectopic fusion of the WWTR1 gene is rarely reported and mainly occurs in the heart.[3] The previous literature reported that epithelioid angiosarcoma could express the CAMTA1 marker, but the above fusion genes were not found.[6,8] Yang et al. compared the detection of CAMTA1 expression in cases of EHE and other vascular tumors using FISH and IHC. The sensitivity and specificity of IHC were 85.7% and 100%, respectively, whereas the sensitivity and specificity of FISH were both 100%.[9] Therefore, the detection of fusion genes is very important to distinguish EHE and epithelioid angiosarcoma.

DIFFERENTIAL DIAGNOSIS

Pulmonary EHE intersects with poorly differentiated adenocarcinoma, epithelioid angiosarcoma, and epithelioid hemangioma, which need to be differentiated according to microscopic morphology, IHC, and molecular detection.

Poorly differentiated adenocarcinoma

Epithelial cells or scattered vacuolar cells with nest-like arrangement in the lung EHE are easily misdiagnosed as poorly differentiated adenocarcinoma. In contrast to EHE, tumor cells have obvious atypia and obvious mitotic images. IHC shows CK pan positivity and vascular marker negativity. Therefore, if CK pan is negative in poorly differentiated tumors, it is suggested to add antigenic markers for identification.

Epithelioid angiosarcoma

In contrast to EHE, epithelioid angiosarcoma is more prone to interstitial hemorrhage, blood lake formation, papillary or fissure growth of tumor cells, obvious large nucleoli, active mitotic imaging, and so on. In addition to classic EHE, vascular lacunar-like structures were found in some areas, with significant atypia and mitotic images. However, both CAMTA1 IHC staining and FISH probes were positive. Therefore, this case was diagnosed as EHE with epithelioid angiosarcoma transformation in some areas.[10-13]

Epithelioid hemangioma

There are few cells in the nest of epithelioid hemangioma with vacuolar cytoplasmic vacuoles and red blood cells in the nest of epithelioid hemangioma, which suggests microvascular formation. Eosinophil infiltration can be seen around erythrocytes. These conditions are easily misdiagnosed as EHE. However, most EHE had no clear formation of vascular lacuna, the stromal was often mucinous cartilage-like, and most of them had no eosinophil infiltration.[14-20]

Epithelioid sarcoma

It often occurs in the limbs with the growth of tumor cells in multiple nodules under the microscope, and necrosis often occurs in the center of the nodule. In contrast to EHE, epithelioid sarcoma cells can express epithelial markers such as AE1/AE3 and epithelial membrane antigen (EMA), and the expression of INI1 is often absent.[21-24]

TREATMENT AND PROGNOSIS

EHE has limited experience in treatment, and more radical treatment may be needed. Therefore, surgical resection should ensure that the cutting edge is negative, supplemented by radiotherapy, chemotherapy, and targeted therapy if necessary.[25] This patient is currently undergoing the third PC chemotherapy regimen. EHE in the lung will indicate a poor prognosis if accompanied by pleural effusion or spindle tumor cell components. In this case, pleural effusion was obvious, and an angiosarcoma area appeared, which may be related to the poor prognosis of the patient.

A rare case of epithelioid hemangioendothelioma transforming from some areas to epithelioid angiosarcoma was discussed. Under a microscope, except for the classic EHE area, vascular lacuna formation was seen in some areas. Tumor cell atypia, mitotic imaging, and proliferative activity were significantly higher than those in the EHE area. IHC techniques and FISH detection confirmed that CAMTA1 gene translocation occurred in this case. Vascular lacunae and tumor cell atypia were found in some areas, suggesting that they may be transformed into epithelioid angiosarcoma.

A case of EHE with epithelioid angiosarcoma of the spine is reported in the literature,[26] but there was no genetic confirmation. From atypical morphology to final accurate diagnosis benefitting from immunohistochemistry and molecular diagnosis, we learned the development process of disease progression. We should not only recognize the classic morphology but also find clues regarding the classic morphology in atypical patients to provide a basis for accurate clinical diagnosis and treatment.

FOLLOW-UP AND PROGNOSIS

The patient received chemotherapy (albumin paclitaxel 420 mg/dL, carboplatin 680 mg/dL) combined with bevacizumab (400 mg/dL) injection intravenously on January 26, 2022. At present, the fourth course of treatment has been carried out. The current situation was stable.

SUMMARY

From atypical morphology to final accurate diagnosis benefitting from immunohistochemistry and molecular diagnosis, we learned the development process of disease progression. We should not only recognize the classic morphology but also find clues regarding the classic morphology in atypical patients to provide a basis for accurate clinical diagnosis and treatment.

ABBREVIATIONS

CT - Computed Tomography

EHE - Epithelioid Hemangioendothelioma

FISH - Fluorescence in situ Hybridization

HE - Hematoxylin-Eosin staining

IHC - Immunohistochemical

NSCLC - Non-small Cell Lung Cancer.

AUTHOR CONTRIBUTIONS

The manuscript was approved by all authors for publication. Substantial contributions to the conception, drafting the work and the acquisition, analysis of data for the work: S Z; Reviewing it critically for important intellectual content, and interpretation of data for the work: C W; Agreement to be accountable for all aspects of the work in ensuring that questions related to the accuracy, complete the Fish for the work, and interpretation of data for the work: W W;Design of the work, reviewing it critically for important intellectual content, and resolved any part of the work appropriately: L H. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

ETHICS APPROVAL AND CONSENT TO PARTICIPATE

The Institutional Review Board of Shanghai Pulmonary Hospital approved this retrospective study (IRB NO. K22-081Y).

Written informed consent was obtained from all the participants prior to the publication of this study.


References

Weiss SW Enzinger FM Epithelioid hemangioendothelioma: A vascular tumor often mistaken for a carcinoma Cancer 19825097081 10.1002/1097-0142(19820901)50:5<970::AID-CNCR2820500527>3.0.CO;2-Z 7093931
Deyrup AT Tighiouart M Montag AG Weiss SW Epithelioid hemangioendothelioma of soft tissue: A proposal for risk stratification based on 49 cases Am J Surg Pathol 2008329247 10.1097/PAS.0b013e31815bf8e6 18551749
Shibayama T Makise N Motoi T Mori T Hiraoka N Yonemori K et al Clinicopathologic characterization of epithelioid hemangioendothelioma in a series of 62 cases: A proposal of risk stratification and identification of a synaptophysin-positive aggressive subset Am J Surg Pathol 20214561626 10.1097/PAS.0000000000001660 33729740
Travis WD Brambilla E Burke AP Marx A Nicholson AG Introduction to the 2015 world health organization classification of tumors of the lung, pleura, thymus, and heart J Thorac Oncol 20151012402 10.1097/JTO.0000000000000630
Moulai N Chavanon O Guillou L Noirclerc M Blin D Brambilla E et al Atypical primary epithelioid hemangioendothelioma of the heart J Thorac Oncol 200611889 10.1016/S1556-0864(15)31539-2 17409853
Anderson T Zhang L Hameed M Rusch V Travis WD Antonescu CR Thoracic epithelioid malignant vascular tumors: A clinicopathologic study of 52 cases with emphasis on pathologic grading and molecular studies of WWTR1-CAMTA1 fusions Am J Surg Pathol 2015391329 10.1097/PAS.0000000000000346 25353289
Xie XF Huang Y Guo JH Zhang W Hou LK Wu CY et al Often misdiagnosed primary pleural epithelioid hemangioendothelioma: A clinicopathological analysis of five cases Zhonghua Bing Li Xue Za Zhi 202049128893
Gong QX Fan QH Ding Y Xiao QX Wang QY Tang JL et al Atypical epithelioid hemangioendothelioma: A clinicopathological analysis of eight cases Zhonghua Bing Li Xue Za Zhi (Chin J Pathol) 2019486205
Yang P Zhang S Yu C Yan W Yang N Li N et al Fluorescence in situ hybridization for WWTR1-CAMTA1 has higher sensitivity and specificity for epithelioid hemangioendothelioma diagnosis Am J Transl Res 2020 1245618
Lyu YT Zhang HY Zhu XL Chen JR Li XF Hu MJ A case of primary epithelioid angiosarcoma of the tonsil in a patient with recurrent hepatocellular carcinoma with pulmonary metastasis Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi (Chin J Otorhinolaryngol Head Neck Surg) 20215686970
Woo JJ Kim Y An JK Lee H Primary pleural epithelioid angiosarcoma manifesting as a loculated hemothorax: A case report and literature review focusing on CT findings Radiol Case Rep 20211630725 10.1016/j.radcr.2021.07.048 34429805
Lemus LF Minervini MH Epithelioid angiosarcoma Causing spinal cord compression Cureus 202113e14325 10.7759/cureus.14325 33968534
Derouane F Brigitte H Placide N Epithelioid angiosarcoma arising after an endovascular aneurysm repair: Case report and review of the literature Acta Clin Belg 202176397401 10.1080/17843286.2020.1742489 32186994
Bi YW Cai RR Wang SY Zhu XZ The clinicopathologic features and differential diagnosis of ocular Kimura disease and epithelioid hemangioma Zhonghua Yan Ke Za Zhi 20215768995
Markidan J Hardy N Kallen M Ma L Epithelioid hemangioma involving large arteries in the skin J Cutan Pathol 20224937780 10.1111/cup.14166 34755378
Yakobson A Alguayn W Shalata W Levin D Kian TA Korngreen A et al Epithelioid hemangioendothelioma and epithelioid hemangioma: Pazopanib as a potential salvage therapy Case Rep Oncol 20211430917 10.1159/000510806 33776722
Ishii A Watanabe S Nishihara C Kakiuchi M Akaike Y Itakura J et al Previously unreported permanent tattoo-associated angiolymphoid hyperplasia with eosinophilia/ epithelioid hemangioma Pathol Int 20217121921 10.1111/pin.13063 33503288
Yasui M Kikuchi Y Fujikura M Morita S Numakura S Watabe S et al Concurrent epithelioid hemangioma and diffuse cavernous hemangioma in the rectum clinically mimicking a malignant tumor: A case report Virchows Arch 202147984751 10.1007/s00428-021-03035-3 33496804
Marton N Ramos-Oliver I Cajal AG de Albert M Dominguez Oronoz R Transarticular epithelioid hemangioma of the ankle-a case of a rare vascular neoplasm Skeletal Radiol 202150126370 10.1007/s00256-020-03687-3 33420574
Covelli C Parente P Icolaro N Dimitri LM Vigna B Popolizio T et al primary epithelioid hemangioma of the central nervous system: A case report and review of the literature J Neuropathol Exp Neurol 2021807179 10.1093/jnen/nlaa163 33411905
Warmke LM Meis JM Sclerosing epithelioid fibrosarcoma: A distinct sarcoma with aggressive features Am J Surg Pathol 20214531728 10.1097/PAS.0000000000001559 32769431
Song L Stashek KM Benyounes A Davis DL Mulligan ME Ng VY et al Epithelioid sarcoma with retained INI1 (SMARCB1) expression Histopathology 2021784646 10.1111/his.14236 32799369
Gounder MM Merriam P Ratan R Patel SR Chugh R Villalobos VM et al Real-world outcomes of patients with locally advanced or metastatic epithelioid sarcoma Cancer 202112713117 10.1002/cncr.33365 33296083
Baker TG Lyons MJ Leddy L Parham DM Welsh CT Epithelioid sarcoma arising in a long-term survivor of an atypical teratoid/rhabdoid tumor in a patient with rhabdoid tumor predisposition syndrome Pediatr Dev Pathol 2021241648 10.1177/1093526620986492 33470921
Errani C Zhang L Sung YS Hajdu M Singer S Maki RG et al A novel WWTR1-CAMTA1 gene fusion is a consistent abnormality in epithelioid hemangioendothelioma of different anatomic sites Genes Chromosome Cancer 2011 5064453 10.1002/gcc.20886 21584898
Hisaoka M Okamoto S Aoki T Yokoyama K Hashimoto H Spinal epithelioid hemangioendothelioma with epithelioid angiosarcomatous areas Skeletal Radiol 2005347459 10.1007/s00256-005-0911-1 15877225


HPV-negative and high-grade finding in Pap cytology

A 69-year-old post-menopausal female with past medical history of idiopathic thrombocytopenic purpura, status post-total abdominal hysterectomy for uterine tumor, and chemotherapy presented with abnormal vaginal bleeding. Computerized tomography (CT) scan chest/ abdomen/pelvis revealed a 2.9 × 2.0 cm mass in the vaginal cuff. High-risk human papillomavirus (HR-HPV) testing was negative. Papanicolaou (Pap)-stained ThinPrep showed atypical hyperchromatic-crowded groups (HCGs) [Figure 1].

Pap stain: (a: ×20; b: ×40) Clusters of epithelial cells with high nucleocytoplasmic ratio; hyperchromatic nuclei; prominent nucleoli; nuclear overcrowding; and indistinct cell borders, (c: ×20; d: ×40) mesenchymal element demonstrating cigar-shaped to spindle cells (arrows) with clusters of inflammatory cells.

QUESTION # 1

The findings in Pap smear [Figure 1] may represent all except

Low-grade squamous intraepithelial lesion (LGSIL)

Atypical glandular cells of uncertain significance (AGUS)

Adenocarcinoma

Carcinosarcoma (Malignant-mixed Mullerian tumor)

ANSWER

a. Low-grade squamous intraepithelial lesion (LGSIL)

LGSIL (Option a) is characterized by distinct cytological features. The cells exhibit nuclear enlargement, typically 3 times the normal size without significantly high nuclear to cytoplasmic (N/C) ratio. Koilocytic changes include hyperchromasia along with coarse chromatin and nuclear membrane irregularities with diagnostic empty perinuclear cytoplasmic space sharply demarcated from peripheral cytoplasm. Binucleation and multinucleation are is frequent. Nucleoli are generally inconspicuous or absent. Low-grade squamous intraepithelial lesion may show increased keratinization seen as dense orangeophilic (atypical parakeratosis).[1]

This is a case of recurrent uterine carcinosarcoma (UCS). The patient presented with chief complaints of abnormal vaginal bleeding for the past 1 month. Four years before this consultation, the patient underwent total abdominal hysterectomy, bilateral salpingo-oophorectomy (BSO), and pelvic lymph node sampling. Evaluation of the hysterectomy of the specimen revealed a stage IB UCS. She underwent chemotherapy; however, after 2 cycles, her thrombocytopenia got exacerbated, and chemotherapy was terminated.

During the present consultation, rectovaginal examination revealed a mass on the left vaginal apex. On CT scan, chest/ abdomen/pelvis revealed a 2.9 × 2.0 cm vaginal mass. There was no evidence of metastatic disease in the chest, abdomen, or pelvis. Pap smear and concurrent left vaginal biopsy were done. Pap smear revealed a biphasic neoplasm composed of atypical glandular cells with a spindle cell component. The malignant glandular component was comprised three-dimensional hyperchromatic crowded cell groups with high N/C ratio, coarse clumped chromatin, and occasional prominent nucleoli, whereas spindle cell components demonstrated high N/C ratio, hyperchromasia, irregular chromatin, and nuclear pleomorphism.

Histopathological examination (H&E) of the left vaginal biopsy [Figure 2a and 2b] revealed high-grade carcinoma with sarcomatoid features. Immunohistochemistry demonstrated [Figure 2c] focal immunoreactivity for cytokeratin CAM 5.2 [Figure 2d] with mutant p53 expression [Figure 2e] and nonimmunoreactivity for desmin [Figure 2f].

Left vaginal biopsy; (a: H&E, ×4; b: H&E, ×10; c: H&E, Zoomed) demonstrated sheets of pleomorphic spindle cells with variable cellularity. (d-f: immunohistochemistry, ×10) focal immunoreactivity for CAM5. 2, aberrant p53 expression, and non-immunoreactivity for desmin.

Based on cytology and vaginal biopsy findings, a diagnosis of vaginal cuff recurrence of carcinosarcoma was made, and the patient underwent external beam radiation therapy and vaginal brachytherapy.

Depending on multiple variables from cytomorphological features sampled in the Pap smear to the interpreter experience adenocarcinoma (Option c) and the carcinomatous component of UCS may have to be interpreted as atypical glandular cells of uncertain significance (AGUS) (Option b). However, the presence of spindle cells and the patient’s history of UCS would point to the definitive interpretation as carcinosarcoma (malignant mixed Mullerian tumor).

DISCUSSION

A biphasic neoplasm showing both a malignant epithelial component and a spindle cell/mesenchymal component on a Pap smear should raise the suspicion for the possibility of carcinosarcoma.[2-5] It is reported that the detection rate of malignant cells in cervical samples of histologically proven UCS ranges from 56% to 70%;[2-5] however, UCS usually lacks the sarcomatous component in Pap smears.

The carcinomatous component of UCS is more readily identifiable than the mesenchymal component [Table 1].[2-8]

Publish metadata.

S. No. Author/Year No. of cases Pap type Findings
1. Tenti et al., 1989[2] 10 Conventional

7 positive smears (2 diagnosed as a mixed tumor at first examination, 1 upon revision).

2. Costa et al., 1992[3] 21 Conventional

Sarcoma component was absent in 4 out of 9 (44%) positive smears available for cytology review.

3. Casey et al., 2003[4] 9 Conventional

Five abnormal Pap smears were available for review:

Epithelial component positive in all five cases.

Mesenchymal component was absent in all five cases

4. Snyder et al., 2004[5] 25 Conventional

60% (15/25) were read as abnormal.

All malignant elements were epithelial.

Two cases (8%) had atypical spindle cells, but no diagnostic sarcoma.

5. Salman et al., 2006[6] 1 SurePath (LBC)

Cervical smear positive for glandular elements and some spindle-shaped cells.

6. Gupta et al., 2014[7] 8 SurePath (LBC)

Malignant epithelial component in all eight cases

Sarcomatous elements in three cases.

7. Hanley et al., 2015[8] 40 4 conventional, 36 liquid based

11 (27.5%) - negative.

5 (12.5%) - atypical squamous cells of undetermined significance, 6 (15%) as AGUS.

16 (40%) as malignant.

2 (5%) as high-grade squamous intraepithelial lesions.

AGUS: Atypical glandular cells of uncertain significance, LBC: Liquid-based cytology

This is an important diagnostic pitfall in cytology as most cases of UCS may be reported as AGUS, squamous cell carcinoma, endometrial adenocarcinoma, poorly differentiated carcinoma, or adenocarcinoma not otherwise specified.[5]

Detecting UCS through cytologic examination poses diagnostic challenges due to several factors. These challenges include sampling errors, random exfoliation of cells, cellular degeneration, and sporadic shedding of mesenchymal component.[4] In addition, frequent association with atrophy can also make it difficult to detect UCS cells in the background of numerous HCGs of parabasal cells (PBC) in atrophic samples during cytologic examination of Pap smears.[7]

In the current case, on initial review, the smear was interpreted as a hypercellular smear with an inflammatory background, cellular degeneration, and numerous malignant glandular cells in clusters and three-dimensional balls [Figure 1a and 1b], raising the possibility of AGUS favoring neoplasia, but HR-HPV testing was negative. After re-reviewing the smear, HCGs composed of a few cigar-shaped spindle cells [Figure 1c] suggestive of a mesenchymal component was identified. The final diagnosis of high-grade carcinoma with sarcomatous features were rendered. The cytologic findings were consistent with the vaginal biopsy results.

It is important to consider that, while HPV testing is a helpful tool in cervical cancer screening, a negative high-risk HPV test does not automatically exclude malignancy, because some malignancies arising in the uterine corpus or adnexa can be identified in cervical smears due to exfoliation, drop metastasis, and/or direct extension to cervix, such as UCS endometrial carcinomas, clear cell carcinoma, high-grade serous carcinoma, and mesonephric adenocarcinoma are not HPV related.

Furthermore, there are other aggressive cancers such as endometrial carcinomas, clear cell carcinoma, high-grade serous carcinoma, mesonephric adenocarcinoma, and adenocarcinomas arising from other sites that show high-grade morphology in Pap smears despite testing negative for HPV.[9,10]

One of the potential reasons for the initial omission of the mesenchymal elements is the frequent association of atrophic changes in the elderly patients with abundant inflammatory cells and cellular degeneration with many HCGs of PBCs in Pap smears. Another possibility could be the inability to detect spindle cells/mesenchymal elements in Pap smear on the first look as the mesenchymal element with atypical spindle cells [as in the current case, Figure 1c, and arrows in Figure 1d] is rarely present due to infrequent shedding and sampling artifacts.

It is crucial to understand that a significant percentage of women with AGUS may turn out to be high-grade pre-invasive squamous disease, adenocarcinoma in situ, adenocarcinoma, or invasive cancers from sites other than the cervix. Owing to the aggressive nature of UCS, an accurate and timely diagnosis is indispensable. An algorithmic approach is suggested for interpreting AGUS cells in relation to UCS Pap smear [Figure 3].

Algorithmic approach for evaluating atypical glandular cells of uncertain significance in post-menopausal women with reference to uterine carcinosarcoma (UCS). This is not for definitive diagnosis of UCS.

ADDITIONAL QUIZ QUESTION Question # 2

Most common mutations in uterine carcinosarcoma?

TP53 mutations

HER2

TERT

KRAS.

Question # 3

Most consistent independent predictor of outcome

Age at presentation

Stage

Angioinvasion

Depth of invasion

Number of mitotic figures.

BRIEF REVIEW OF THE TOPIC

UCS, previously known as malignant mixed Müllerian tumor, is a rare and aggressive biphasic tumor in the female reproductive system, accounting for <5% of uterine tumors. It is characterized by the presence of high-grade epithelial and mesenchymal components. UCS is commonly diagnosed in postmenopausal women with a history of bleeding with a median age of presentation of 62 years.[11] The most common locations within the female genital tract include the uterus and ovaries. Cases of UCS originating from the cervix, vagina, ovary, fallopian tube, and peritoneum are also reported.[11]

It is biologically a de-differentiated endometrial carcinoma with its own pathogenesis and molecular profile.[12] Several hypotheses to explain the origination of UCS have been proposed. The collision hypothesis suggested that UCS forms when separate epithelial and mesenchymal tumors collide and merge within the uterus. On the other hand, the combination hypothesis posits that UCS arises from a single stem cell capable of divergent differentiation, giving rise to both epithelial and mesenchymal components within the tumor. Finally, the conversion hypothesis, which is the most widely accepted theory proposed that UCS is a metaplastic carcinoma that carcinomatous cells can undergo a conversion process where they transform into sarcomatous cells through epithelial to mesenchymal transition (EMT).[13,14] This conversion is associated with high scores of EMT gene signatures and is likely influenced by epigenetic alterations at microRNA promoters, histone gene mutations, and gene amplifications.[14]

TP53/p53 abnormalities are present in a majority of the UCS. The proportion of replicative DNA polymerase epsilon (POLE) and hypermutated microsatellite instability-high is related to the epithelial component, being approximately 25% and 3% in endometrioid and non-endometrioid components.[15] The stage at diagnosis follows a bimodal distribution, approximately 40–50% of cases are detected at an early stage (International Federation of Gynecology and Obstetrics [FIGO] Stages I and II), while about 50–60% are detected at an advanced stage (FIGO stages III and IV). At the time of diagnosis, up to 30–40% of patients display lymph node metastases. In addition, about 10% of patients present with distant metastatic spread, with lung as the commonly affected site. These findings highlight the aggressive nature of UCS and the importance of early detection and intervention. The 5-year survival rate of UCS is approximately 30% (Stage I and II: 59%; Stage III: 25%; Stage IV: 9%).[16]

Histologically, the malignant cell in UCS exhibits a biphasic nature; the carcinomatous component is usually endometrioid or serous, but clear and undifferentiated may be seen. The sarcomatous component is usually high-grade sarcoma NOS, but heterologous elements (rhabdo, chondro, osteo, and lipo) might be encountered.[17,18]

Recommended treatment for UCS involves surgical staging with total hysterectomy with BSO, pelvic lymphadenectomy, para-aortic lymph node sampling, and peritoneal washings.[19]

SUMMARY

The Pap smear can serve as both a screening and diagnostic tool.

It is essential to consider UCS as one of the possible potential differentials, particularly in post-menopausal women presenting with vaginal bleeding.

An algorithmic approach in the case of AGUS favoring neoplasia can facilitate correct interpretation of UCS in Pap smears.

Comprehensive evaluation, incorporating clinical history and histological biopsies, should be considered while interpreting the result of Pap smears with AGUS.

It is essential to report on both the carcinomatous and sarcomatous components for proper management.

Answers for Question 2 through 3:

2. a

3. b


References

Solomon D Davey D Kurman R Moriarty A O'Connor D Prey M et al The 2001 Bethesda system: Terminology for reporting results of cervical cytology JAMA 200228721149 10.1001/jama.287.16.2114 11966386
Tenti P Babilonti L La Fianza A Zampatti C Carnevali L Sonnino P et al Cytology of malignant mixed mesodermal tumor of the uterus: Experience of 10 cases Eur J Gynaecol Oncol 1989101258
Costa MJ Tidd C Willis D Cervicovaginal cytology in carcinosar-coma [malignant mixed mullerian (mesodermal) tumor] of the uterus Diagn Cytopathol 199283340 10.1002/dc.2840080108 1312925
Casey MB Caudill JL Salomao DR Cervicovaginal (Papanicolaou) smear findings in patients with malignant mixed Mullerian tumors Diagn Cytopathol 2003282459 10.1002/dc.10276 12722119
Snyder MJ Robboy SJ Vollmer RT Dodd LG An abnormal cervicova-ginal cytology smear in uterine carcinosarcoma is an adverse prognostic sign: Analysis of 25 cases Am J Clin Pathol 20041224349 10.1309/KUA1FQ3BQRC8YG4V 15362375
Salman WD Sharief S Howat AJ Pleomorphic spindle cells and hyperchromatic crowded groups in a cervical smear Cytopathology 2006173968 10.1111/j.1365-2303.2006.00345.x 17168924
Gupta N Dudding N Smith JH Eight cases of malignant mixed Müllerian tumor (carcinosarcoma) of the uterus: Findings in SurePath™ cervical cytology Diagn Cytopathol 2014421659 10.1002/dc.22910 22865798
Hanley KZ Oprea-Ilies G Ormenisan C Seydafkan S Mosunjac MB Atypical findings on cervicovaginal smears correlate with cervical involvement by malignant mixed Müllerian tumors of the uterus Acta Cytol 20155931924 10.1159/000439159 26315658
Pirog EC Lloveras B Molijn A Tous S Guimerà N Alejo M et al HPV prevalence and genotypes in different histological subtypes of cervical adenocarcinoma, a worldwide analysis of 760 cases Mod Pathol 201427155967 10.1038/modpathol.2014.55 24762548
Alexander C White M Maleki Z Rodriguez EF HPV-ISH-negative invasive cervical squamous cell carcinoma: Histologic and Pap test results Acta Cytol 20196341723 10.1159/000500595 31195388
Matsuo K Ross MS Machida H Blake EA Roman LD Trends of uterine carcinosarcoma in the United States J Gynecol Oncol 201829e22 10.3802/jgo.2018.29.e22 29400015
Cantrell LA Blank SV Duska LR Uterine carcinosarcoma: A review of the literature Gynecol Oncol 20151375818 10.1016/j.ygyno.2015.03.041 25805398
McCluggage WG Malignant biphasic uterine tumours: Carcinosarcomas or metaplastic carcinomas? J Clin Pathol 2002553215 10.1136/jcp.55.5.321 11986333
Cherniack AD Shen H Walter V Stewart C Murray BA Bowlby R et al Integrated molecular characterization of uterine carcinosarcoma Cancer Cell 20173141123 10.1016/j.ccell.2017.02.010 28292439
Bogani G Ray-Coquard I Concin N Ngoi NY Morice P Caruso G et al Endometrial carcinosarcoma Int J Gynecol Cancer 20233314774 10.1136/ijgc-2022-004073 36585027
Gonzalez Bosquet J Terstriep SA Cliby WA Brown-Jones M Kaur JS Podratz KC et al The impact of multi-modal therapy on survival for uterine carcinosarcomas Gynecol Oncol 201011641923 10.1016/j.ygyno.2009.10.053 19896181
Silverberg SG Major FJ Blessing JA Fetter B Askin FB Liao SY et al Carcinosarcoma (malignant mixed mesodermal tumor) of the uterus. A Gynecologic Oncology Group pathologic study of 203 cases Int J Gynecol Pathol 19909119 10.1097/00004347-199001000-00001 2152890
Lopez-Garcia MA Palacios J Pathologic and molecular features of uterine carcinosarcomas Semin Diagn Pathol 20102727486 10.1053/j.semdp.2010.09.005 21309261
Nemani D Mitra N Guo M Lin L Assessing the effects of lymphadenectomy and radiation therapy in patients with uterine carcinosarcoma: A SEER analysis Gynecol Oncol 2008111828 10.1016/j.ygyno.2008.05.016 18674808


An uncommon vascular lesion over the right hand

A 45-year female presented with progressively enlarging, 2 × 1 cm, soft, mobile, non-tender swelling over the dorsum of the right hand around the wrist joint for 6 months. Fine needle aspiration cytology are shown in Figure 1.

(a) Isolated cell clusters with plump nuclei displaying “hobnailing” (May Grunwald-Giemsa stain ×100). (b) There is moderate pleomorphism with distinct nucleoli (Papanicolaou stain ×400). (c) A venous wall with intravascular organized thrombus (Hematoxylin and Eosin stain ×400). (d) Thrombus invaginated by anastomosing delicate papillae with a hyalinized core lined by endothelial cells (Hematoxylin and Eosin stained cell-block section ×100). (e) Plump endothelial cells with hobnail nuclei (Hematoxylin and Eosin stained cell-block section ×400).

Q1. What is your interpretation?

Hemangioma

Angiosarcoma

Papillary endothelial hyperplasia (PEH)

Hobnail hemangioendothelioma.

Answer: c. Papillary endothelial hyperplasia (PEH)

Common sites of involvement by PEH include the head-and-neck, lip, tongue, buccal mucosa, and limbs. Usually presents as a reddish-blue nodular lesion in the skin or mucosa.[1] On histology, they present as an intravascular lesion with residual organizing thrombus, anastomosing vascular channels, and papillary formations with hyaline or fibrous stalks lined by plump endothelial cells (histiocyte or epithelial-like).[2-5]

Q2. Which among the following finding is characteristically seen in PEH

Hobnail

Thrombus

Nuclear atypia

Necrosis.

The differential diagnoses considered include vasoformative lesions such as hemangioma, angiosarcoma, and hobnail hemangioendothelioma. Hemangioma and angiosarcoma commonly involve the skin and subcutis of the head-and-neck region. Angiosarcomas exhibit marked nuclear atypia, necrosis, and mitoses. They are never confined to the intravascular location.[1] Hobnail hemangioendothelioma (retiform type) is a rare intermediate-grade (locally aggressive) tumor of vascular origin affecting distal extremities. It consists of numerous elongated vessels (retiform) involving the dermis and extending into the subcutis. They display “hobnail” endothelial cells similar to PEH.[6] However, they lack the organized thrombus seen in histology sections.[6]

Q3. Which of the following is a reactive, non-neoplastic process.

Arteriovenous malformation

Hereditary hemorrhagic telangiectasia

PEH

Hobnail hemangioma.

Q4. Papillary endothelial hyperplasia is not a common feature of PEH:

Usually begins within a thrombosed blood vessel

They usually have abundant thrombotic material

They show papillae are lined by plump, normochromatic endothelial cells, in a single, non-stratified layer

PEH shows frequent mitotic figures.

Papillary endothelial hyperplasia is a reactive, non-neoplastic lesion representing an exuberant organization and recanalization of the thrombus.[3] It occurs in normal vessels as well as in preexisting vascular lesions such as varices, hemorrhoids, pyogenic granulomas, hematomas, and angiosarcomas. The clinical presentation is non-specific, so histopathological examination is ultimately required to establish the diagnosis.

A BRIEF REVIEW OF THE TOPIC

PEH is an exuberant intravascular, endothelial cell proliferation mimicking a neoplastic tumor of vascular origin with papillary tufting and “hobnail” morphology.[1] Although initially described as a neoplastic lesion by Masson,[7] it is now considered as reactive vascular proliferation with an organized thrombus.[8] In its pure form, as in our case, it occurs in dilated veins of the head, neck, limbs, and trunk. It can also engraft in pre-existing vasoformative lesions. In the early lesions, numerous small, delicate papillae with a hyalinized core lined by plump endothelial cells invaginate into the lumen with tufting growth and “hobnail” morphology as in our case.[2-5] During the late stage, the fusion of the papillae results in an anastomosing network of vessels embedded in a loose, meshlike stroma of connective tissue.[1] Rupture of the vein can result in the passive extension of the lesion into the surrounding soft tissue.[1] The prognosis is generally excellent. Essentially, all cases are treated and cured by simple excision.[1] Recurrent lesions are generally secondarily superimposed on preexisting vasoformative lesions.[1]

SUMMARY

Papillary endothelial hyperplasia carries a significant potential for misdiagnosis as angiosarcoma. Cytopathological diagnosis of this uncommon reactive vascular lesion is difficult only on FNA findings. A vascular lesion may be considered, especially when polygonal cells with plump nuclei and “hobnail” morphology are seen in a hemorrhagic background. The cytological differential diagnoses to be considered include the common neoplastic vascular lesions. Histomorphology of the resected specimen helps provide conclusive evidence in excluding the neoplastic nature of the vascular lesion.


References

Weiss SW Goldblum JR Papillary endothelial hyperplasia (vegetant intravascular hemangioendothelioma, intravascular angiomatosis) Enzinger and Weiss’s Soft Tissue Tumors St. Louis Mosby 20207447
GarciaMacias MC Abad M Alonso MJ Flores T Bullón A Masson’s vegetant intravascular hemangioendothelioma. Fine needle aspiration cytology, histology and immunohistochemistry of a case Acta Cytol 1990341758
Novak JA Ferguson DJ Komorowski RA Fine needle aspiration cytology of papillary endothelial hyperplasia. A case report Acta Cytol 1999436636 10.1159/000331163 10432892
Suh KS Shin KS Park IA Intravascular papillary endothelial hyperplasia of the neck masquerading as malignancy on fineneedle aspiration cytology Diagn Cytopathol 200329147 10.1002/dc.10207 12827709
Karim RZ Merani R Shannon K Watson G Papillary endothelial hyperplasia of the orbit: Report of a case highlighting a pitfall on fine needle aspiration biopsy Acta Cytol 20075120710 10.1159/000325718 17425205
Weiss SW Goldblum JR Papillary endothelial hyperplasia (vegetant intravascular hemangioendothelioma, intravascular angiomatosis) Enzinger and Weiss’s Soft Tissue Tumors St. Louis Mosby 20207727
Masson MP Hemangioendotheliome vegetant intravasculaire Bull Soc Anat (Paris) 19239351724
Clearkin KP Enzinger FM Intravascular papillary endothelial hyperplasia Arch Pathol Lab Med 19761004414


Unusual findings on fine-needle aspiration cytology of a retroperitoneal mass

A 77-year-old male presented with intermittent abdominal pain. CT scan revealed a 4.7 cm retroperitoneal mass. A subsequent fine-needle aspiration (FNA)with a core biopsy revealed basophilic clumps of acellular homogenous material on Diff Quik stains performed during rapid on site evaluation [Figure 1a]. Pap stain showed greenish round deposits [Figure 1b], whereas cell block showed dense hyalinization and amorphous eosinophilic material amidst benign liver parenchyma [Figure 1c and d].

(a) Retroperitoneal fine-needle aspiration (FNA): Diff Quik Stain showing acellular, blue, amorphous material. (b) Retroperitoneal FNA: Pap stain showing acellular, greenish, rounded amorphous material. (c) Retroperitoneal FNA: Cell block with H and E stain showing hyaline deposits of amorphous eosinophilic material, amidst benign liver parenchyma (Low power view). (d) Retroperitoneal FNA: Cell block with H and E stain showing hyaline deposits of amorphous eosinophilic material (High power view).

What is the diagnosis?

Soft-tissue tumor

Solitary fibrous tumor

Collagenous spherulosis

Amyloid

Answer:

d) Amyloid

The most common diagnosis of retroperitoneal masses is a soft-tissue tumor such as a liposarcoma. Other common entities that can lead to retroperitoneal mass formation are lymphomas. Collagenous nodules as seen in our patient on H and E section can be also seen in some cases of retroperitoneal fibrosis. However, in our patient, the collagenous areas were striking and amorphous and immediately raised the possibility of amyloid deposits. The presence of inflammatory cells and plasma cells also favored the diagnosis of amyloid.

The diagnosis was confirmed by positive Congo staining [Figure 2] with apple green birefringence [Figure 3]. Serum protein electrophoresis and immunofixation were negative, but his serum lambda-free light chains were elevated at 109.3 mg/L with a low of kappa/lambda of 0.21 prompting referral to hematology. Liquid chromatography-tandem mass spectrometry was performed on peptides extracted from micro dissected areas of the paraffin-embedded specimen and detected a peptide profile consistent with amyloid light chain (AL) (lambda)-type amyloid deposition, supporting the diagnosis of AL amyloidosis. The patient’s abdominal pain had subsided, and bone marrow biopsy was deferred for later. There was not enough evidence to determine if the amyloidosis was systemic or localized.

Retroperitoneal fine-needle aspiration: Congo red stain showing brownish red acellular, amorphous deposits.

Retroperitoneal fine-needle aspiration: Congo red stain showing apple green birefringence under polarized light.

Amyloidosis is a disease that occurs from the deposition of mis-folded proteins in organs and tissues. Amyloid refers to a group of abnormal proteins that share a ß-pleated sheet structure.[1] This structure makes the proteins insoluble and unsusceptible to enzymatic digestion as well as responsible for its physical attributes such as its amorphous homogenous eosinophilic material and “apple-green” birefringence of Congo red stains under polarized light.[1]

There are different types of amyloid deposits, but AL (A = amyloid, L = light chain) is the most common and is usually seen to affect older men. In AL amyloidosis, plasma cells produce unstable quantities of monoclonal free light chains.[2] AL amyloidosis can be of the kappa or lambda type. These free light chains join to form amyloid fibrils, which the body cannot eliminate easily.[2] Amyloid fibrils start to build up in tissues and organs, inhibiting their function over time. Amyloids can precipitate in the same location, in which they are created resulting in localized amyloidosis. Amyloids can also spread throughout the bloodstream to other organs resulting in systemic amyloidosis.[3]

Which new specific and sensitive test can confirm the presence of amyloid and also specify the type (AA, AL [Kappa or Lambda], and transthyretin/hereditary type)?

Congo red staining

Immunohistochemistry

Liquid chromatography-tandem mass spectrometry

Electron microscopy

Answer(s):

c) Liquid chromatography-tandem mass spectrometry

AL amyloidosis is typically diagnosed by staining the sample with Congo red and observing for apple-green birefringence. Immunohistochemistry is not useful as it often results in non-specific staining and the sensitivity of the technique is low. Using this process, diagnosis of the specific kind of amyloidosis and determining if it is systemic or localized is difficult. However, technological advancements in mass spectrometry have made it more efficient for diagnosing amyloidosis. Mass spectrometry can analyze specific proteins allowing for a diagnosis of not only amyloidosis but also the type of amyloidosis, giving more options of treatment to the patient. Findings from mass spectrometry can also provide a general diagnosis independent of the application of Congo red staining.[4] In addition, mass spectrometry can be easily performed on formalin fixed paraffin-embedded tissue.[4]

ADDITIONAL QUESTIONS

What are the symptoms of AL amyloidosis?

Shortness of breath

Chronic kidney disease

Constipation

Arrhythmia

All of the above

Answer(s):

d. All of the above

AL amyloidosis is generally found in older men. General symptoms of AL amyloidosis include abnormal bleeding, dizziness, fatigue, shortness of breath, and muscle weakness.[5] AL amyloidosis also hinders the function of specific organs. A build-up of amyloid proteins in the kidneys can cause chronic kidney disease (renal AL amyloidosis).[5] Accumulation of amyloid proteins in the heart (AL cardiac amyloidosis) can cause heart palpitations, arrhythmia, shortness of breath, fatigue, as well as thickening of heart tissues increasing the chance of heart failure.[5] In the gastrointestinal tract, escalation in amyloid proteins can cause weight loss, abdominal pain, constipation, diarrhea, gastric reflux, and vomiting.[5] Amyloid proteins can also develop in the central nervous system causing changes in blood pressure, erectile dysfunction, pain in the hands, feet, and lower legs, as well as confusion.[5] Unlike some forms of amyloidosis, AL amyloidosis cannot be transmitted genetically.[3]

What treatments are implicated on a patient with AL amyloidosis?

Chemotherapy

No treatment

Bone marrow transplant

Removal of the mass

All of the above

Answer(s):

e). All of the above

There is no known cure for systemic AL amyloidosis. Chemotherapy, like that used in cancer patients, is typically prescribed to slow down the build-up of amyloid proteins by killing cells that produce them.[6] Another method is bone marrow transplant, also known as a stem cell transplant. Patients with localized amyloidosis do not receive treatment, instead, the mass is removed if it affects organ function; otherwise, the mass can be left in the body.

What is the latest method of confirming a clinical diagnosis of amyloidosis?

Blood test

Fat pad biopsy with Congo red staining and electron microscopy

Anterior fat pad FNA biopsies with mass spectrometry/ proteomics

Rectal biopsies

Answer:

c). Anterior fat pad FNA biopsies with mass spectrometry/ proteomics

Earlier, abdominal fat biopsies for confirming amyloidosis relied on electron microscopy and Congo red staining.[7] Now, better tests for diagnosis are available, and FNA samples can be aspirated successfully[8,9] and processed using laser dissection, mass spectrometry, and proteomics.[10] Rectal mucosal biopsies have been replaced by anterior abdominal wall fat pad FNA technique as shown in videos.[8,9]

SUMMARY

AL amyloidosis is a rare condition typically found in older male patients. Initial diagnosis of AL amyloidosis usually involves Congo red staining, looking for apple-green birefringence. Mass spectrometry recognizes all amyloid types (AL vs. AA vs. transthyretin/hereditary) and can easily be performed on formalin fixed, paraffin embedded tissue from core biopsies, and cell blocks. It confirms the diagnosis, identifies specific mutations, and helps guide appropriate treatment.


References

Immunohistochemistry in Amyloidosis Available from: https://www.propath.com/diagnostic-services/resources/2022/11/amyloidosis/?q=immunohistochemistry%20in%20amyloidosis [Last accessed on 2023 Mar 11]
Understanding Free Light Chains (FLCs) Available from: https://www.amyloidosis.org.uk/about-amyloidosis/alamyloidosis/understanding-free-light-chains-flcs [Last accessed on 2023 Mar 11]
Vaxman I Visram A Pasvolsky O Kumar S Dispenzieri A Buadi F et al Retroperitoneal involvement with light chain amyloidosis-Case series and literature review Leuk Lymphoma 20216231622 10.1080/10428194.2020.1832670 33118418
Dasari S Theis JD Vrana JA Rech KL Dao LN Howard MT et al Amyloid typing by mass spectrometry in clinical practice: A comprehensive review of 16,175 samples Mayo Clin Proc 202095185264 10.1016/j.mayocp.2020.06.029 32861330
Symptoms of AL Amyloidosis Available from: https://www.myamyloidosisteam.com/resources/symptoms-of-alamyloidosis [Last accessed on 2023 Mar 11]
Amyloidosis Available from: https://www.hopkinsmedicine.org/health/conditions-and-diseases/amyloidosis
Devata S Hari P Markelova N Li R Komorowski R Shidham VB Detection of amyloid in abdominal fat pad aspirates in early amyloidosis: Role of electron microscopy and Congo red stained cell block sections Cytojournal 2011811 10.4103/1742-6413.82278 21760829
Shidham VB Hunt B Jaradeh SS Barboi AC Devata S Hari P Performing and processing FNA of anterior fat pad for amyloid J Vis Exp 2010441747 10.3791/1747 21085098
Shidham VB Updates in processing of anterior fat pad aspirate for amyloid (with video and sketches) Cytojournal 20201715 10.25259/Cytojournal_31_2020 33093851
Vrana JA Gamez JD Madden BJ Theis JD Bergen HR 3rd Dogan A Classification of amyloidosis by laser microdissection and mass spectrometry-based proteomic analysis in clinical biopsy specimens Blood 200911449579 10.1182/blood-2009-07-230722 19797517


Cytomorphology of a brain lesion and its pitfall

A 25-year-old man presented with complaints of giddiness and severe headache for 20 days. Magnetic resonance imaging showed a solid mass lesion of size 56 × 57 mm in the left frontal lobe. Intraoperative tumor cavity fluid (2 mL, clear) was aspirated and sent for cytological examination. Cytocentrifuge smears prepared were cellular [Figure 1a-c].

(a-c) (GIEMSA ×40) Smears showing tumor cells in clusters and singly scattered having high N:C ratio and opened up chromatin and pleomorphism. (a) Inset shows atypical mitoses pointed by solid arrow.

QUESTION 1 What is your interpretation?

Degenerated cells

Adenocarcinoma

Glioma

Small blue round cell tumor.

ANSWER TO QUESTION 1

The correct cytopathological interpretation is d. Small blue round cell tumor.

EXPLANATION

Cytocentrifuge smears prepared were cellular comprising of monomorphic tumor cells arranged in clusters and nests. Individual tumor cells are round to ovoid having scant to moderate amount of delicate cytoplasm. Nuclei were regular, smooth with opened up chromatin, showing mild-to-moderate degree of pleomorphism [Figure 1a-c]. Atypical mitosis was noted. However, rosette or acini formation or necrosis or moulding was not seen.

The cells were cohesive with no acini formation, delicate chromatin and no nucleoli thereby ruling out adenocarcinoma (Option b).

No glial matrix or fibrillary processes or rosenthal fibres or vessels or calcification or cellular pleomorphism or endothelial cell proliferation were displayed hence possibility of glioma was also excluded from the study (Option c).

Degenerated cells (Option a) were seen but most of the cells were preserved and had distinct cell boundary with nuclear margins and cellular details as discussed briefly above. Based on findings described above, a provisional diagnosis of small blue round blue cell tumor was considered. Cell block and immunocytochemistry could not be performed due to exhaustion of fluid while making smears for routine examination.

Excision surgery was performed and histopathological examination revealed a tumor [Figure 2a and b].

(a) Tumor cells arranged in nest, chords and trabeculae (H & E ×10); (b) Tumor cells having round nucleus and powdery chromatin (H & E ×40).

QUESTION 2

All of the following are among the differentials except

Germ cell tumor

Astrocytoma

Melanoma

Neuroendocrine tumor.

EXPLANATION

Histopathological examination revealed tumor cells arranged in nests, chords and trabeculae showing moderate degree of pleomorphism. Cells were round to oval with stippled nuclear chromatin [Figure 2a and b]. Areas of hemorrhage and focal areas of necrosis were also seen. Mitotic figures of 2–3/10 high power field noted. The various differential included are neuroendocrine neoplasm (NEN), germ cell tumor, melanoma, and lymphoma.

Astrocytoma (option b) will show tumor cells having irregular nuclei with variable degree of atypia along with presence of glial processes. Cells can have variable cellular morphology in a fibrillary background which is not seen in this case.

Initial panel of immunohistochemistry (IHC) was applied which included Glial fibrillary acidic protein (GFAP), LCA, SALL4, CK7, CK20, CD117, desmin, myogenin, CD99, HMB 45, and S-100. All above markers came out to be negative except focal pan-cytokeratin positivity [Figures 3a-e]. A secondary panel of synaptophysin and chromogranin applied was positive. KI67 proliferation index was >20%.

Immunohistochemistry: Strong positive synaptophysin (a), Positive Chromogranin (b), Pancytokeratin focal positive (c), and Glial fibrillary acidic protein negative (d), Ki67 >20% (e) (×40).

QUESTION 3

Which marker helped in excluding the primary nature of the lesion?

Synaptophysin

Chromogranin

Pan-cytokeratin

GFAP.

EXPLANATION

GFAP is positive in intermediate filament of astrocytes. In the present case, it was negative in the tumor cells.

Synaptophysin and chromogranin are markers for neuroendocrine cells whereas pancytokeratin indicates the epithelial nature of the lesion.

QUESTION 4

All further IHCs can be put to check for the metastatic site of NEN except

Insulinoma associated protein 1 (INSM-1)

CDX2- Caudal type homeobox2 (CDX2)

Thyroid transcription factor 1 (TTF-1)

Pancreatic battery (Insulin, gastrin, somatostatin, glucagon, etc.).

EXPLANATION

Insulinoma associated protein 1 (INSM-1) is a nuclear marker of neuroendocrine differentiation with better sensitivity and specificity as compared to synaptophysin, chromogranin, and CD56.

CDX2 helps in location metastasis from colon whereas TTF-1 from lung.

On extended IHC, the cells were focally positive for TTF-1 [Figure 4] while negative for Napsin-A and CDX2.

Tumor cells are positive for TTF-1 (×40).

FURTHER FOLLOW UP OF THE CASE

A Positron emission tomography/Computed tomography (PET/CT) was performed which showed metabolically active soft tissue density lesion measuring 2.5 × 1.7 × 2.5 cm in the upper lobe of left lung in the para mediastinal aspect with metabolic activity in the mediastinal lymph nodes. The above PET/CT findings further confirmed the histopathological diagnosis of tumor arising in lung.

BRIEF REVIEW

NEN can develop at any of the initial mentioned systems where the neuroendocrine cells are present.[1] Incidence of central nervous system (CNS) metastasis is very rare and accounts 1.5–5% of all patients[2] where NEN source found to be in 1.3–1.4% in all cases of CNS metastasis.[3] It is always a challenging task to detect primary focus, because patients have specific symptoms when the tumor size is small. Radiological investigations including somatostatin scintigraphy and PET/CT proved to be very useful. Metastatic disease in addition to differentiation and proliferation rates is an important prognostic factor in NEN. Presence of brain metastasis is usually found to be characteristic of a systemic dissemination and disease progression.

Cytological features of neuroendocrine tumors are described in literature that includes monomorphic population of cells, loosely cohesive fragments, medium in size having abundant cytoplasm, and very few mitotic figures. Occasional rosette formation can be seen. Nuclei were regular, smooth with opened up chromatin, showing mild-to-moderate degree of pleomorphism [Figure 1]. Occasional cells showed prominent nucleolus. Atypical mitosis was seen. Due to variation in the cytomorphological features of neuroendocrine tumor, they mimic a variety of tumors. The features range from discrete cells to tight cohesive clusters, hyperchromatic nuclei with basophilic to granular to scant cytoplasm. Nuclear moulding may or may not be seen. Pleomorphism can be seen.[4]

In the present case, diagnosis of metastatic neuroendocrine tumor Grade 3 was made. TTF-1 positivity pointed out the brain tumor was metastatic, which was later confirmed by PETCT that exhibited a primary lesion in the lung. The grading of neuroendocrine tumors (NET) is based on Ki67 proliferation index. However, in neuroendocrine carcinomas, also it is advised for distinguishing it from Grade 3 NETs.[5] Features favoring neuroendocrine carcinoma are dirty smear with areas of necrotic debris and very scant cytoplasm in small cell type whereas abundant in large cell type. The population of cells having salt and paper chromatin, severe nuclear fragility, and many atypical mitoses.[2,6] Only limited previous case reports are available discussing NEN initial presentation as brain metastasis,[7-11] one of which was reported as a primary brain NEN of third ventricle adjacent to which in the paraventricular nucleus resides group of neuroendocrine cells.[12] Mostly by the time patient presents with brain metastasis either metastasis to lung happens or develop primary lung tumor. The metastasis to brain occurs due to hematogenous spread of the tumor. Leading cause of death in patients with CNS NENs is secondary to systemic disease progression and a better 10-year overall survival rate is seen in primary brain NENs.[13] Imaging workup holds importance where CT imaging has sensitivity of 95% in identification of primary tumor.[14] Etoposide -platinum chemotherapy is the treatment of choice in grade 3 NEN.[15] In metastatic brain NENs, radiation therapy and surgery are found to be beneficial but more research is required for proper management of primary brain NENs.[16]

SUMMARY

Metastatic disease is a major prognostic factor in NEN s in addition to differentiation and proliferation rate. Brain metastasis of neuroendocrine carcinoma is a rare entity but the possibility of it should be borne in mind for accurate diagnosis. In the present study, patient has been on regular follow-up for 9 months and is living disease free while continuing on etoposide chemotherapy.


References

Shalaby AM Kazuei H Koichi H Naguib S Al-Menawei LA Assessment of intracranial metastases from neuroendocrine tumors/carcinoma J Neurosci Rural Pract 201674359 Retraction in: J Neurosci Rural Pract 2017;8:322 10.4103/0976-3147.182779 27365963
Nicholson SA Ryan MR A review of cytologic findings in neuroendocrine carcinomas including carcinoid tumors with histologic correlation Cancer 20009014861 10.1002/1097-0142(20000625)90:3<148::AID-CNCR3>3.0.CO;2-9 10896328
Hlatky R Suki D Sawaya R Carcinoid metastasis to the brain Cancer 2004101260513 10.1002/cncr.20659 15495181
Gupta S Gupta P Rohilla M Gupta N Srinivasan R Rajwanshi A et al Neuroendocrine carcinomas: Cytological mimics and diagnostic dilemmas Diagn Cytopathol 2020484405 10.1002/dc.24386 31976618
Duan K Mete O Algorithmic approach to neuroendo-crine tumors in targeted biopsies: Practical applications of immunohistochemical markers Cancer Cytopathol 201612487184 10.1002/cncy.21765 27529763
Mete O Hannah-Shmouni F Kim R Stratakis C Inherited neuroendocrine neoplasms Asa SL La Rosa S Mete O The Spectrum of Neuroendocrine Neoplasia Cham, Switzerland Springer Nature 202140960 10.1007/978-3-030-54391-4_18
Maiuri F Cappabianca P De Caro MD Esposito F Single brain metastases of carcinoid tumors J Neurooncol 20046632732 10.1023/B:NEON.0000014517.56866.bc 15015664
Hussein AM Feun LG Savaraj N East D Hussein DT Carcinoid tumor presenting as central nervous system symptoms. Case report and review of the literature Am J Clin Oncol 1990132515 10.1097/00000421-199006000-00015 2189289
Greene KA Anson JA Martinez JA Spetzler RF Johnson PC Multicystic metastatic carcinoid to brain: Case report J Neurooncol 1993171520 10.1007/BF01054269 8120567
Porter DG Chakrabarty A McEvoy A Bradford R Intracranial carcinoid without evidence of extracranial disease Neuropathol Appl Neurobiol 200026298300 10.1046/j.1365-2990.2000.00257.x 10886688
Nakamura Y Shimokawa S Ishibe R Ikee T Taira A Pulmonary carcinoid found in a patient who presented with initial symptoms of brain metastasis: Report of a case Surg Today 2001315102 10.1007/s005950170111 11428603
Reed CT Duma N Halfdanarson T Buckner J Primary neuroendocrine carcinoma of the brain BMJ Case Rep 201912e230582 10.1136/bcr-2019-230582 31537610
Hallet J Law CH Cukier M Saskin R Liu N Singh S Exploring the rising incidence of neuroendocrine tumors: A population-based analysis of epidemiology, metastatic presentation, and outcomes Cancer 201512158997 10.1002/cncr.29099 25312765
Kirshbom PM Kherani AR Onaitis MW Feldman JM Tyler DS Carcinoids of unknown origin: Comparative analysis with foregut, midgut, and hindgut carcinoids Surgery 1998124106370 10.1067/msy.1998.93105 9854584
Lacombe C De Rycke O Couvelard A Turpin A Cazes A Hentic O et al Biomarkers of response to etoposide-platinum chemotherapy in patients with grade 3 neuroendocrine neoplasms Cancers (Basel) 202113643 10.3390/cancers13040643 33562726
Mallory GW Fang S Giannini C Van Gompel JJ Parney IF Brain carcinoid metastases: Outcomes and prognostic factors J Neurosurg 201311888995 10.3171/2013.1.JNS121556 23394337


A rare and challenging case of uterine mass successfully reported in a cervical smear

A 65-year-old postmenopausal woman, gravida 7 and para 5 from a rural hospital, presented with the lower abdominal pain associated with dysuria. A contrast-enhanced computed tomography was done, which showed a mass occupying the uterus and part of the mass is protruding into the upper vagina. A Papanicolaou (Pap) smear test was performed. The images are shown in [Figure 1].

(a) Contrast-enhanced computed tomography scan axial image at the region of the pelvis showed a large heterogeneous enhancing mass occupying the uterus and protruding into the upper vagina (arrows). (b) Cytological cervical smears examination showed tissue fragments of round to oval hyperchromatic nuclei with scant cytoplasm in necrotic debris background (Pap, ×10). (c) Cells with abundant granular to glassy cytoplasm and nuclei with coarse irregularly distributed chromatin and macroprominent nucleoli (Pap, ×40). (d) Large bizarre cells with strap-like, rhabdoid morphology (Pap, ×20).

QUESTION

Q1. What is your diagnosis for the above findings?

Squamous cell carcinoma.

Endocervical adenocarcinoma

Carcinosarcoma

Neuroendocrine carcinoma.

The answer is (c).

Uterine carcinosarcoma, also known as malignant mixed Mullerian tumor (MMMT), is a rare and aggressive form of corpus uteri tumor. The tumor is composed of two cellular components: Epithelial and mesenchymal. The cervical smear findings showed both cellular components which raised the suspicion of carcinosarcoma. Endocervical adenocarcinoma can be a differential; however, the presence of large rhabdoid cells and tissue fragments of round to oval hyperchromatic nuclei with scant cytoplasm cells excluded adenocarcinoma. Squamous cell carcinoma (SCC) was excluded since the spindle cells that are present in this case look different from pleomorphic cells such as “tadpole” and “fiber” cells that are seen particularly in keratinizing type of SCC. Also, the lack of high-grade squamous intraepithelial lesion which did not favor the diagnosis of SCC. Hence altogether, the final report concluded that carcinosarcoma is the most likely correct diagnosis.

Further workup of the case

The patient underwent total hysterectomy with bilateral salpingo-oophorectomy and right and left lymph node. Gross examination of the resected specimen demonstrated a completely occupied endometrial cavity by a large polypoid soft tan mass. Furthermore, the mass extended through the cervix and protruded from the external cervical OS [Figure 2a]. Histopathological examination of the tumor mass revealed an admixture of spindle cells and epithelioid cells. The spindle cell component showed hyperchromatic pleomorphic nuclei and indistinct cell borders. It was admixed with varying proportions of large polygonal cells and strap-like cells. The epithelioid component was comprised of sheets of round cells with moderately hyperchromatic pleomorphic nuclei showing brisk mitosis and scattered spiral-like arterioles. Large areas of necrosis and hemorrhage were also seen. Additional sections of posterior uterine wall showed atrophic endometrium with irregular variably sized glands lined by columnar epithelial cells with marked nuclear atypia and prominent nucleoli [Figure 2b-d].

(a) Gross examination of the total hysterectomy specimen showed a completely occupied endometrial cavity by a large polypoid soft tan mass attached to fundus, anterior and posterior wall and also extends through the cervix. (b) Histological sections of resected uterus showed sarcomatous area (H&E, ×4), (c) serous carcinoma (H&E, ×10), and (d) rhabdomyosarcomatous morphology (H&E, ×10).

ADDITIONAL QUIZ QUESTIONS

Q2. What are the following immunohistochemistry markers will be positive to confirm the diagnosis of carcinosarcoma?

CD56, desmin, α-smooth muscle actin (SMA), MyoD-1, AE1/AE3, and tumor protein 53 (P53)

Synaptophysin, chromogranin, and AE1/AE3

CEA, vimentin, and ER

P16, CK5/6, and P63

The answer is (a).

Immunohistochemical markers on sarcomatous components were positive for cluster of differentiation 56, and desmin and focal positivity for α-SMA and MyoD-1 [Figure 3a-d]. In the malignant epithelial components, AE1/AE3 and P53 showed positivity in more than 80% of the cells in keeping with serous carcinoma [Figure 3e].

Immunohistochemical analysis of uterine carcinosarcoma. (a) The sarcomatous component showed positive for CD56 (×20), (b) Desmin (×10), and (c) partially positive α-Smooth Muscle Actin (α-SMA) muscle actin (×20), and (d) MyoD-1 (×40). (e) The carcinomatous component stained positive for P53 (×10).

Q3. All of following are heterologous component of carcinosarcoma, except for?

Osteosarcomatous

Liposarcomatous

Angiosarcomatous

Leiomyosarcoma

The answer is (d).

BRIEF REVIEW OF THE TOPIC

MMMT, also known as carcinosarcoma, is a rare and aggressive form of corpus uteri tumor. The tumor is composed of two cellular components: Epithelial and mesenchymal which commonly arise from the endometrial cavity.[1] Despite carcinosarcoma cases currently comprising <5% of all uterine tumors,[2] there is an evidence to suggest that there is an increase in the incidence of carcinosarcomas.[3] Hence, it becomes increasingly important for cytologists to be aware of the cytological features for diagnosing this form of tumor. Our cytological findings showed highly atypical epithelial and spindle cells, suggestive of a biphasic tumor type. In addition, a tumor diathesis is reported to be characteristic features of carcinosarcoma in cytological cervical smears.[4,5]

Our cytological features overlapped with previously described reports,[4-6] indicating that carcinosarcoma should always be considered in cases where malignant epithelial and spindly cells are found on a dirty background, especially in women presenting with postmenopausal bleeding.[7] In contrary, other studies have indicated that sarcomatous components are rare findings in cytological smears which make diagnosis of carcinosarcoma difficult.[6,8] A study by Hanley et al. showed that around 72.5% of carcinosarcoma patients had an abnormal Pap smear; however, the majority lacked cytological features consistent with a sarcomatous component.[9] In addition, a study showed that Pap smear test has low overall sensitivity (41.8%), but higher specificity (60.8%) for detecting uterine glandular lesions, which suggest that cytological findings should be confirmed by histological and immunochemical markers.[10]

Uterine carcinosarcoma is a malignancy of two components; epithelial (carcino-) and mesenchymal (-sarcoma), and it can be classified as homologous or heterologous types. The homologous type involves a sarcomatous component of intrauterus such as fibrosarcoma, endometrial stromal sarcoma, and leiomyosarcoma, whereas heterologous component involves cells of extrauterine connective tissues such as rhabdomyosarcoma and chondrosarcoma.[11] In the present case, the malignant epithelial components are positive for P53 and p16 in keeping with serous carcinoma. While the sarcomatous component with rhabdoid differentiation was positive for MyoD-1 and desmin and negative for h-caldesmon, demonstrating a heterologous uterine carcinosarcoma, consistent with previously reported case.[12]

This case was challenging since the Pap smear request was received from a peripheral hospital with limited clinical and radiological details. Furthermore, this tumor commonly occurs in postmenopausal women, with one of the most frequent clinical manifestations being postmenopausal bleeding. However, in this case, the patient presented with the lower abdominal pain and dysuria. Nevertheless, the presence of all the classic cytological features of carcinosarcoma enabled a successful diagnosis of this rare tumor in a Pap smear.

SUMMARY

Carcinosarcoma of the uterine wall may extend to cervix and present in cytological cervical (Pap) smears. Awareness of the possibility facilitates an optimal diagnosis and appropriate management.


References

Akahira JI Tokunaga H Toyoshima M Takano T Nagase S Yoshinaga K et al Prognoses and prognostic factors of carcinosarcoma, endometrial stromal sarcoma and uterine leiomyosarcoma: A comparison with uterine endometrial adenocarcinoma Oncology 20067133340 10.1159/000107107 17690557
Matsuo K Ross MS Machida H Blake EA Roman LD Trends of uterine carcinosarcoma in the United States J Gynecol Oncol 201829e22 10.3802/jgo.2018.29.e22 29400015
Liao CI Caesar MA Lee D Chan A Darcy KM Tian C et al Increasing incidence of uterine carcinosarcoma: A United States cancer statistics study Gynecol Oncol Rep 202240100936 10.1016/j.gore.2022.100936 35169605
Costa MJ Tidd C Willis D Cervicovaginal cytology in carcinosarcoma [malignant mixed mullerian (mesodermal) tumor] of the uterus Diagn Cytopathol 199283340 10.1002/dc.2840080108 1312925
Casey MB Caudill JL Salomão DR Cervicovaginal (papanicolaou) smear findings in patients with malignant mixed müllerian tumors Diagn Cytopathol 200328 2459 10.1002/dc.10276 12722119
Gupta N Dudding N Smith JH Eight cases of malignant mixed müllerian tumor (carcinosarcoma) of the uterus: Findings in SurePath™ cervical cytology Diagn Cytopathol 2014421659 10.1002/dc.22910 22865798
Bongiovanni M Vitale VA Grassi P Mazzucchelli L Malignant mixed müllerian tumor with an extensive squamous-cell carcinomatous component: A potential pitfall in liquid-based cervical cytology Acta Cytol 2014584138 10.1159/000364851 25059755
Snyder MJ Robboy SJ Vollmer RT Dodd LG An abnormal cervicovaginal cytology smear in uterine carcinosarcoma is an adverse prognostic sign: Analysis of 25 cases Am J Clin Pathol 20041224349 10.1309/KUA1FQ3BQRC8YG4V 15362375
Hanley KZ Oprea-Ilies G Ormenisan C Seydafkan S Mosunjac MB Atypical findings on cervicovaginal smears correlate with cervical involvement by malignant mixed müllerian tumors of the uterus Acta Cytol 20155931924 10.1159/000439159 26315658
Bansal B Gupta P Gupta N Rajwanshi A Suri V Detecting uterine glandular lesions: Role of cervical cytology Cytojournal 2016133 10.4103/1742-6413.177156 27014363
Adachi Y Nonogaki H Minamiguchi S Li M Ikehara S Carcinosarcoma of the uterus: A case report Mol Clin Oncol 201645713 10.3892/mco.2016.744 27073665
Seow DY Liquid-based papanicolaou smear: Unusual cytological features of a uterine mass Cytojournal 2020172 10.25259/Cytojournal_89_2019 32256668


Unusual case of parietal scalp swelling without palpable swelling in head and neck region

A 73-year-old woman presented with the swelling in the left parietal region of the scalp, measuring 3 × 3 cm, which was cystic in consistency, and the overlying skin was normal without any palpable swelling in head and neck region. Clinical picture, contrast-enhanced computed tomography (CECT), and cytomorphology of fine-needle aspirate are shown in Figure 1.

(a) Swelling over the left parietal scalp region; (b) contrast-enhanced computed tomography brain shows destructive mildly enhancing bony mass lesion; (c) aspirate from scalp swelling showing round cells with moderate amount of cytoplasm in sheets and groups with mild nuclear atypia (× 400, Papanicolaou stain); and (d) round cells arranged in small groups and clusters with central nuclei and moderate amount of granular cytoplasm (×400, May–Grunwald– Giemsa stain).

QUESTION 1

What is your most probable diagnosis?

Metastatic renal cell carcinoma

Primary adnexal tumor of skin with apocrine differentiation

Granular cell tumor

Metastatic Hurthle cell carcinoma

ANSWER

d. Metastatic Hurthle cell carcinoma.

The scalp swelling was [Figure 1a] on CECT brain revealed a destructive, mildly enhancing bony mass lesion with intra- and extra-cranial extension in the left parietal bone [Figure 1b]. The aspirate from scalp swelling of moderate cellularity showing round cells with central nucleus with nuclear dysplasia in the form of high nucleocytoplasmic ratio, nuclear enlargement, mild-to-moderate pleomorphism, and binucleation [Figures 1c, d and 2a, b]. In addition, some of the groups of cells also had presence of fire flares. The thyroid function test was within normal limits. No mass lesion was seen in kidneys on ultrasound. CECT neck showed few hypodense and calcified nodules in bilateral thyroid lobes. Ultrasound-guided aspirate from thyroid showed similar morphology as seen in scalp swelling [Figure 3a and b]. Final diagnosis was metastatic Hurthle cell carcinoma (HCC) of thyroid.

(a) May–Grunwald–Giemsa stained smear from scalp swelling showing the cohesive sheet of round cells with large central nuclei and moderate granular cytoplasm. Occasional binucleation also noted (×400) and (b) Papanicolaou stained smear showing round cells with moderate amount of granular cytoplasm in sheet with well-defined cell border (×400).

(a and b) Aspirate from thyroid showing the sheet of Hurthle cells with moderate amount of granular cytoplasm in sheet with well-defined cell border with central round nucleus and occasional binucleation (×400).

QUESTION 2

Who gave the description of Hurthle cells?

Max Askanazy

Karl Hurthle

James Ewing

ANSWER

a. Max Askanazy.

HCC is a rare differentiated thyroid carcinoma of follicular origin, and it accounts for approximately 3–7% of all thyroid carcinomas.[1] Max Askanazy, in 1898, described the Hurthle cells/Oncocytic cells in Graves’ disease and Karl Hurthle described the C cells. James Ewing, in 1919, wrote the textbook “Neoplastic Diseases,” described large, granular eosinophilic cells in thyroid carcinoma, and named them Hurthle cells of the thyroid alveoli. Since then, the “Hurthle cell” term has been used in the literature for the cells that were originally described by Max Askanazy, and it is a misnomer.[2]

QUESTION 3

In the recent, the World Health Organization (WHO) classification of Tumors of Endocrine Organs published in 2017, how the HCC is classified?

Variant of follicular carcinoma

Separate entity

HCC has traditionally been classified as a variant of follicular carcinoma of the thyroid, but it was reclassified as a separate entity under the differentiated thyroid carcinomas in the 4th WHO classification of Tumors of Endocrine Organs in 2017, because it has a distinct clinical presentation, pathological features, and genetic profile.[3] The present case is being reported due to the rarity of HCC in the thyroid and even rarer is its primary presentation as scalp swelling without any palpable mass in the thyroid.

BRIEF REVIEW OF THE TOPIC

Hurthle cells are also known as oncocytic/oxyphilic/ eosinophilic cells. Hurthle cell change is observed in a variety of thyroid entities ranging from benign to malignant (including lymphocytic/Hashimoto thyroiditis, nodular goiter, long-standing chronic hyperthyroidism, Hurthle cell adenoma, and HCC), and it is not specific to any single entity.[4] Hurthle cell tumors with malignant behavior show metastasis to the lungs, bones, lymph nodes, and brain.[5] Bone metastasis has been reported in 10–40% of cases, with skull bone metastases only in 2.5–5.8% of cases.[6] Lymph node metastasis is also observed in various studies, ranging from 9% to 20%.[7] Sometimes, the primary presentation of thyroid carcinoma is a metastatic lesion, and the present case is one such rare example. If the cytomorphology shows a follicular pattern along with the classic malignant nuclear features in fine-needle aspiration (FNA) of metastatic sites, diagnosis of HCC on FNA is also possible. HCC can be misdiagnosed as some oncocytic neoplasms from other organs are also present with Hurthle/oncocytic cells with granular cytoplasm, central nucleus, and bland chromatin. In such cases, it is imperative to take into consideration the other clinical and radiological features before making a definitive diagnosis. In cutaneous metastatic lesions, apocrine cells of sweat glands, granular cell tumors, and adnexal tumors of skin with apocrine differentiation may create confusion with Hurthle cells.[8] Granular cell tumor, on cytology, shows tumor cells predominantly in cohesive/syncytial pattern, as well as scattered singly with ill-defined abundant granular cell borders. Tumor cell nuclei show pleomorphism ranging from round/oval to spindle cells. These cells are slightly smaller in size compared to cells of HCC and tumor cells of HCC have well-defined cytoplasm.[9] Cytomorphology of metastatic renal cell carcinoma may show round to oval tumor cells in clusters, sheets, acini, and papillaroid fragments with pale to clear foamy and vacuolated cytoplasm. Some of the cells may show adherence to endothelial cells. There can be mild-to-moderate cellular and nuclear pleomorphism. Foamy cytoplasm on cytology in some cases can help it to differentiate from HCC. However, ancillary studies and radiology are required for making a definite diagnosis.[10] Primary adnexal tumor of skin includes clear cell hidradenoma which shows biphasic cell population comprising of round to polygonal cells with eosinophilic cytoplasm and vesicular nuclei; other population of cells include clear cells containing glycogen with eccentric nuclei.[10] HCC is usually treated with total thyroidectomy and skull metastasis by excision of the lesion.[6] However, there are no consistent guidelines or recommendations for the treatment of skull metastasis.[6] Total surgical clearance of foci of tumor can be achieved with total thyroidectomy and follow-up of the patient for recurrence or metastasis can be done using thyroglobulin marker.[6]

SUMMARY

As discussed in the present case, HCC can present as an isolated metastatic disease without palpable thyroid swelling. In such cases, cytomorphological features can point toward a possible primary in the thyroid and lead to an accurate and timely diagnosis. The case also highlights the importance of cytology, radiology, as well as clinical features to arrive at a definitive diagnosis and appropriate treatment in such rare cases.


References

Harel G Hadar T Segal K Levy R Sidi J Hurthle cell carcinoma of the thyroid gland. A tumor of moderate malignancy Cancer 19865716137 10.1002/1097-0142(19860415)57:8<1613::AID-CNCR2820570829>3.0.CO;2-3 3948132
Caturegli P Ruggere C Karl Hurthle! Now, who was he? Thyroid 2005151213 10.1089/thy.2005.15.121 15753669
LIoyd R Osamura R Kloppel G Rosai J WHO Classification of Tumours of Endocrine Organs 4th ed Lyon International Agency for Research on Cancer 2017
Pu RT Yang J Wasserman PG Bhuiya T Griffith KA Michael CW Does Hurthle cell lesion/neoplasm predict malignancy more than follicular lesion/neoplasm on thyroid fine-needle aspiration? Diagn Cytopathol 2006343304 10.1002/dc.20440 16604553
Stojadinovic A Hoos A Ghossein R Urist MJ Leung DH Spiro RH et al Hurthle cell carcinoma: A 60-year experience Ann Surg Oncol 20029197203 10.1245/aso.2002.9.2.197 11888879
Wong GK Boel R Poon WS Ng HK Lytic skull metastasis secondary to thyroid carcinoma in an adolescent Hong Kong Med J 2002814951
Coca-Pelaz A Rodrigo JP Shah JP Sanabria A Al Ghuzlan A Silver CE et al Hürthle cell carcinoma of the thyroid gland: Systematic review and meta-analysis Adv Ther 202138514464 10.1007/s12325-021-01876-7 34423400
Lallu S Naran S Bethwaite P Fine needle aspiration cytology of unsuspected metastatic hurthle cell carcinoma of the thyroid and its pitfalls: A report of two cases Diagn Cytopathol 20073543943 10.1002/dc.20660 17580356
Chen Q Li Q Guo L Li S Jiang Y Fine needle aspiration cytology of a granular cell tumor arising in the thyroid gland: A case report and review of literature Int J Clin Exp Pathol 20147518691
Chandra S Raina MK Shukla SK Kusum A Nautiyal V Diagnostic rescue of a silent scalp swelling by fine-needle aspiration Cytojournal 20191619 10.4103/cytojournal.cytojournal_40_18 31576200


Prominent emperipolesis in breast lesion: A diagnostic challenge

A middle-aged woman presented with a lump in her left breast for 8 months. She complained of multiple episodes of intense itching in the lesion along with pus discharge in the past 8 months. May-Grunwald-Giemsa and Papanicolaou stained smears are shown in [Figure 1].

(a) Epithelioid cell granuloma with scattered neutrophils (Papanicolaou ×400), (b) Emperipolesis of neutrophils by histiocyte (lower half) and epithelioid cell granuloma (Papanicolaou ×1000), (c) Giant cell, scattered neutrophils, and histiocytes in the background (MGG ×100), (d) Giant cell and histiocytes (MGG ×400).

QUIZ QUESTIONS

Q1. What is the probable diagnosis?

Tuberculosis

Rosai-Dorfman disease

Idiopathic granulomatous mastitis (IGM)

Sarcoidosis

Answer: C.

Idiopathic granulomatous mastitis (IGM)

Cytological diagnosis of granulomatous mastitis is challenging. Differential diagnoses include tuberculosis, fungal infections, sarcoidosis, and histiocytic disorders including Rosai-Dorfman disease. Cytological features of granulomatous mastitis include the presence of epithelioid cell granulomas, multinucleated giant cells, histiocytes with phagocytosed neutrophils, and numerous neutrophils.

Q2. Which type of cell is phagocytosed by histiocytes in IGM?

Lymphocytes

Neutrophils

Eosinophils

All of the above

Answer: D. Studies have shown that the presence of neutrophils is an important diagnostic clue in diagnosing granulomatous mastitis, as in the present case, where histocytes showing emperipolesis of neutrophils along with the presence of numerous neutrophils in the background led to the diagnosis in this case. In contrast, Rosai-Dorfman disease shows emperipolesis of lymphocytes. However, in some cases of IGM, lymphocytes and eosinophils have also been seen in the smear or biopsy.

Q3. Based on various studies, which bacteria is associated with IGM?

Corynebacterium

Pseudomonas

Atypical mycobacteria

All of the above

Answer: D. There are many studies which have identified Corynebacterium as the dominant bacteria associated with IGM. But Streptococcus, Pseudomonas, Atypical Mycobacteria, and Actinomyces have also been identified. Various other etiology of IGM include autoimmunity and abnormal hormonal levels.

Q4. What is the treatment modality for IGM?

Steroid

Immunosuppressive therapy

Surgical excision

All of the above

Answer: D. Patients of IGM have good prognosis with steroids, antibiotics, and immunosuppressive therapy. In some refractory cases, where medical management failed, surgical excision is offered.

BRIEF REVIEW OF THE TOPIC

Granulomatous mastitis is a rare benign inflammatory entity described by Kessler and Wolloch in 1972.[1] It commonly affects young women of childbearing age usually within a few years of lactation.[2,3] The most common presentation includes a painful nodular lump along with induration and redness.[4] The clinical presentation may mimic malignancy. Cytological diagnosis of the entity remains challenging due to overlapping features with many other conditions including tuberculosis, Rosai-Dorfman disease, and even malignancy.[1,5]

Various risk factors were studied, but none were specifically related to granulomatous mastitis. Possible etiological factors include an inflammatory response to epithelial damage, Corynebacterium kroppenstedtii infection, autoimmune disease, foreign body reaction, and a previous history of antipsychotic medications. In addition, the correlation between granulomatous mastitis and breastfeeding and childbirth was also studied.[3,4] The radiological features of the entity are non-specific.[6] The present case did not have a history of medications. She had a history of breastfeeding 8 years ago. Cytological diagnosis of granulomatous mastitis is challenging. Differential diagnoses include tuberculosis, fungal infections, sarcoidosis, and histiocytic disorders including Rosai-Dorfman disease. Cytological features of granulomatous mastitis include the presence of epithelioid cell granulomas, multinucleated giant cells, and numerous neutrophils. The presence or absence of necrosis is not very specific as few studies demonstrated necrosis in all the cases while few studies report necrosis in only a few cases.[2-5,7] Studies have shown that the presence of neutrophils is an important diagnostic clue in diagnosing granulomatous mastitis, as in the present case where histiocytes showing phagocytosed neutrophils along with the presence of numerous neutrophils in the background led to the diagnosis.[2,7,8] The prominence of emperipolesis is not a common feature. Apart from infectious causes, the major differential diagnosis considered was Rosai-Dorfman disease of the breast, which also shows an abundance of histiocytes showing emperipolesis. However, the literature search revealed that the type of cells ingested by histiocytes differs in both cases. In the case of Rosai-Dorfman disease, it is the lymphocytes that are being found in the cytoplasm of histiocytes in contrast to neutrophils which are found in granulomatous mastitis.[8] Other differential diagnoses include sarcoidosis, which shows naked granulomas in the absence of necrosis, which was not the case here. ZiehlNeelsen stain was performed to rule out tuberculosis, which did not reveal any acid-fast bacilli. Management of granulomatous mastitis includes antibiotics such as doxycycline, corticosteroids with or without surgery, or simple follow-up. Few refractory cases have shown improvement with methotrexate, although consensus on the optimal treatment method is lacking.[9]

SUMMARY

We discussed a case of granulomatous mastitis presenting in a middle-aged non-lactating woman with no previous history of psychiatric medications, contraceptive pills, or trauma who presented with a nodular lesion. The cytologic diagnosis is based on finding histiocytes with neutrophils, giant cells, and epithelioid cell granulomas with or without necrosis and excluding other common differential diagnoses such as tuberculosis and fungal infections. As far as IGM is concerned, there is no standard management approach. Therefore, treatment strategies should be tailored to the needs of each patient based on the culture report and response to initial medical management which avoids aggressive management.


References

Kessler E Wolloch Y Granulomatous mastitis: A lesion clinically simulating carcinoma Am J Clin Pathol 1972586426 10.1093/ajcp/58.6.642 4674439
Al-Khaffaf B Knox F Bundred NJ Idiopathic granulomatous mastitis: A 25-year experience J Am Coll Surg 200820626973 10.1016/j.jamcollsurg.2007.07.041 18222379
Johnstone KJ Robson J Cherian SG Cheong JW Kerr K Bligh JF Cystic neutrophilic granulomatous mastitis associated with Corynebacterium including Corynebacterium kroppenstedtii Pathology 20174940512 10.1016/j.pathol.2017.01.006 28442140
Wolfrum A Kümmel S Theuerkauf I Pelz E Reinisch M Granulomatous mastitis: A therapeutic and diagnostic challenge Breast Care (Basel) 2018134138 10.1159/000495146 30800035
Tse GM Poon CS Law BK Pang LM Chu WC Ma TK Fine needle aspiration cytology of granulomatous mastitis J Clin Pathol 20035651921 10.1136/jcp.56.7.519 12835297
Sripathi S Ayachit A Bala A Kadavigere R Kumar S Idiopathic granulomatous mastitis: A diagnostic dilemma for the breast radiologist Insights Imaging 201675239 10.1007/s13244-016-0497-2 27164916
Helal TE Shash LS Saad El-Din SA Saber SM Idiopathic granulomatous mastitis: Cytologic and histologic study of 65 Egyptian patients Acta Cytol 20166043844 10.1159/000448800 27607182
Rajyalakshmi R Akhtar M Swathi Y Chakravarthi R Reddy JB Priscilla MB Cytological diagnosis of RosaiDorfman disease: A study of twelve cases with emphasis on diagnostic challenges J Cytol 2020374652 10.4103/JOC.JOC_4_19 31942098
Martinez-Ramos D Simon-Monterde L Suelves-Piqueres C Queralt-Martin R Granel-Villach L Laguna-Sastre JM et al Idiopathic granulomatous mastitis: A systematic review of 3060 patients Breast J 201925124550 10.1111/tbj.13446 31273861


Pancreatic head mass: To Whipple or not to Whipple

The patient was a 57-year-old female with a 3.8 cm, solid, lobulated, and ill-defined pancreatic head lesion suspected to represent a primary pancreatic malignancy. An endoscopic ultrasound (EUS) was performed and EUS-guided fine needle aspiration (FNA) of the pancreatic mass was obtained with rapid on-site evaluation (ROSE). The direct smears showed the following [Figure 1].

(a) and (b) Fragments of fibrous tissue with dense mononuclear cell infiltrate (Papanicolaou stain, ×20 and ×100, respectively). (c) and (d) Dispersed background lymphomononuclear cells and high-power view of plasma cells. Lymphocytic counts of 27/60X field were identified. Plasma cells were less numerous (8/60X field). (Papanicolaou, ×20 and ×100, respectively). (e) and (f) Ductal epithelium with slight nuclear enlargement and disorganization, consistent with mild atypia (Papanicolaou, ×40 and DiffQuik, ×40, respectively).

QUESTION 1

What is your interpretation?

Negative for malignancy; mucinous cyst debris of uncertain etiology

Positive (for malignancy); malignant glandular and squamous cells consistent with adenosquamous carcinoma

Suspicious (for malignancy); rare markedly atypical epithelial cells suspicious for adenocarcinoma accompanied by fragments of desmoplastic stroma

Atypical; cellular stromal elements with mononuclear cells and mild ductal epithelial atypia.

ANSWER TO QUESTION NUMBER 1

The correct cytologic interpretation is: D. Atypical; cellular stromal elements with mononuclear cells and mild ductal epithelial atypia.

The FNA showed numerous fragments of fibrous tissue with a dense lymphomononuclear cell infiltrate present both within the stromal fragments and in the background. Lymphocytic counts of 27/60X field were identified. Plasma cells were less numerous (8/60X field). Immunocytochemical staining for immunoglobulin G4 (IgG4) performed on air-dried smears was non-contributory. Groups of ductal epithelium with mild nuclear atypia were noted. Material was not available for cell block. The case was signed out as, “Atypical; mild ductal epithelial atypia and chronic fibroinflammatory changes.”

ADDITIONAL DETAILS, FOLLOW-UP AND BRIEF DISCUSSION

The patient denied alcohol consumption but had a remote history of tobacco use. Prior to detection of the lesion, she had visited the emergency room on two occasions over a 3-month period due to epigastric pain accompanied by weight loss and anorexia. She eventually underwent cephalic duodenopancreatectomy. Macroscopic examination of the specimen revealed an ill-defined, tan, 4 cm lesion within the pancreatic head. Histologic sections of the lesion [Figure 2] showed dense lymphoplasmacytic infiltrates with perineural accentuation, storiform fibrosis, and acinar atrophy. Nonobliterative lymphoplasmacytic phlebitis and non-necrotizing obliterative arteritis were observed. Immunohistochemistry revealed over 50 IgG4-positive plasma cells per high power field and an IgG4 to IgG ratio of over 50%. These findings proved histologically highly suggestive of IgG4-related autoimmune pancreatitis (AIP).

(a) Dense lymphoplasmacytic infiltrate with marked perineural accentuation (H&E, ×4). (b) Obliterative arteritis with recanalization and transmural lymphoplasmacytic inflammatory infiltrate (H&E, ×20). (c) Fibrosis exhibiting a storiform-pattern (H&E, ×20). (d) Numerous immunoglobulin G4 (IgG4)-positive plasma cells (IgG4 immunohistochemistry, ×40).

The patient’s serum IgG4 level preoperatively was 85 mg/dL and postoperatively rose to 122 mg/dL. Both values were insufficient to meet the serological diagnostic criterion of 136 mg/dL for IgG4-related disease.[1] However, clinical follow-up was significant for persistent enlargement of abdominal lymph nodes as well as elevated alkaline phosphatase and gamma-glutamyl transferase, presumed to represent IgG4-related lymphadenopathy and cholangiopathy. Prednisone and methotrexate therapy were initiated with subsequent resolution of lymphadenopathy and normalization of liver enzymes. The patient is asymptomatic 2 years after surgery.

It is important to be aware of mass-forming inflammatory lesions of the pancreas such as AIP which may clinically and radiologically simulate pancreatic malignancy sometimes leading to unnecessary surgical resection. Although EUS guided FNA (EUS-FNA) is often a useful adjunct in the diagnosis of pancreatic lesions, allowing for correct classification of many cases, the preoperative diagnosis of AIP based on EUS-FNA is challenging, and no widely accepted cytologic criteria have been reported.[2,3]

Furthermore, benign features suggestive of AIP do not rule out false negative results induced by sampling error.[4]

ADDITIONAL QUIZ QUESTIONS

Q2. All of the following are suggestive of Type 1 AIP over Type 2 AIP, EXCEPT:

Systemic disease

Frequent relapses

Swift response to immunosuppression

Inflammatory bowel disease (IBD).

Q3. All are pathological characteristics of Type 1 AIP, EXCEPT:

Lymphoplasmacytic inflammation with storiform fibrosis

Preferential involvement of pancreatic tail

IgG4+/IgG+ plasma cell ratio of >40%

Obliterative phlebitis.

Q4. Which of the following is true regarding the cytomorphologic findings of Type 1 AIP:

Fragments of cellular fibrous stroma are common

Ductal epithelial atypia precludes diagnosis of AIP

Cytologic findings are highly specific and alone are sufficient for a definitive diagnosis

On FNA, AIP is indistinguishable from chronic pancreatitis, NOS.

ANSWERS TO ADDITIONAL QUIZ QUESTIONS AND BRIEF REVIEW OF THE TOPIC

Q2. (d); Q3. (b); Q4. (a).

Q2. (d) AIP is a pancreatobiliary-centric inflammatory disease, typically marked by corticosteroid responsiveness and classified in two groups. Type 1 AIP often affects elderly males, associates extrapancreatic manifestations of systemic IgG4-related disease (including cholangitis, sialadenitis, retroperitoneal fibrosis, or lymphadenopathy), shows frequent relapses and elevation of serum IgG4 in approximately 80% of cases.[5] In contrast, Type 2 AIP is considered to be an isolated pancreatic disorder and does not show elevation of serum IgG4. Recurrence is rare. Interestingly, Type 2 AIP is linked to inflammatory bowel disease (ulcerative colitis or Crohn’s disease), seen in up to 16–30% of Type 2 AIP cases.[5,6]

Q3. (b) AIP most often involves the head of the pancreas.[7] Type 1 AIP is histologically characterized by a dense lymphoplasmocytic infiltrate, storiform fibrosis, obliterative phlebitis, and increased numbers of IgG4 positive plasma cells, typically >50/high-power field, and IgG4+/IgG+ ratio of >40%.[8] Lymphoplasmacytic arteritis and a preferentially perineural distribution of inflammation have also been described.[8,9] Type 2 AIP is histologically characterized by periductal inflammation with granulocytic epithelial lesions and relative paucity of IgG4-positive plasma cells.[10]

Q4. (a) EUS-FNA alone is widely considered to be insufficient to make a definitive diagnosis of AIP, probably at least partially due to a lack of architectural integrity.[11] In fact, International Consensus Diagnostic Criteria for AIP (2011) list core/surgical biopsy specimens as preferable for diagnosis of AIP.[12] Despite this, certain cytological findings in conjunction with clinicoradiological clues can aid in establishing a diagnosis AIP in certain cases. A recent retrospective review of AIP FNA results found that cellularity of stromal fragments was significantly higher in AIP than in the control group.[2] Furthermore, stromal fragments with embedded lymphocytes (>30/60x) were seen in almost 40% of AIP cases versus 0% in chronic pancreatitis, NOS.[2] Other authors have found that AIP is often reported as “atypical” on FNA, probably due to ductal epithelial atypia secondary to surrounding inflammatory and fibrotic changes.[4] Performance of IgG4 immunolabeling on the cell block material and/or pre-operative identification of elevated serum IgG4 levels may provide valuable information. Unfortunately, as occurred in the present case, not all cases of Type 1 AIP demonstrate elevated serum IgG4 and cell block material was not available for evaluation.

SUMMARY

Awareness of AIP’s potential to clinically and radiologically simulate pancreatic malignancy is important to avoid unnecessary surgery.

Although AIP on FNA lacks specific cytomorphologic features, cellular stromal fragments, mild ductal epithelial atypia and prominent lymphocytic infiltrate are common findings and may support the diagnosis in the proper context.

Corticosteroid responsiveness and elevated serum IgG4 are useful clues which, in conjunction with suggestive cytomorphology, may reduce gratuitous duodenopancreatectomy.


References

Umehara H Okazaki K Kawa S Takahashi H Goto H Matsui S et al The 2020 revised comprehensive diagnostic (RCD) criteria for IgG4-RD Mod Rheumatol 20213152933 10.1080/14397595.2020.1859710 33274670
Deshpande V Mino-Kenudson M Brugge WR Pitman MB Castillo CF Warshaw AL et al Endoscopic ultrasound guided fine needle aspiration biopsy of autoimmune pancreatitis: Diagnostic criteria and pitfalls Am J Surg Pathol 200529146471 10.1097/01.pas.0000173656.49557.48 16224213
Yoon SB Moon SH Song TJ Kim JH Kim MH Endoscopic ultrasound-guided fine needle aspiration versus biopsy for diagnosis of autoimmune pancreatitis: Systematic review and comparative meta-analysis Dig Endosc 202133102433 10.1111/den.13866 33030283
Holmes BJ Hruban RH Wolfgang CL Ali SZ Fine needle aspirate of autoimmune pancreatitis (lymphoplasmacytic sclerosing pancreatitis): cytomorphologic characteristics and clinical correlates Acta Cytol 20125622832 10.1159/000336135 22555522
Shinagare S Shinagare AB Deshpande V Autoimmune pancreatitis: A guide for the histopathologist Semin Diagn Pathol 201229197204 10.1053/j.semdp.2012.07.007 23068298
Tsen A Alishahi Y Rosenkranz L Autoimmune pancreatitis and inflammatory bowel disease: An updated review J Clin Gastroenterol 20175120814 10.1097/MCG.0000000000000737 27779612
Raina A Yadav D Krasinskas AM McGrath KM Khalid A Sanders M et al Evaluation and management of autoimmune pancreatitis: Experience at a large US center Am J Gastroenterol 20091042295306 10.1038/ajg.2009.325 19532132
Deshpande V Zen Y Chan JK Yi EE Sato Y Yoshino T et al Consensus statement on the pathology of IgG4-related disease Mod Pathol 201225118192 10.1038/modpathol.2012.72 22596100
Farris AB 3rd Basturk O Adsay NV Pancreatitis, other inflammatory lesions, and pancreatic pseudotumors Surg Pathol Clin 2011462550 10.1016/j.path.2011.03.004 26837491
Chari ST Kloeppel G Zhang L Notohara K Lerch MM Shimosegawa T Histopathologic and clinical subtypes of autoimmune pancreatitis: the Honolulu consensus document Pancreatology 20101066472 10.1159/000318809 21242705
Majumder S Chari ST EUS-guided FNA for diagnosing autoimmune pancreatitis: Does it enhance existing consensus criteria? Gastrointest Endosc 2016848057 10.1016/j.gie.2016.05.046 27742043
Shimosegawa T Chari ST Frulloni L Kamisawa T Kawa S Mino-Kenudson M et al International consensus diagnostic criteria for autoimmune pancreatitis: Guidelines of the International Association of Pancreatology Pancreas 2011403528 10.1097/MPA.0b013e3182142fd2 21412117


Inguinal swelling in a young female: An unusual finding

A 21-year-old female presented with gradually increasing swelling over right inguinal region for the past 1 month. She denied any history of leg trauma, pain, or fever. On physical examination, a firm, non-tender nodular mass lesion measuring 1 × 0.5 × 0.5 cm was palpable over the right inguinal region. No other lymphadenopathy or nodular swelling was found elsewhere in the body. Fine needle aspiration was performed from the lesion, after informed consent of the patient and smears were prepared [Figures 1 and 2].

Microphotograph panel of smears from the right inguinal swelling are cellular and show (a) large fragments of bladder wall and (b and c) granular fibrillary sheath-like material in a background of histiocytes, scattered eosinophils, neutrophils. (d and e) rounded bladder wall fragments tiny dense pyknotic nuclei. (f) scattered multinucleated giant cell. (a: ×4, b-d: ×10, e and f: ×40) (H&E).

1. What is the interpretation based on the clinical history and findings of fine needle aspiration?

Hydatidosis

Cysticercosis

Spargana

Filariasis.

ANSWER

The correct cytological interpretation is b. Cysticercosis.

EXPLANATION

Aspirate was fluidy in this case and the smears showed large fragments of bladder wall identified as fibrillary bluish material woven into rounded multiple subcuticular cells with interspersed tiny blue pyknotic nuclei surrounded by dense inflammatory infiltrate. The background showed sheets of histiocytes, scattered eosinophils, neutrophils, and few multinucleated giant cells [Figures 1 and 2]. These features are characteristic of Cysticercosis. Cysticercosis is caused by larval stage of Taenia solium, while adult form causes Taeniasis.

Microphotograph panel of smears (a and b) showing large fragments of bladder wall in a background of dense inflammatory infiltrate (c and d) fibrillary bluish material woven into rounded multiple subcuticular cells with interspersed tiny blue pyknotic nuclei surrounded by dense inflammatory infiltrate. (a-c: ×10, d: ×40) (MGG).

The most common cytologic mimic of cysticercosis is other cestodes including hydatid cyst (Echinococcus granulosus), Coenuri and Spargana. The differentiation between these requires careful examination of the bladder wall and hooklets. The bladder wall is thin and membranous in cysticercosis while thick and lamellated in a hydatid cyst. The hooklets of cysticercosis are sized 130–170 m while hydatid hooklets range between 22 m and 44 m.[1]

Filariasis (Option D) is another vector borne disease and shows microfilariae. Most common causative agents of human filariasis include Wuchereria bancrofti, Brugia malayi, Brugia timori, and Loa loa.

ADDITIONAL QUIZ QUESTIONS

Q2. What is the mode of transmission of the disease?

Feco-oral contamination

Droplet transmission

Inhalation

Sexual contact.

Q3. All of the following statement are true for cysticercosis, except?

Cysticercosis is caused by larval stage of T. solium

Humans cannot be both definitive and intermediate hosts

Inflammatory response is usually associated with cysticercosis

Viable cysticerci can persist for several months to years in humans.

ANSWERS TO THE ADDITIONAL QUIZ QUESTIONS

Answers: Q2-a, Q3-b.

EXPLANATION FOR ADDITIONAL QUIZ QUESTIONS

The life-cycle of T. solium involves two hosts – the intermediate hosts which are most commonly pigs and harbor the larval stage, and the definitive host which are humans, sheltering the adult form. However, humans can become accidental intermediate hosts. The adults tapeworm enters the human body by eating the larval form present in the meat of the intermediate host.[2]

BRIEF REVIEW OF THE TOPIC

A myriad of parasitic infections continue to pose health issues in tropical climates. In endemic countries, like India, cysticercosis is a common infection. The WHO experts in 2014, ranked T. solium as food borne parasite of greatest global concern.[2] Cysticercosis can involve skin, subcutaneous tissue, muscles, intermuscular fascia, lymph nodes, brain, and various other organs.

Most subcutaneous cysticerci present as painless nodular or cystic swellings, that are easily amenable to fine needle aspiration. A definitive diagnosis of cysticercosis can be established by cytological examination and identification of the variable morphology in viable and degenerating/ calcified lesions. Microscopically, viable cysts demonstrates fragments of the bladder wall of the larva identified as loose granular fibrillary material, often thrown into rounded wavy folds with small round dark subcuticular or tegumental cells with small pyknotic nuclei, refractile claw shaped hooklets, and scolices, while degenerating lesions can demonstrate scattered hooklets and calcareous corpuscles.[3,4] Background consists of inflammatory infiltrate comprising of eosinophils, polymorphs, histiocytes, and giant cell reaction.

Cysticercus cellulosae can regulate T-Cell response and interacts with the host immune system by excreting and secreting antigens, thereby escaping the host immune attacks and establish a persistent infection.[5]

Serological tests and radiological investigations such as computed tomography scan and magnetic resonance imaging are also sensitive for diagnosing cysticercosis. Lentil lectin glycoprotein enzyme linked immunoelectrotransfer blot assay is the assay of choice for serodiagnosis.[6]

SUMMARY

Cysticercosis is an infection caused due to larval stage of the parasite T. solium. The clinical presentation is highly variable ranging from nodules occurring in the skin, subcutis, muscles, intermuscular fascia, and various other organs including the central nervous system. The index case emphasizes the awareness of possibility of parasitic infestation during the evaluation of isolated nodular and cystic swellings yielding fluidy or necrotic material on fine needle aspiration.


References

Bhardwaj S Rather G Fine needle aspiration cytology of cysticercosis: A study of 30 cases J Cytol 2019361821 10.4103/JOC.JOC_77_17 30745734
World Health Organization Multicriteria-based ranking for risk management of food-born parasites Microbiological Risk Assessment Series Geneva World Health Organization 201423
Rajwanshi A Radhika S Das A Jayaram N Banerjee CK Fine-needle aspiration cytology in the diagnosis of cysticercosis presenting as palpable nodules Diagn Cytopathol 199175179 10.1002/dc.2840070515 1954832
Kala P Khare P Fine-needle aspiration cytology as a diagnostic modality for cysticercosis: A clinicocytological study of 137 cases J Cytol 2014316872 10.4103/0970-9371.138665 25210232
Fan X Zhang Y Ouyang R Luo B Li L He W et al Cysticercus cellulosae regulates T-cell responses and interacts with the host immune system by excreting and secreting antigens Front Cell Infect Microbiol 202111728222 10.3389/fcimb.2021.728222 34540719
Rodriguez S Wilkins P Dorny P Immunological and molecular diagnosis of cysticercosis Pathog Glob Health 201210628698 10.1179/2047773212Y.0000000048 23265553


Cervical lymphadenopathy with dual pathology: Interesting finding

A 48-year-old male presented with a rapidly enlarging painless firm to hard, mobile, and non-tender lymph node approximately 2 cm deep in the left level II cervical region. Fine needle aspiration of the lymph node was performed (Figure 1).

(a) Cellular smear showing discrete population of round to polygonal cells with pyknotic nuclei and dense cytoplasm with smoothly curved filarial worm covered with a hyaline sheath and a pointed tail. (b) Higher magnification showing worm along with tumour cells with squamoid differentiation. (c) Cellular smear showing discrete population of tumor cells having squamoid appearance and of transverse section of gravid female worm with numerous ovoid eggs. (d) Higher magnification of smoothly curved worm covered with a hyaline sheath and a pointed tail, devoid of nuclei. (a) MGG ×10, (b) MGG ×40, (c) H&E ×10, (d) H&E ×40.

Q1. What is your interpretation?

Metastatic squamous cell carcinoma with microfilaria of Wuchereria bancrofti

Metastatic adenocarcinoma with microfilaria of W. bancrofti

Metastatic squamous cell carcinoma with microfilaria of Brugia malayi

Metastatic adenocarcinoma with microfilaria of B. malayi

Answer: a. Metastatic squamous cell carcinoma with microfilaria of Wuchereria bancrofti

The cytological findings are consistent with metastatic squamous cell carcinoma with accompanying microfilaria in a necro-haemorrhagic background. The filarial worm is smoothly curved, covered with a hyaline sheath and a pointed tail. The body has multiple discrete evenly spaced nuclei with no terminal nuclei in the tail. The microfilaria is identified as Wuchereria bancrofti owing to characteristic smoothly curved worm with pointed tail, devoid of nuclei (Figure 1)

Table 1 elaborates the key differentiating features of commonly occurring sheathed filarial nematodes.

Key differentiating features of commonly occurring sheathed filarial nematodes.

Nematode Wuchereria bancrofti Brugia malayi Loa loa
Length 244–296 µ 177–230 µ 231–250 µ
Cephalic space Short Long Short
Tail Pointed tail, devoid of nuclei Two distinct nuclei on tail tip Continuous discrete row of nuclei at the tail tip
Periodicity Nocturnal Nocturnal Diurnal

Q2. Which of the following is not a characteristic feature of W. bancrofti?

Pointed tail, devoid of nuclei

Two distinct nuclei on tail tip

Cephalic space is short

Length of microfilaria ranges from 244 to 296 µ.

Answer: b

Q3. What could be the possible reason for lodgement of filaria in the metastatic sites?

Increased vascularity of the tissues

Transmigration of microfilaria along with metastatic tumor emboli

Decreased host immune response

All of the above.

Answer: d

BRIEF REVIEW OF THE TOPIC

Filariasis is a vector borne public health issue and is endemic all over India. In India, W. bancrofti accounts for majority of the total filarial infections.[1] Filarial nematodes can be detected in peripheral blood, body fluids, urine, sputum, lymph node, thyroid, parotid, scrotum, and subcutaneous tissues along with a wide variety of metastatic sites.[2-4] Causative agents of human filariasis include W. bancrofti, B. malayi, Brugia timori, Loa loa, Mansonella perstans, Mansonella ozzardi, Mansonella streptocerca, and Onchocerca volvulus. Of all of the human filarial nematodes, W. bancrofti has the widest geographic distribution.[5] The existence of filarial nematodes in association with metastatic deposits could be coincidental or may involve a different pathogenetic mechanisms. Filarial parasites exert profound immunoregulatory effects on the host immune system with both parasite-antigen specific and more generalized levels of immune modulation.[6] The immune-modularity effects of microfilaria can be responsible for delayed post-operative healing after neck dissection.

Few authors have speculated that increased vascularity of the tissues and transmigration of microfilaria along with metastatic tumor emboli, decreased host immune response could be a possible reasons for lodgement of filarial in the metastatic sites.[3,7,8]

SUMMARY

This case highlights the importance of diligent screening of all cytology smears in endemic regions as the occurrence of microfilaria in cervical nodes of patients with oral cavity malignancy requiring neck dissection can have different implications in post-operative healing.


References

Park K Epidemiology of communicable diseases Park's Textbook of Preventive and Social Medicine 26th ed Jabalpur, India Bhanot Publishers 2021
Kishore M Kumar V Kaushal M Gomber A Cytology of achylous hematuria: A clue to an underlying uncommon clinical scenario Cytojournal 20181530 10.4103/cytojournal.cytojournal_11_18 30662516
Pantola C Kala S Agarwal A Khan L Microfilaria in cytological smears at rare sites coexisting with unusual pathology: A series of seven cases Trop Parasitol 20122613 10.4103/2229-5070.97244 23508347
Rekhi B Kane SV Microfilariae coexisting with a follicular lesion in thyroid aspirate smears in an uncommon case of a retrosternal thyroid mass, clinically presenting as malignancy Cytojournal 201184 10.4103/1742-6413.76732 21383959
Mathison BA Couturier MR Pritt BS Diagnostic Identification and differentiation of microfilariae J Clin Microbiol 201957e0070619 10.1128/JCM.00706-19 31340993
Babu S Nutman TB Immunology of lymphatic filariasis Parasite Immunol 20143633846 10.1111/pim.12081 24134686
Vij M Kumari N Krishnani N Microfilaria in liver aspiration cytology: An extremely rare finding Diagn Cytopathol 2011395212 10.1002/dc.21483 21695804
Gahine R Das A Jain VK Agrawal A Rare association of microfilaria with poorly differentiated mucin-secreting metastatic carcinoma in liver aspirate cytology BMJ Case Rep 201912e231618 10.1136/bcr-2019-231618 31645405


Granulomas on cervical Pap smear: “Forget me not”

A 62-year-old female presented with the complaints of heaviness in the lower abdomen for 1 year. She was post-menopausal from the past 15 years. No prior history of fever or cough was present. On gynecological examination, her uterus was anteverted and cervix showed mild erosion. Routine Papanicolaou test was done and stained smears showed features as shown in Figure 1.

(a and b) Smears show well-formed collection of epithelioid cells forming granulomas (red arrow). The adjacent area shows parabasal cells and inflammation comprising of neutrophils and lymphocytes. (×100, Papanicolaou [PAP] stain); (c) Smear shows a multinucleated giant cell (red arrow) (×100, PAP stain); (d) Smear shows a beaded rod-shaped acid-fast bacillus (red arrow) (×1000 oil immersion, Ziehl Neelson stain).

1. What is your probable diagnosis?

Sarcoidosis

Granuloma Inguinale

Tuberculosis

Syphilis.

Answer to Question 1: (c)

The granulomas [Figure 1a and b] and multinucleated giant cell [Figure 1c] can be seen in all of the above conditions; however, positive acid-fast bacilli on Ziehl Neelson stain (ZN) are a characteristic feature of Mycobacterium tuberculosis.

Tuberculous bacilli are seen as a pink rod shaped slender bacilli on ZN stain [Figure 1d].

2. What is the mode of spread for cervical tuberculosis?

Hematogenous spread

Lymphatic spread

Primary infection

All of the above.

Answer to Question 2: (d)

Cervical tuberculosis (TB) can spread by any route – Hematogenous spread, lymphatic spread, or direct extension from the primary focus in the body. Rarely, primary route can be the cause of infection introduced by the male partner suffering from TB of genitourinary tract.

3. Which stain is used for the confirmation of TB infection?

Periodic Acid Schiff stain

Ziehl Neelson stain – 20%

Ziehl Neelson stain – 05%

Ziehl Neelson stain – 01%.

Answer to Question 3: (b)

The concentration of sulfuric acid varies in the ZN stain. About 20% of acid is used for M. tuberculosis, 5% of acid is used for Mycobacterium leprae, and 1% of acid is used for actinomyces and nocardia.

REVIEW OF THE TOPIC

TB is a significant cause of high morbidity and mortality and is more common in developing countries. TB of the female genital tract is a rare disease and more commonly involves the upper genital tract (fallopian tubes and endometrium) as compared to lower genital tract.[1,2] TB of cervix is very rare as the stratified squamous epithelium is resistant to the tubercular bacilli.[3] Cervical TB accounts for 0.1–0.65% of all the TB cases and 5–24% of the genital tract TB cases.[1,2] The incidence of TB is on rise.[1] It can occur at any age but commonly affects women of reproductive age group.[4,5] The two common organisms responsible for genital TB are M. tuberculosis or Mycobacterium bovis.[1] Genital tract TB usually occurs from the hematogenous spread of infection from the primary focus. Cervical TB almost always occur secondary to TB of fallopian tube or endometrium and is typically associated with pulmonary TB.[1,2] It can occur by either hematogenous spread, lymphatic spread, or by direct extension from the primary focus.[1,6,7] Rarely, primary route can be the cause of infection introduced by the partner suffering from TB of genitourinary tract.[2,4] Chowdhury suggested that sputum may also be source of infection when it is used as a sexual lubricant.[4] The symptoms of genital TB can range from constitutional symptoms to infertility.[1,2] The common presentations include abdominal pain, menstrual irregularities, discharge per vaginum, postmenopausal bleeding, etc.[2] Grossly TB can present as papillary or vegetative growth, multiple tiny nodules or ulcer, and on imaging studies (hysterosalpingography or ultrasonography) of cervix, there can be seen diverticular outpouching of cervix with feathery appearance or cervical distortion etc.[1,3] These features can be easily misinterpreted as cervical carcinoma.[1] Microscopic examination of a cervical biopsy is characterized by well-formed epithelioid cell granulomas which may be associated with multinucleated Langhans type giant cell reaction and caseation necrosis. Lymphoplasmacytic infiltrates are seen at the rim of granulomas.[2] TB of cervix needs to be differentiated from other granulomatous diseases, for example, lymphogranuloma venereum, sarcoidosis, schistosomiasis, amoebiasis, brucellosis, foreign body granuloma due to suture, crystal or cotton, and carcinoma cervix.[2,8-11] The confirmation can be done by demonstration of acid fast bacilli of TB on ZN staining or isolation of mycobacterium.[2] However, ZN stain and culture may be negative in one-third of the cases. In these cases, identification of typical granulomas is sufficient for the diagnosis of TB, if other causes of granulomatous cervicitis are excluded from the study.[1,2] Newer diagnostic techniques such as enzyme linked immunosorbent assay or polymerase chain reaction may aid in the diagnosis of TB.[8,9] The treatment includes anti tubercular therapy. The granulomas disappear after the treatment; however, fertility is low in patients even after the treatment due to subsequent healing by fibrosis and adhesions.[4,5] The best method of preventing TB is Bacillus of Calmette Guerin vaccination and healthy lifestyle.[5]

SUMMARY

Genital TB is a chronic disease in females with low-grade symptoms. It commonly involves fallopian tubes and endometrium. Cervix is an uncommon location for genitourinary TB. The gross appearance and imaging studies of cervical TB may mislead as cervical carcinoma. Therefore, microscopic examination and positive ZN stain and culture are diagnostic. TB may lead to fibrosis and adhesion and is a major cause of infertility among females. Therefore, screening for genital TB must be a part while evaluating for the menstrual irregularities or infertility. Early diagnosis and further treatment are the key to avoid the complications associated with TB.


References

Paprikar M Biswas M Bhattacharyav S Sodhi B Mukhopadhyay I Tuberculosis of cervix Med J Armed Forces India 2008642978 10.1016/S0377-1237(08)80129-3 27408176
Singh S Gupta V Modi S Rana P Duhan A Sen R Tuberculosis of uterine cervix: A report of two cases with variable clinical presentation Trop Doct 2010401256 10.1258/td.2009.090423 20305116
Lamba H Byrne M Goldin R Jenkins C Tuberculosis of the cervix: Case presentation and a review of the literature Sex Transm Infect 200278623 10.1136/sti.78.1.62 11872864
Chowdhury NN Overview of tuberculosis of the female genital tract J Indian Med Assoc 1996943456 345-6, 361
Lester CW Tuberculosis of uterine cervix Am J Surg 19363357480 10.1016/S0378-4320(36)90030-1
Gupta B Shree S Rajaram S Goel N Genital tuberculosis: Unusual presentations Int J Mycobacteriol 201653579 10.1016/j.ijmyco.2016.06.017 27847026
Moussa B Valentin K Adama O Aziz DA Idrissa Z Goumbri LO Tuberculosis of the uterine cervix: About a case and literature review Open J Obstet Gynecol 201667349 10.4236/ojog.2016.612091
Grace GA Devaleenal DB Natrajan M Genital tuberculosis in females Indian J Med Res 201714542536
Kesharwani H Mohammad S Pathak P Tuberculosis in the female genital tract Cureus 202214e28708 10.7759/cureus.28708
Kamal MM The Pap smear in inflammation and repair Cytojournal 20221929 10.25259/CMAS_03_08_2021 35673696
Gupta D Kaushal M Jain S Epithelioid cell granulomas in urine cytology smears: A diagnostic approach Cytojournal 20221934 10.25259/Cytojournal_82_2020 35673699


Duct tales of a parotid gland swelling

An Indian male in his mid-30s complaining of a progressively enlarging right parotid swelling for the past 5 years, with a history of exposure to tuberculosis from a close contact. The ultrasound (USG) report suggests a retention cyst, measuring 5.2 × 4.4 × 2 cm. Direct fine-needle aspiration (FNA) yielded 1 mL of thick, yellowish, turbid fluid, and swelling was reduced slightly after aspiration [Figure 1a-c].

(a and b) Direct FNA smears show cells in papillaroid architecture in an abundant proteinaceous background (MGG, a: ×400, b: ×1000). (c) Large cohesive sheet of benign looking cells, showing oncocytic changes and minimal nuclear irregularity (MGG, ×4000).

QUESTION # 1

What is the most likely interpretation of FNA using the Milan system? [Figure 1a-c]

Non-neoplastic

Atypia of undetermined significance

Neoplasm: Benign

Neoplasm: Salivary gland neoplasm of uncertain malignant potential (SUMP)

Malignant.

ANSWER TO QUESTION # 1

c. Neoplasm: Benign.

FNA smears were cellular and showed benign-looking epithelial cells arranged in large cohesive sheets, clusters, and groups. They exhibit mild degenerative nuclear changes and abundant eosinophilic granular cytoplasm. Background showed abundant eosinophilic proteinaceous material along with a few neutrophils, lymphocytes, macrophages, and debris. There was no evidence of mitosis, necrosis, or atypia in the smears examined. To interpret and report the case, “The Milan System for Reporting Salivary Gland Cytopathology” was used; the cytomorphological features were suggestive of Category IV A – Benign Salivary Gland Neoplasm.

On inquiry, the patient showed a previous (3 months prior) USG report and fine-needle aspiration cytology (FNAC), which were done outside. The USG report was suggestive of a retention cyst and the FNAC was consistent with the radiological findings of a “Retention cyst.”

QUESTION # 2

An excisional biopsy was performed after 12 days of FNAC. On the basis of gross and microscopic findings, what is the diagnosis? [Figure 2a-d]

Cystadenoma

Sclerosing polycystic adenosis

Acinic cell carcinoma (papillary cystic pattern)

Mammary analog secretory carcinoma

Low-grade intraductal carcinoma.

(a) Gross specimen of superficial parotidectectomy show solid cystic cut surface. (b) Cystically dilated ducts with tufted, micropapillary anastomosing proliferations of epithelial ductal cells; tumor area with normal serous salivary gland (H&E, ×200). (c and d) Intraductal proliferations with Roman Bridges and “pseudocribriform” papillary architecture of tumor cells, displaying mild to moderate pleomorphism (H&E, c: ×1000, d: ×4000).

ANSWER TO QUESTION # 2 AND FOLLOW-UP OF CASE

e. Low-grade intraductal carcinoma.

There was no evidence of necrosis grossly. Multiple H&E stained sections examined under the microscope showed a salivary gland neoplasm consisting of solid and cystic areas. Cysts showed intracystic and intraductal proliferation of neoplastic epithelial cells arranged in papillary, micropapillary, and “pseudocribriform” architecture, displaying false punched out spaces and “Roman bridge” formation. The cysts cavities were filled with proteinaceous secretions. Tumor cells exhibit mild-to-moderate pleomorphism, vesicular chromatin, inconspicuous 1–2 nucleoli, and a moderate to abundant amount of eosinophilic cytoplasm. At places, a few cells showed apocrine changes. Myoepithelial cells were noted. Adjoining stroma showed areas of hemorrhage and lymphoid cell proliferation, well demarcated from normal salivary gland acini. Tiny foci of necrosis were identified; however, an invasion was not seen. Mitosis was exceedingly rare. No lymphovascular or perineural invasion was noted. Surgical margins were free of tumor cells. Four intraparotid lymph nodes identified microscopically showed features of reactive lymphoid hyperplasia.

To conclude the diagnosis, whole tissue was processed to screen for invasion, which was not identified on the total of 19 sections passed. Thereafter, a diagnosis of “Low-grade-Intraductal Carcinoma” was awarded. Immunohistochemistry (IHC) was performed by application of S100, which came out negative for tumor cells.

Post-operative course

The post-operative course of the patient was fair, and facial nerve palsy was not noted. No swelling recurs at the surgical site or in the nearby regional area on physical examination or radiological investigation, stating that, there is no recurrent disease 12 months after surgery.

QUESTION # 3

What is/are the diagnostic feature of low-grade intraductal carcinoma?

“Pseudocribriform” architecture displaying false punched-out spaces and “Roman bridge” formation without invasion

Intracystic and intraductal proliferation of neoplastic epithelial cells arranged in a papillary and micropapillary pattern with invasion

Absence of S100 IHC positivity

Lymphovascular or perineural invasion

Absence of myoepithelial cells and presence of rare mitosis.

ANSWER TO QUESTION # 3

a. “Pseudocribriform” architecture displaying false punched-out spaces and “Roman bridge” formation without invasion.

QUESTION # 4

What are the major pitfalls observed in the cytohistomorphological correlation of this case?

Gross aspiration of thick yellow fluid, followed by slight reduction of size of the swelling

USG findings suggestive of retention cyst

Long-standing history of 5 years

Murky fluid in background and no evidence of atypia/malignancy noted

All of the above.

ANSWER TO QUESTION # 4

e. All of the above.

This case was mislabeled as a retention cyst on USG and was unrecognized and under-diagnosed on FNAC done outside in a private laboratory despite being an extremely rare entity. On both occasions, FNA procedures done outside as well as in our department yielded similar fluidy aspirates [Table 1].

The clues or features favoring on FNAC performed in our laboratory are as follows.

Neoplastic etiology Benign in nature/against malignant etiology
1. High cellularity 1. Fluid aspiration on two attempts
2. Swelling size 2. Size mildly reduced after aspiration
3. Progressive 3. Cohesive cell arrangement
4. Fixed and firm in consistency 4. No definite atypia
5. Non-tender, no signs of inflammation 5. No mitosis
6. No necrosis
7. Murky fluid or proteinaceous background
8. No hemorrhage
9. Ultrasound report

FNAC: Fine-needle aspiration cytology

Although making a definite diagnosis of this tumor on FNA smears is challenging, at least can be suggested as salivary gland neoplasm. The Category IV A, B, and C of Milan’s system is highly overlapping. The presence of above-mentioned cytomorphological findings is of a salivary gland multicystic neoplasm; however, exact diagnosis is almost impossible. However, raising the diagnostic possibility of salivary gland neoplasm rather than just a cyst in a pre-operative FNA would be of great help to the surgeon so that the complete resection was planned. The distinguished nomenclature relies solely on histopathological examination of stromal invasion that cannot be evaluated on pure cytological grounds.

Warthin’s tumor was one of the differential diagnoses considered due to the classical background (consisting of few lymphocytes) and cystic nature of the tumor, as well as the few oncocytic looking cells; however, the florid lymphoid background was absent in our case. The lymphocytic background is sometimes very misleading, as in the present case, as hitting an intrasalivary gland lymph node gives a similar picture on a smear, even when associated with oncocytic looking cells.

QUESTION # 5

What are the most obvious biopsy findings describing the present case’s prognosis?

Cystic areas

Mild atypia

Absence of necrosis

Negative surgical margins and absence of stromal invasion

All of the above.

ANSWER TO QUESTION # 5

d. Negative surgical margins and absence of stromal invasion.

BRIEF REVIEW OF THE TOPIC

Low-grade intraductal carcinoma (LG-IC) is a rare salivary gland malignant tumor.[1] To the best of our knowledge, only 56 cases have been reported in the archives to date. The United States accounts for half of all reported cases, followed by Japan (26%), Canada, China, Korea, Taiwan, Czech Republic, and Italy.[1-5] So far, only one case has been documented in India as an incidental diagnosis on an excision biopsy of a 56-year-old female, who was clinically and radiologically diagnosed with a pleomorphic adenoma of the palate.[6]

In 1996, Delgado et al.,[7] mentioned LG-ICs for the first time in a case series, where it was described as a “mass forming lesion with morphological features analogous to mammary ductal hyperplasia and intact myoepithelial cell layer.” Since then, the nomenclature of LG-IC has been reviewed at times. It was renamed from “Low-grade cribriform cystadenocarcinoma” or “Low-grade salivary duct carcinoma” to “Salivary duct carcinoma in situin 2005.[8,9] In 2017, the World Health Organization Head and Neck tumors reclassified the entities LGCCC and salivary duct carcinoma in situ collectively as “Intraductal carcinoma,” with low-grade and high-grade features, respectively.[10-12]

In the majority of cases, LG-ICs are seen in a wider age range of 27–93 years, with a slight female (M: F = 1:1.3) predilection.[2-5] The most common site is the parotid gland (>80%), with rare occurrences in minor salivary glands. The clinical presentation of LG-IC is mostly a circumscribed cystic mass with indolent behavior. Exceptionally, LGIC can be intranodal, having arisen from salivary gland inclusions in the lymph node. Macroscopically, LG-ICs are well-circumscribed, non-encapsulated, multicystic lesions; a size varying from 0.7 to 5.3 cm, and contain serous to hemorrhagic fluid.[1] Histologically, following variants have been described:

Intercalated duct-like (most common)[13]

Apocrine[11]

Mixed/hybrid

Possibly oncocytic, but may be a variant of intercalated duct type.[14]

Complete surgical excision with preservation of the facial nerve is the most commonly practiced management at present. LG-IC has an excellent prognosis after complete excision, with no metastasis or mortality at a follow-up of 2–12 years, regardless of nuclear grade. However, recurrence can occur as a result of incomplete resection, positive surgical margins, or metastasis. The previous systemic reviews illustrate that adjuvant radiotherapy is not justified for tumor resections with negative margins, even in the presence of a close margin. However, it may be advised in cases of positive margins or invasive tumors.[1]

A systematic review performed by Giovacchini et al.[1] revealed the heterogeneous morphology of tumor cells lacking cellular or nuclear pleomorphism, prominent nucleoli, significant mitotic activity, and necrosis. Furthermore, these tumors had no local or perineural invasion.[1,2,7,15]

Nakazawa et al.,[4] Jeong et al.,[16] and Kokabu et al.,[17] showed FNA reports suggestive of a cystic lesion with a mildly irregularly shaped nucleus with inconspicuous nucleoli and little atypia; some had single, large, and clear cytoplasmatic vacuoles. Background showed scattered lymphocytes, neutrophils, macrophages, and eosinophils. The FNA morphology is closely similar to our case findings with specific backgrounds.

Kuo et al.[18] identified the following LG-IC multicystic architecture differential diagnoses: cystadenocarcinoma, cystadenoma, salivary duct carcinoma in situ/high-grade intraductal carcinoma, conventional salivary duct carcinoma, acinic cell carcinoma (Papillary Cystic Pattern), mammary analog secretory carcinoma, sclerosing polycystic adenosis, ductal adenoma with striated duct differentiation, and intercalated duct lesion.

According to the literature, FNAC has lower sensitivity to cliché the diagnosis of LG-IC as malignant, with only four FNAs out of all cases reported to date showing malignant neoplasms,[11,17,19] 23% of cases reported as benign,[12,20,21] and 46% of cases reported as SUMP.[3-5,17,22]

Immunohistochemically, Giovacchini et al.[1] reviewed that total 55 cases revealed that 92% cases showed tumor cells positive for S-100, 18% for cytokeratinAE-1/AE-3, 14.5% for mammaglobin, 67% for GCDFP-15, 94% for calponin, 86% for DOG1, 91% for CK14, 95% for SMA, 70% for CEA, and 11% for CK7. Instead, 95% of tumor cells were negative for Her-2/neu, 62% for androgen receptor, 90% for ER, 90% for PR, and 75% for p53. In LG-IC, GATA-3 was generally negative; it may be useful in distinguishing LG-IC from mammary secretory carcinoma, metastatic squamous cell carcinoma, mucoepidermoid carcinoma, salivary duct carcinoma, and Warthin tumor, which are usually positive.[23] The role of IHC is found to be supportive, however not specific, as well as controversial in the diagnosis for LG-IC.

SUMMARY

LG-ICs are unique in their cytohistomorphological complexity. On histomorphology, they have classical non-invasive pseudocribriform and Roman bridges-like architectural patterns, which are similar to the pattern seen in low-grade intraductal breast carcinoma. This specific tumor is considered the counterpart of low-grade intraductal breast carcinoma, which is highly debatable and still not confirmed. On FNAC, the overlapping cytomorphological features make its identification as a malignant tumor more difficult. FNAC plays a pivotal role in detecting both benign and malignant etiologies, the latter in those not suitable for attempted curative surgery or with recurrent disease before palliative treatment, and can also reduce the rate of salivary gland surgery by one-half to one-third. The crux of the case reported is the importance of FNA with good skills to identify neoplastic etiology, especially indolent malignant tumors like LG-IC when cytomorphological features are benign-looking. However, histopathological examination is considered the “gold standard” to arrest the diagnosis of LG-ICs, and after complete excision, it carries an excellent prognosis.


References

Giovacchini F Bensi C Belli S Laurenti ME Mandarano M Paradiso D et al Low-grade intraductal carcinoma of salivary glands: A systematic review of this rare entity J Oral Biol Craniofac Res 2019996110 10.1016/j.jobcr.2018.11.003 30555776
Kusafuka K Itoh H Sugiyama C Nakajima T Low-grade salivary duct carcinoma of the parotid gland: Report of a case with immunohistochemical analysis Med Mol Morphol 20104317884 10.1007/s00795-009-0479-2 20857268
Kimura M Mii S Sugimoto S Saida K Morinaga S Umemura M Low-grade cribriform cystadenocarcinoma arising from a minor salivary gland: A case report J Oral Sci 2016581459 10.2334/josnusd.58.145 27021553
Nakazawa T Kondo T Yuminomochi T Nakazawa K Ishii Y Mochizuki K et al Fine-needle aspiration biopsy of low-grade cribriform cystadenocarcinoma of the salivary gland Diagn Cytopathol 20113921822 10.1002/dc.21405 21319326
Ohta Y Hirota Y Kohno Y Kishimoto K Norose T Ohike N et al Cytology of low-grade cribriform cystadenocarcinoma in salivary glands: Cytological and immunohistochemical distinctions from other salivary gland neoplasms Diagn Cytopathol 2016442415 10.1002/dc.23411 26875597
Devi A Narwal A Kamboj M Gupta M Pandiar D Low grade intraductal carcinoma of palate: An extremely rare entity with review of literature August 2020 Indian J Otolaryngol Head Neck Surg 20227422227 10.1007/s12070-020-02092-5 36452681
Delgado R Klimstra D Albores-Saavedra J Low grade salivary duct carcinoma. A distinctive variant with a low grade histology and a predominant intraductal growth pattern Cancer 19967895867 10.1002/(SICI)1097-0142(19960901)78:5<958::AID-CNCR4>3.0.CO;2-8
Seethala RR Stenman G Update from the 4th edition of the world health organization classification of Head and neck tumours: Tumors of the salivary gland Head Neck Pathol 2017115567 10.1007/s12105-017-0795-0 28247227
Barnes L Eveson JW Reichart P Sidransky D Pathology and Genetics Head and Neck Tumors Geneva World Health Organization 2005
Weinreb I Intraductal carcinoma of salivary gland (So-Called low-grade cribriform cystadenocarcinoma) arising in an intraparotid lymph node Head Neck Pathol 201153215 10.1007/s12105-011-0256-0 21442195
Weinreb I Tabanda-Lichauco R Van Der Kwast T Perez-Ordoñez B Low-grade intraductal carcinoma of salivary gland: Report of 3 cases with marked apocrine differentiation Am J Surg Pathol 200630101421 10.1097/00000478-200608000-00013 16861974
Nishijima T Yamamoto H Nakano T Hatanaka Y Taguchi KI Masuda M et al Low-grade intraductal carcinoma (low grade cribriform cystadenocarcinoma) with tumor-associated lymphoid proliferation of parotid gland Pathol Res Pract 20172137069 10.1016/j.prp.2017.02.019 28551385
Weinreb I Bishop JA Chiosea SI Seethala RR Perez-Ordonez B Zhang L et al Recurrent RET gene rearrangements in intraductal carcinomas of salivary gland Am J Surg Pathol 20184244252 10.1097/PAS.0000000000000952 29443014
Bishop JA Nakaguro M Whaley RD Ogura K Imai H Laklouk I et al Oncocytic intraductal carcinoma of salivary glands: A distinct variant with TRIM33-RET fusions and BRAF V600E mutations Histopathology 20217933846 10.1111/his.14296 33135196
Brandwein-Gensler M Hille J Wang BY Urken M Gordon R Wang LJ et al Low-grade salivary duct carcinoma: Description of 16 cases Am J Surg Pathol 20042810404 10.1097/01.pas.0000128662.66321.be 15252310
Jeong JY Ahn D Park JY Fine-needle aspiration cytology of low-grade cribriform cystadenocarcinoma with many psammoma bodies of the salivary gland Korean J Pathol 2013474815 10.4132/KoreanJPathol.2013.47.5.481 24255638
Kokabu S Nojima J Kayano H Yoda T Low-grade cribriform cystadenocarcinoma of the palatal gland: A case report Oncol Lett 20151024537 10.3892/ol.2015.3528 26622870
Kuo YJ Weinreb I Perez-Ordonez B Low-grade salivary duct carcinoma or low-grade intraductal carcinoma? Review of the literature Head Neck Pathol 201375967 10.1007/s12105-013-0460-1 23821212
Obokata A Sakurai S Hirato J Sakamoto K Takekoshi T Aoki J Cytologic features of low-grade cribriform cystadenocarcinoma of the submandibular gland: A case report Acta Cytol 20135720712 10.1159/000345910 23406647
Arai A Taki M Mimaki S Ueda M Hori S Low-grade cribriform cystadenocarcinoma of the parotid gland: A case report Auris Nasus Larynx 2009367258 10.1016/j.anl.2009.04.005 19477092
Wakabayashi N Umezawa H Matsumoto NM Endo Y Naito Z Ogawa R Low-grade cribriform cystadenocarcinoma: A review of the literature and case report Plast Reconstr Surg Glob Open 20175e1306 10.1097/GOX.0000000000001306 28507867
Ko YS Koo JS Cytomorphological findings and histological correlation of low-grade cribriform cystadenocarcinoma of salivary gland in fine-needle aspiration: A case study Korean J Pathol 2013475925 10.4132/KoreanJPathol.2013.47.6.592 24421856
Schwartz LE Begum S Westra WH Bishop JA GATA3 immunohistochemical expression in salivary gland neoplasms Head Neck Pathol 201373115 10.1007/s12105-013-0442-3 23604756


An extremely rare case of axillary accessory breast swelling with uncommon association of methicillin-resistant

A 34-year-old female presented to the surgery department with persistent nodular left axillary swelling and pain for 15 days. A fine needle aspiration biopsy was performed from the left axilla and image of the smear is depicted below.

QUESTION

Q1. What is the interpretation/diagnosis?

Breast carcinoma

Granulomatous mastitis (GM) in accessory axillary breast

Fibrocystic change

Phyllodes tumor.

ANSWER

b- GM in accessory axillary breast.

EXPLANATION

Cytological findings showed cellular aspirate with sheets of benign ductal epithelial cells, few non-necrotizing epithelioid cell granulomas, numerous foamy macrophages, neutrophils, and multinucleated giant cells showing emperipolesis [Figures 1a-c]. Ziehl-Neelsen stain and TrueNat for acid-fast bacilli were also performed which was negative, excluding Tuberculosis. Based on cytological findings, the diagnosis of GM in the accessory axillary breast was considered. Methicillin-resistant Staphylococcus aureus was later isolated in the bacterial culture performed on aspirate sample. Following the administration of antibiotics, the lesion significantly improved.

(a-c) Fine needle aspirate biopsy smear from left axillary swelling stained with papanicolaou and may-grunwald-giemsa stain.

ADDITIONAL QUIZ QUESTIONS

Q2. Which of the following is true about GM?

Usually occurs in women of reproductive age, and most cases occur around 2 years after breastfeeding

Malignant disease of the breast

It is an untreatable condition

None.

Q3. Which of the following disease is reported in the accessory breast?

Phyllodes

Mastitis

Fibroadenoma

All of the above.

Q4. What are the other granulomatous diseases of the breast?

Tuberculous mastitis

Sarcoidosis

Both

Galactocele.

ANSWERS TO ADDITIONAL QUIZ QUESTIONS

Answers: Q2-a, Q3-d, Q4-c.

BRIEF REVIEW OF THE TOPIC

GM is an unusual chronic inflammatory condition of the breast that is characterized by breast masses, erythema, abscesses, indurations, and tenderness. It usually occurs in pregnant women within 5 years of giving birth.[1] GM was described as a distinct entity in 1972 by Kessler and Wolloch.[2] In general, accessory breast tissue extends from the axilla to the pubic area along the embryonic mammary ridge.[3] The disease processes that affect accessory breast tissue are similar to those that affect normal breast tissue. The most frequent diseases reported in the accessory breasts are cancers followed by mastitis, fibroadenoma, phyllodes tumors, and fibrocystic change.[4] The majority of cases of GM occur within the first 2 years after breastfeeding, while GM during pregnancy is rare.[5] Idiopathic GM manifests histologically as non-caseating granulomas and is chronic, rare, and inflammatory. Typically, it presents as an inflamed, tender mass on the breast.[6] In our case, the patient had given birth to her first child 3 years ago, and there was no history of breast trauma/use of oral contraceptives/any family history of breast cancer. Clinical examination revealed a tender swelling of approximately 1.5 cm in size with redness in the left axilla. Ultrasonography findings were suggestive of left axillary lymphadenitis. The patient was exposed to a number of risk factors, including delivery and breastfeeding, which both contribute to GM and accessory breast tissue formation.[6,7] The diagnosis of GM remains challenging for clinicians. Cytological examination of fine-needle aspiration biopsy can ensure the diagnosis of this disease, despite the fact that it often mimics breast cancer.[8] Patients with GM are often subject to prolonged disease courses with substantial negative impacts on quality of life during diagnosis and treatment. Therefore, diagnosing and treating them remain a challenge for clinicians as well as patients. To the best of our knowledge, till date, only two other cases of GM in accessory breast tissue have been reported in the literature. At present, there is no consensus regarding the etiology and management of GM.[9] In our case, the patient was treated with oral antibiotics and the patient’s condition improved.

SUMMARY

The diagnosis of GM on the accessory axillary breast should be considered in women with pain and swelling along the milk line and having recent history of delivery and breastfeeding. As far as GM is concerned, there is no standard management approach. Therefore, treatment strategies should be tailored to the needs of each patient.


References

Brown KL Tang PH Postlactational tumoral granulomatous mastitis: A localized immune phenomenon Am J Surg 19791383269 10.1016/0002-9610(79)90397-0 464240
Kessler E Wolloch Y Granulomatous mastitis: A lesion clinically simulating carcinoma Am J Clin Pathol 1972586426 10.1093/ajcp/58.6.642 4674439
Bartsich SA Ofodile FA Accessory breast tissue in the axilla: Classification and treatment Plast Reconstr Surg 201112835e-6 10.1097/PRS.0b013e3182173f95 21701310
Pfeifer JD Barr RJ Wick MR Ectopic breast tissue and breast-like sweat gland metaplasias: An overlapping spectrum of lesions J Cutan Pathol 1999261906 10.1111/j.1600-0560.1999.tb01827.x 10335896
Wolfrum A Kümmel S Theuerkauf I Pelz E Reinisch M Granulomatous mastitis: A therapeutic and diagnostic challenge Breast Care (Basel) 2018134138 10.1159/000495146 30800035
Lai EC Chan WC Ma TK Tang AP Poon CS Leong HT The role of conservative treatment in idiopathic granulomatous mastitis Breast J 200564546 10.1111/j.1075-122X.2005.00127.x 16297091
Khanna S Mishra SP Kumar S Khanna AK Carcinoma in accessory axillary breast BMJ Case Rep 20152015bcr2015210944 10.1136/bcr-2015-210944 26260957
Macansh S Greenberg M Barraclough B Pacey F Fine needle aspiration cytology of granulomatous mastitis. Report of a case and review of the literature Acta Cytol 1990343842
Freeman M Lewis CD Lower E Khan S Shaughnessy E Refractory granulomas of breast: Benign or malignant disease J Clin Oncol 201432 Suppl 2621 10.1200/jco.2014.32.26_suppl.21


Rapid on-site evaluation of a solitary lung nodule in a patient with remote history of hysterectomy: Cytologic findings and diagnostic challenges

A 75-year-old female patient was referred to our institution with chronic upper gastrointestinal symptoms. A computed tomography (CT) scan of the abdomen and chest revealed a well-demarcated, ovoid nodule in the right, lower lobe of the lung measuring 17 mm. She had a remote history of hysterectomy for endometrial sarcoma 26 years ago. A transbronchial fine needle aspiration biopsy and rapid on-site assessment of smears were performed. The smears were moderately cellular comprising very uniform population of small, round to oval cells with very scant cytoplasm disposed as single cells and occasional clusters associated with occasional metachromatic matrix [Figure 1a and b]. The Papanicolaou stain showed cells with moderately hyperchromatic nuclei with evenly distributed chromatin and inconspicuous nucleoli. The cell block showed groups of small cells arranged around thin-walled vessels and eosinophilic and hyaline matrix. There were no mitoses, necrosis, or hemorrhage [Figure 1c and d].

(a) Cytologic smear of lung nodule on rapid on-site assessment (Diff Quik, ×200). (b) Discrete and clusters of cells with pale matrix (Diff Quik, ×400). (c) Papanicolaou stain showing monotonous small round to oval cells ×400. (d) Cell block showing sheets of small cells with hyperchromatic nuclei (H and E ×200).

Q1. What is the most likely diagnosis?

Lung hamartoma

Non-Hodgkin lymphoma

Small cell carcinoma

Carcinoid tumor

Metastatic low-grade endometrial stromal sarcoma (ESS)

Answer to Q1: Option e

Explanation: The cytologic features of lung hamartoma are characterized by paucicellular smears, prominent chondroid matrix, bronchial epithelial cells, sheets of small round cells, and occasionally fat cells.[1] In our case, there were no bronchial cells, cartilage, or adipose tissue. The small round cells with hyperchromatic nuclei mimicked a non-Hodgkin lymphoma. However, the presence of cohesive cell groups and absence of lymphoglandular bodies make this diagnosis unlikely. Small cell carcinoma was ruled out by the absence of nuclear molding, chromatin smearing, necrosis, and mitoses.

Metastatic ESS lacks any characteristic features on cytology and is frequently mistaken for a benign lesion or carcinoid tumor. A history of hysterectomy 26 years ago in our patient is an important clue to the diagnosis. The cytology smears were characterized by small, round to oval cells with no visible cytoplasm and no definite pattern of the arrangement of the tumor cells. Spindle cells were not evident and necrosis, mitoses, and hemorrhage were absent.

ESS is a very rare tumor of the endometrium comprising 1.0% of all uterine malignancies.[2] The current World Health Organization classifies these groups of neoplasms into four distinct categories: Endometrial stromal nodule, low-grade ESS, high-grade ESS, and undifferentiated uterine sarcoma. Low-grade ESS accounts for 86% of all ESS[2] and usually affects perimenopausal women. The tumor is characterized by tongue like invasion into the myometrium. Microscopically, the tumor resembles proliferative phase endometrium with small to oval cells surrounding blood vessels and low mitotic index. Late recurrence and metastasis can occur even in early-stage disease requiring long-term follow-up.[2] Distant metastasis to the lung is typically in the form of multiple nodules.

Q2. Which immunohistochemical panel would be best to confirm the diagnosis of low-grade ESS?

Chromogranin, synaptophysin, CK5/6, INSM1, and ki67

CK7, TTF1, Napsin A, p63, and p40

CD10, ER, PR, WT1, AR, and interferon-induced transmembrane protein 1 (IFITM1)

P16, desmin, SMA, and beta-catenin

Answer: c

Explanation: There is no single marker that is specific for low-grade-ESS; the use of a panel of immunohistochemical markers can help in making the diagnosis besides detailed clinical history. The tumor is typically positive for CD10, ER, PR, WT1, AR, and IFITM1. The neuroendocrine and smooth muscle markers were negative in our case [Figure 2].

(a) Immunohistochemical stain for CD10 ×200. (b) Immunohistochemical stain for WT1 ×200.

Q3. What is the most common cytogenetic abnormality associated with low-grade ESS?

t(7;17) (p16;q21)

t(6;7) (p21;p15)

t(10;17) (q22;p13)

t(x;17)(p21-p11;q23)

Answer: a

Explanation: Different types of chromosomal abnormalities have been described in low-grade ESS and cytogenetic analyses have been used to confirm the diagnosis. Different types of translocation have been reported with t(7;17) translocation being the most distinctive cytogenetic hallmark of low-grade ESS with fusion of two genes JAZF1 and SUZ12 (2, 3).

Q4 What is the most common site of metastasis of lowgrade ESS besides lung?

Bone

Kidney

Brain

Abdomen

Answer: d

Explanation: The lung and abdomen are the most frequent sites of metastasis and recurrence.

Our case was discussed at the multidisciplinary conference for further management. Due to her age and oligo metastatic disease, the patient opted for close follow-up, and anti-hormonal therapy was recommended.

BRIEF REVIEW OF THE TOPIC

Endometrial stromal tumors are rare tumors that arise from the endometrial stroma. Low-grade ESS typically occurs in perimenopausal women and has a very indolent course with excellent prognosis.[3] Distant metastasis can develop many decades after the initial diagnosis.[3] Pulmonary metastasis from low-grade ESS usually manifests as multiple nodules.[4-6] Very rarely, however, it can present as a solitary lesion, which can pose significant diagnostic dilemma. Due to its rarity and bland appearance, low-grade ESS has been mistaken for benign lesions or inflammatory processes.[4,6-9] The diagnostic dilemma is even more challenging in cytology specimen and small biopsies.[5]

The cytologic features of metastatic low-grade ESS have rarely been described in the literature since it is rarely subjected to fine-needle aspiration.[4,8-10] In the lung, it has been mistaken for carcinoid tumor, hamartoma, and endometriosis.[4,5,9] A predominance of bland spindle cells can also be mistaken for leiomyoma and other benign spindle cell neoplasms.[5,7,8]

The diagnostic challenges of low-grade ESS in cytology have been emphasized by others, and in most case reports of metastatic low-grade ESS, a definite diagnosis was achieved only after performing molecular studies. Zaharopoulos et al.[10] first described a case of ESS that metastasized to the lung. The fine-needle aspiration biopsy of the lung lesion revealed small cells with scant cytoplasm and low mitosis. An ultrastructural study was performed which confirmed the diagnosis of metastatic ESS. Satoh et al.[9] emphasized the diagnostic dilemma of metastatic low-grade ESS in transbronchial fine-needle aspiration (FNA) cytology where the initial diagnosis was thought to be a non-neoplastic lesion. On surgical resection, a diagnosis of pulmonary endometriosis was considered due to the presence of bland and round to oval cells with positive expression for ER and PR. There was no previous history of gynecologic malignancy; however, a cytogenetic and FISH analysis demonstrated a t(7;17) translocation, which confirmed the diagnosis of ESS.

Ronen et al.[5] reported a case of metastatic ESS in a patient who presented with multiple lung nodules. The FNA smears showed bland, oval to spindle cells with moderate cytoplasm, and delicate vessels, but no matrix component. The differential diagnosis included carcinoid tumor and diffuse neuroendocrine hyperplasia; however, the IHC stains for neuroendocrine markers were negative, and CD10 was positive. A definitive diagnosis could not be rendered because of the limited nature of the cytology specimen. The nodule was subsequently resected, and histology revealed tumor cells with smooth muscle differentiation and positive staining for ER, PR, and desmin. On further clinical investigation, a history of recent hysterectomy for fibroids at another hospital was obtained. On re-review of slides from the hysterectomy specimen, a small focus of low-grade ESS with similar morphology to the lung metastasis was noted confirming the diagnosis.

Mindiola-Romero et al.[8] described a similar clinical scenario of low-grade ESS metastatic to the lung in a patient who presented with multiple lung nodules. A CT-guided core biopsy with rapid on-site assessment demonstrated cells with oval and spindled nuclei with mild-to-moderate atypia and scant matrix component. The core biopsy demonstrated bland spindle cells. Immunohistochemical workup for spindle cell neoplasms was performed using markers for desmin, S100, STAT6, CD34, and SOX-10 which were all negative. This prompted a molecular analysis that detected a fusion between exon3 of JAZF1 and exon 2 of SUZ12 supporting the diagnosis of low-grade ESS. Subsequently, immunohistochemical stains for ER, PR, and CD10 were positive corroborating the diagnosis. On further clinical investigation, the patient had a history of hysterectomy 25 years ago for ESS.

Metastatic low-grade ESS presenting as a solitary lung nodule as observed in our case is very rare. The predominance of small, round to oval cells with bland nuclei was thought to be consistent with a carcinoid tumor. Negative expression for neuroendocrine markers prompted review of the previous clinical history and additional IHC workup. Positive expression for ER, PR, CD10, and WT1 markers and a remote history of hysterectomy 26 years ago for endometrial sarcoma facilitated in making the right diagnosis. It must be emphasized that none of the above markers are specific for ESS; however, positive staining for WT1 has been shown to be a useful marker for differentiating extrauterine ESS from other potential mimics.[11] IFITM1 is a novel marker for endometrial stroma cells with a higher specificity than CD10.[12]

SUMMARY

This report highlights the diagnostic pitfall of metastatic LGESS in cytology specimens. The importance of clinical history and appropriate use of ancillary tests cannot be overemphasized. Awareness of the various cytomorphologic features and potential mimics is important.


References

Hughes JH Young NA Wilbur DC Renshaw AA Mody DR Cytopathology Resource Committee CoAP Fine-needle aspiration of pulmonary hamartoma: A common source of false-positive diagnoses in the college of American pathologists interlaboratory comparison program in nongynecologic cytology Arch Pathol Lab Med 20051291922 10.5858/2005-129-26-PISGFC
Capozzi VA Monfardini L Ceni V Cianciolo A Butera D Gaiano M et al Endometrial stromal sarcoma: A review of rare mesenchymal uterine neoplasm J Obstet Gynaecol Res 202046222136 10.1111/jog.14436 32830415
Chiang S Oliva E Recent developments in uterine mesenchymal neoplasms Histopathology 20136212437 10.1111/his.12048 23240674
Aubry MC Myers JL Colby TV Leslie KO Tazelaar HD Endometrial stromal sarcoma metastatic to the lung: A detailed analysis of 16 patients Am J Surg Pathol 2002264409 10.1097/00000478-200204000-00005 11914621
Ronen S Narula N Koizumi JH Hunt B Giorgadze T Low-grade endometrial stromal sarcoma presenting as multiple pulmonary nodules: A potential pitfall in fine needle aspiration and core biopsy specimens-a cytological-pathological correlation Ann Diagn Pathol 2018363843 10.1016/j.anndiagpath.2018.06.004 30055523
Satoh Y Ishikawa Y Miyoshi T Mukai H Okumura S Nakagawa K Pulmonary metastases from a low-grade endometrial stromal sarcoma confirmed by chromosome aberration and fluorescence in-situ hybridization approaches: A case of recurrence 13 years after hysterectomy Virchows Arch 20034421738 10.1007/s00428-002-0731-9 12596070
Mahadeva R Stewart S Wallwork J Metastatic endometrial stromal sarcoma masquerading as pulmonary lymphangioleiomyomatosis J Clin Pathol 1999521478 10.1136/jcp.52.2.147 10396245
Mindiola-Romero AE Liu X Dillon JL Talarico M Smith G Zhang L et al Metastatic low-grade endometrial stromal sarcoma after 24 years: A case report and review of recent molecular genetics Diagn Cytopathol 202149E99105 10.1002/dc.24601 32910526
Satoh Y Ishikawa Y Furuta N Tsuzuku M Difficulty of cytodiagnostic approaches to pulmonary metastases from a case of low grade endometrial stromal sarcoma Acta Cytol 20034711401
Zaharopoulos P Wong JY Lamke CR Endometrial stromal sarcoma: Cytology of pulmonary metastasis including ultrastructural study Acta Cytol 1982264954
Sumathi VP Al-Hussaini M Connolly LE Fullerton L McCluggage WG Endometrial stromal neoplasms are immunoreactive with WT-1 antibody Int J Gynecol Pathol 2004232417 10.1097/01.pgp.0000130051.04396.13 15213600
Zhao W Cui M Zhang R Shen X Xiong X Ji X et al IFITM1, CD10, SMA, and h-caldesmon as a helpful combination in differential diagnosis between endometrial stromal tumor and cellular leiomyoma BMC Cancer 2021211047 10.1186/s12885-021-08781-w 34556086


Breast lump: “Keep me in your differentials”

A 31-year-old female with tender lump in left breast lump 4 months with pain and intermittent yellow colored nipple discharge (2 months) [Figure 1a]. Fine-needle aspiration yielded 10 ml of yellowish pus like material with cytological findings shown in [Figure 1b , c, and c (inset)].

(a) Left breast with erythema. (b,c, and c (inset). FNA smear showing inflammatory cells comprising of neutrophils, foamy macrophages, and degenerated cells along with cotton ball like colony in the center with radiating filaments [b, c, and c(inset): MGG; b, X10; c, X40; c(inset) Zoomed].

What is the diagnosis based on cytomorphology?

Actinomyces

Tuberculosis

Non-Hodgkin’s lymphoma

Ductal carcinoma.

Answer to the first question:

1. a. Actinomyces

What is the special stain used in such cases?

PAS

ZN

Gram

All of the above

None of the above.

What is the most common causative agent of breast abscess?

Staphylococcus

Streptococcus

Enterococcus

Pseudomonas.

REVIEW OF THE TOPIC

Actinomycosis is a chronic infection caused by Actinomyces species characterized by abscess formation, tissue fibrosis, and draining sinuses. The spectrum of infections caused by Actinomyces species ranges from classical invasive actinomycosis to a less invasive form of superficial skin and soft-tissue infection.[1] It is an often relapsing granulomatous infection that characteristically crosses tissue planes and forms abscesses and sinus tracts.[2] First described by Ammentrop in 1893, primary actinomycosis of the breast is very rare and most commonly caused by Actinomyces israelii which normally inhabits the mouth, colon, and vagina.[3-5]

Breast actinomycosis is primary when inoculation occurs through the nipple. Secondary actinomycosis of the breast refers to the extension of a pulmonary infection through the thoracic cage in a process that can affect the ribs, muscles, and finally the breast.[6] Primary actinomycosis infections are commonly caused by A. israelii. Actinomyces neuii is a less common cause of classical actinomycosis.[7] Actinomyces of breast is an uncommon occurrence with very limited case reports in the literature.[8-10] Most often, the diagnosis of Actinomyces breast is made based on cytological findings. Differentiating causative agents of breast abscess is challenging. Knowledge of this organism at this rare site is essential so that it could be considered in the differentials. Distinguishing breast actinomycosis from tuberculosis and other potential infectious conditions is possible through pathological examination.[11] Cytomorphology helps in making the diagnosis. Microbiological examination aids in the confirmation of the same. Although breast is a rare site for Actinomyces infection, cytopathologists must be aware of its occurrence and consider this in their differentials while diagnosing breast lumps.

Answers to the additional questions:

2. c. Gram

3. a. Staphylococcus.

COMPETING INTEREST STATEMENT BY ALL AUTHORS

There is no conflict of interest in this paper.

AUTHORSHIP STATEMENT BY ALL AUTHORS

Each author has participated sufficiently in the work and takes public responsibility for appropriate portions of the content of this article. All authors read and approved the final manuscript. Each author acknowledges that this final version was read and approved.

ETHICS STATEMENT BY ALL AUTHORS

FNAC was done after proper consent as routine diagnostic test.

LIST OF ABBREVIATIONS (In alphabetic order)

PAS- Periodic Acid Schiff

ZN- Ziehl Nelson

EDITORIAL/PEERREVIEW STATEMENT

To ensure the integrity and highest quality of CytoJournal publications, the review process of this manuscript was conducted under a double-blind model (the authors are blinded for reviewers and vice versa) through automatic online system.


References

Bing AU Loh SF Morris T Hughes H Dixon JM Helgason KO Actinomyces species isolated from breast infections J Clin Microbiol 201553324755 10.1128/JCM.01030-15 26224846
Fred HL Aisenberg GM van Dijk HA Actinomycosis of the breast and lungs Proc (Bayl Univ Med Cent) 201932656 10.1080/08998280.2018.1540251 30956585
Salmasi A Asgari M Khodadadi N Rezaee A Primary actinomycosis of the breast presenting as a breast mass Breast Care (Basel) 201051057 10.1159/000301599 20847823
Attar KH Waghorn D Lyons M Cunnick G Rare species of Actinomyces as causative pathogens in breast abscess Breast J 2007135015 10.1111/j.1524-4741.2007.00472.x 17760673
Kamal MM The Pap smear in inflammation and repair Cytojournal 20221929 10.25259/CMAS_03_08_2021 35673696
De Barros N Issa FK Barros AC D'Avila MS Nisida AC Chammas MC et al Imaging of primary actinomycosis of the breast AJR Am J Roentgenol 200017417846 10.2214/ajr.174.6.1741784 10845523
Leenstra BS Schaap CC Bessems M Renders NH Bosscha K Primary actinomycosis in the breast caused by Actinomyces neuii. A report of 2 cases IDCases 20178702 10.1016/j.idcr.2017.03.014 28462153
Yalav O Topal U Ünal AG Dalci K Primary actinomycosis of the breast in postmenopausal women Ann Ital Chir 20198S2239253X19031232 31869310
Gosavi AV Anvikar AR Sulhyan KR Manek DD Primary actinomycosis of breast-A diagnosis on cytology Diagn Cytopathol 2016446935 10.1002/dc.23499 27238823
Lacoste C Escande MC Jammet P Nos C Breast Actinomyces neuii abscess simulating primary malignancy: A case diagnosed by fine-needle aspiration Diagn Cytopathol 2009373112 10.1002/dc.21044 19217059
Thambi R Devi L Sheeja S Poothiode U Primary breast Actinomyces simulating malignancy: A case diagnosed by fine-needle aspiration cytology J Cytol 2012291979 10.4103/0970-9371.101173 23112463


Diagnostic difficulties in evaluation of primary malignant lesions of thyroid: A study of cytomorphology, histopathology, and immunohistochemistry

18-year-old male presented with complaints of midline neck swellings since 1½ years associated with history of loss of weight. On examination, it was soft to firm in consistency. Contrast-enhanced computed tomography (CECT) neck was suggestive of neoplastic etiology. Cytopathological findings following ultrasound (USG)-guided fine-needle aspiration and cytology (FNAC) are shown in [Figure 1].

(a-c) Smear showing atypical spindle cell arranged in dyscohesive clusters and swirls. The cells reveal hyperchromatic nuclei with moderate amount of cytoplasm (MGG, ×400); (d) Smear showing a biphasic lesion comprising of both epithelial component (arrow) forming acini and spindle cells (MGG, ×100).

QUESTION 1

Which of the following is the LEAST LIKELY diagnosis?

Medullary Carcinoma Thyroid

Primary Synovial Sarcoma (SS)

Spindle Epithelial Tumor with Thymus-Like Differentiation (SETTLE)

Papillary Carcinoma

Undifferentiated (Anaplastic) Thyroid Carcinoma

Immature Teratoma of thyroid

Answer: d. Papillary Carcinoma

EXPLANATION

Papillary carcinoma thyroid (PTC) cannot be the differential diagnosis in this case as cytologically PTC reveals syncytial cell aggregates and sheets with distinct anatomical borders. Papillary fragments with or without a fibrovascular core can also be seen. The cells typically show intranuclear inclusions and nuclear grooves.

However, medullary carcinoma, SETTLE, undifferentiated (anaplastic) carcinoma, immature teratoma of thyroid, and SS can all reveal spindle cell pattern. Although primary SS of thyroid is not a well-known entity and is very uncommon, the possibility cannot be ruled out.

The patient presented with a midline neck swelling [Figure 2] since a year and a half associated with loss of weight. His thyroid function tests were within normal limits. USG showed an oval hypoechoic lesion with central cystic area in the right lobe of thyroid [Figure 3]. This was followed by a radionuclide thyroid scan which showed a euthyroid gland with hypofunctioning nodule in the upper pole of the right lobe of thyroid gland [Figure 4]. CECT neck showed a well-defined heterogeneously enhancing solid cystic lesion in the right lobe with adjacent extensions and mass effect. Solid component showed significant post-contrast enhancement with mass attenuation suggestive of neoplastic etiology.

Patient presented with a soft to firm midline neck swelling.

Ultrasonography of thyroid showing an oval hypoechoic lesion with central cystic area in the right lobe of thyroid.

Radionuclide thyroid scan showing a euthyroid gland with hypofunctioning nodule in the upper pole of the right lobe of thyroid gland.

On cytology, the smears were cellular and revealed a biphasic lesion consisting of spindle cell and epithelial components. The atypical spindle cells were arranged in dyscohesive clusters and forming whorled pattern. Individual cells revealed hyperchromatic nuclei with scant tapering cytoplasm. Interspersed in between were seen epithelial cells arranged in acinar pattern having round to oval nuclei with some showing prominent nucleoli [Figure 1]. Focal areas suggestive of amyloid were identified. However, they stained negative for Congo red [Figure 5]. Diagnosis of undifferentiated carcinoma (the Bethesda Category VI) was considered with the following possibilities:

Primary SS – Biphasic

Medullary Carcinoma Thyroid

Smears showing negative congo red stain (Congo red, ×100).

The patient, then, underwent a total thyroidectomy and the specimen was sent for histopathological examination (HPE). On gross examination [Figure 6], we received a thyroidectomy specimen comprising two lobes measuring 3.5 × 2 × 0.5 cm and 3.2 × 1.8 × 1 cm, respectively, and isthmus measuring 1.5 × 1 × 0.4 cm. Attached skeletal muscle measured 4 × 3.5 × 1.5 cm. Cut section showed tumor mass in the right lobe measuring 2 × 1.5 cm with solid-cystic areas.

Total thyroidectomy specimen with tumour mass showing solid-cystic areas.

Microscopy [Figure 7] showed a cellular neoplasm composed predominantly of spindle-shaped cells arranged in fascicles. The cells revealed ovoid to spindle-shaped nuclei with fine granular chromatin, inconspicuous nucleoli, and 3–4 mitoses per high-power fields. Scattered small tubules and papillae [Figures 8 and 9] lined by cuboidal epithelium having uniform ovoid nuclei were present imperceptibly admixed with the spindle-shaped cells. No extrathyroidal extension and invasion into extrathyroidal skeletal muscle were noted. The above-mentioned features further strengthened the suspicion of a spindle cell sarcoma, suggestive of SS.

(a and c) Section showing spindle cells arranged in fascicles and whorls (H and E, ×100); (b and d) Higher magnification of the same (H and E, ×400); (e) Section showing epithelial cells arranged in papillae and tubules (H and E, ×100); (f) Higher magnification of the same (H and E, ×400).

Section showing epithelial cells arranged in papillae and tubules (H and E, ×100).

Higher magnification of the same (H and E, ×400).

IHC studies were done in a referral center. The tumor was immunoreactive for CK, CD99, and epithelial membrane antigen (EMA).

CK – Immunoreactive score 4+ in lesional cells

CD 99 – Immunoreactive score 3+ in lesional cells

EMA – Immunoreactive score 3+ in lesional cells.

Non-immunoreactive score 0 in lesional cells – CK7, Calretinin, Synaptophysin, TTF-1, and CK5/6.

ADDITIONAL QUESTIONS

Q2. Which of the following is the definitive diagnosis?

Medullary Carcinoma Thyroid

Primary SS

SETTLE

Papillary Carcinoma

Undifferentiated (Anaplastic) Thyroid Carcinoma

Immature Teratoma of thyroid

Q3. Which of the following IHC markers is not specific for this entity?

TLE1

h-caldesmon

EMA

Cytokeratin (CK)

Q4. Which of the following molecular pathologies is diagnostic of this entity?

Gain of function mutation in RET proto-oncogene

TP53 mutations

t(X;18)(p11.q11) translocation

Point mutations

Answers to additional questions:

Answer 2: b. Primary SS

Answer 3: a. TLE1

Answer 4: c. t(X;18)(p11.q11) translocation

EXPLANATION Answer 2 <italic>SETTLE versus Primary SS</italic>

SETTLE can be differentiated from SS on the basis of lower nuclear grade, glomeruloid glandular structures, stromal hyalinization, and diffuse expression of high molecular weight CK. The abovementioned features were absent in the present case.

<italic>Medullary carcinoma versus Primary SS</italic>

Cytological smears of medullary carcinomas are cellular and the cells may show variable patterns including plasmacytoid, small cells, or spindle cells. They may also variably exhibit amyloid or coarse red cytoplasmic granularity. In contrast to SS, these are positive for TTF-1 (weak to moderate), while the amyloid is positive for Congo red, both of which were negative in the present case.

<italic>Undifferentiated (anaplastic) carcinoma versus Primary SS</italic>

The patients with anaplastic carcinomas (AC) are usually women who present after 60 years of age. Cytological examination of AC shows the presence of necrotic background and highly pleomorphic malignant cells. The cells may range from spindle/squamoid cells to multinucleate and bizarre giant cells.

<italic>Immature teratoma of thyroid versus Primary SS</italic>

Although immature teratomas of thyroid can also be considered as a differential diagnosis, very few cases have been reported, with an average age of 43 years. Although in these cases also FNA reveals dominance of immature spindle cells since there was absence of immature neural tissue in the present case; thus, the possibility of being a malignant teratoma was excluded from the study. In addition to this, our patient was of 18 years of age and the FNA smears revealed a biphasic tumor comprising of both; spindle cell as well as epithelial cell component.

Answer 3

SSs show a moderate/strong nuclear expression for TLE1 which is a transcriptional corepressor. However, it is not specific for this entity alone. TLE 1 may also be seen in malignant nerve sheath tumor and solitary fibrous tumor. Other non-specific markers include bcl2 and CD99. Although a majority of SS show membranous positivity for these, they are not specific.

Answer 4

SSs are characterized by SYT-SSX1, SYT-SSX2, or SYT-SSX4 fusion gene. This is a result of t(X;18)(p11;q11) translocation which leads to the fusion of SS18 on chromosome 18 to one of the SSX genes- SSX1, SSX2, or SSX4.

Based on the above findings and discussion, a final diagnosis of the primary SS of thyroid was given.

BRIEF REVIEW OF TOPIC

SS is an exceedingly rare malignant mesenchymal neoplasm accounting for about 10% of soft-tissue sarcomas.[1] These are an extremely rare, malignant group of tumors characterized by chromosomal translocation t(X;18)(p11.2;q11.2) which results in the expression of SYT-SSX gene transcript.[2] SS is thought to arise from multipotent stem cells and not synovium.[3,4] Thus, they can occur at any site.[3,4] However, these usually arise in the deep soft tissue of the upper as well as lower extremities (70%), more often in the para-articular areas, followed by the trunk (15%) and the head and neck region (7%).[2] They are classified either as biphasic, composed of both spindle cell and epithelial cell component, or may be monophasic spindle cell type.[2,5]

The primary SS of thyroid is a rare, high grade, and aggressive tumor. To the best of our knowledge, only 13 cases have been reported in the literature so far.[1] It commonly presents in young adults and adolescents[1,6] and men are affected more than women with a 2:1 sex ratio.[1] The presenting symptoms usually include an asymptomatic rapidly growing neck mass.[1] The symptoms may also include hoarseness, dysphagia, dysphonia, and excessive salivation which may all be a result of compression effects by a rapidly growing tumor.[1] This unusual location makes the diagnosis of SS difficult.[6]

Radiological examination usually falls short in differentiating it from other thyroid malignancies. However, it can contribute in the assessment of location, size, vascularization, and local invasion.[1]

Pre-operative diagnosis based on FNAC is usually not supportive, although HPE may be contributory to some extent.[7] The conclusive diagnosis of this entity relies mainly on the diagnostic molecular studies such as fluorescence in situ hybridization.[5] However, immunohistochemistry may show positivity for EMA, CKs, and focal S100 expression (40% cases).[2] H-caldesmon is always negative.[2] Strong nuclear positivity is seen for TLE1 and membranous staining for BCl2 and CD99, but these markers are not specific.[2]

SUMMARY

The primary SS of thyroid is an extremely uncommon entity; however, the possibility cannot be entirely ruled out. Cytological findings, in our case, were strongly in favor of the primary SS. Although the definitive diagnosis was possible only on IHC studies, however molecular profiling is mandatory for further evaluation.


References

Seyed-Alagheband SA Sharifian M Adeli OA Sohooli M Shekouhi R Primary synovial sarcoma of thyroid gland: A case report and review of literature Int J Surg Case Rep 202185106245 10.1016/j.ijscr.2021.106245 34330070
Suurmeijer AJ Ladanyi M Nielsen TO Synovial sarcoma The WHO Classification of Tumours Editorial Board editors WHO Classification of Tumors: Soft Tissue and Bone Tumours 5th ed France International Agency for Research on Cancer 20192903
Park CS Kim Y Jeong EH Kim NI Choi YD Cytologic features of primary monophasic synovial sarcoma of the thyroid gland Cytojournal 20171424 10.4103/cytojournal.cytojournal_14_17 29259650
Akerman M Ryd W Skytting B Scandinavian Sarcoma Group Fine-needle aspiration of synovial sarcoma: Criteria for diagnosis: Retrospective reexamination of 37 cases, including ancillary diagnostics. A Scandinavian Sarcoma Group study Diagn Cytopathol 2003282328 10.1002/dc.10265 12722117
Ghafouri A Anbara T Mir A Lashkari M Nazari M Thyroid synovial sarcoma: A case report Acta Med Iran 2013516972
Srivastava P Anand N Husain N Primary synovial sarcoma thyroid: An unusual presentation J Pathol Nepal 20201017768 10.3126/jpn.v10i2.28358
Lal RK Hazarika B Julaha M Primary synovial sarcoma of thyroid gland: A case report Int J Otorhinolaryngol Clin 2020122730 10.5005/jp-journals-10003-1350


Touch preparation from a pancreatic core biopsy

A 50-year-old male presented with abdominal pain and weight loss. Abdominal CT showed a 5 cm round mass in the head of the pancreas. An ultrasound-guided core needle biopsy was performed. Touch prep of the needle cores performed for rapid onsite evaluation [Figure 1].

The touch prep showed loose clusters of cells with occasional vague acinar formations (a and b), abundant cytoplasm, occasional nucleoli, and mild-to-moderate anisonucleosis (c).

Q1. What is your interpretation of the touch preparation?

Atypical epithelial cells, suspicious for carcinoma

Well-differentiated neuroendocrine tumor

Negative for malignancy, favor chronic pancreatitis

Gastrointestinal contaminant (duodenal epithelium).

Answer

The correct cytological interpretation is

a. Atypical epithelial cells, suspicious for carcinoma.

EXPLANATION

A diagnosis of “suspicious for carcinoma,” A is appropriate when some or most features of carcinoma are present but fall qualitatively or quantitatively short of a definitive diagnosis for carcinoma. In this case, the touch prep shows variably sized loose clusters and single cells with acinar formation. The cells show abundant granular cytoplasm. The nuclei show mild-to-moderate anisonucleosis with occasional prominent nucleoli, and salt-and-pepper chromatin. At the time of rapid onsite evaluation, “suspicious for carcinoma” is the most appropriate diagnosis. The differential diagnosis would include pancreatic acinar cell carcinoma (ACC) and pancreatic neuroendocrine neoplasm.

Pancreatic well-differentiated neuroendocrine tumors show loosely cohesive cell clusters and single cells. Cytological preparations from these tumors may also demonstrate pseudo rosette formation that is impossible to distinguish from the acinar formation noted in ACC. Anisonucleosis is minimal, and the nuclear chromatin may show a classic salt-and-pepper appearance, clumping, or even prominent nucleoli.

A diagnosis of negative for malignancy, favor chronic pancreatitis, would be appropriate in the absence of neoplastic cells and when features of chronic pancreatitis are present. Cytological preparations may show inflammatory cells (predominantly macrophages, but also lymphocytes and occasional neutrophils). There may be rare ductal cells with mild-to-moderate atypia and fibrotic acinar tissue.

Duodenal contamination would consist of sheets of uniform epithelial cells with occasional goblet cells.

ADDITIONAL QUIZ QUESTIONS

Q2. H&E stained sections are prepared from the tissue cores [Figure 2] and show a solid, cellular neoplasm. Which of the following is the least likely diagnosis?

Neuroendocrine neoplasm

Solid pseudopapillary neoplasm

Pancreatic ACC

Islet cell pseudohypertrophy in chronic pancreatitis.

The core biopsy showed nests of cells with only mild nuclear anisocytosis and occasional inconspicuous nucleoli.

Q3. Special stains and immunohistochemical preparations show the following results: Positive trypsin, BCL-10, and beta-catenin (membranous) with weak and focal positivity for neuroendocrine markers (synaptophysin, chromogranin, and CD56), PAS-D highlighted intracytoplasmic zymogen granules, and CD10 was negative [Figure 3]. What is the final diagnosis?

Well-differentiated neuroendocrine tumor

Solid pseudopapillary neoplasm

Pancreatic adenocarcinoma with predominantly acinar differentiation

Pancreatoblastoma.

(a) Trypsin, (b) Bcl-10, (c) PAS-D, (d) Synaptophysin, (e) Beta-catenin, and (f) CD-10.

Answers to additional quiz questions:

Q2: d; Q3: c

EXPLANATION FOR ADDITIONAL QUIZ QUESTIONS

The H&E stained biopsy cores show a solid cellular neoplasm. This brings up a differential diagnosis of a neuroendocrine neoplasm, solid pseudopapillary neoplasm, pancreatic ACC, and pancreatoblastoma.

Pancreatic well-differentiated neuroendocrine tumors demonstrate a nested, trabecular, or infiltrating (in the case of neuroendocrine carcinoma) growth pattern. Nuclei are round and uniform. The nuclear chromatin classically shows a salt-and-pepper appearance; however, nuclear clumping and prominent nucleoli can also be seen.

Solid pseudopapillary neoplasms can form solid masses with degenerative cystic changes. The cells are loosely cohesive and may form pseudopapillae. Other features that may be present in pancreatic solid pseudopapillary tumors are cytoplasmic vacuoles, hyaline globules, and foamy histiocytes.

Pancreatic ACC shows a solid or lobular growth pattern with acinar formations. Cells show prominent cytoplasm with intracytoplasmic granules. The nuclei show mild anisonucleosis with prominent nucleoli.

Islet cell pseudohypertrophy in chronic pancreatitis can be mass-forming; however, this would occur in a background of atrophy with obliteration of pancreatic parenchyma. Acinar formation and abundant cytoplasm with intracytoplasmic granules are not features of islet cell pseudohypertrophy.

The addition of immunostains and special stains would support an impression of pancreatic ACC. A PAS/D stain highlights intracytoplasmic zymogen granules. Positive staining for trypsin, BCL-10, and membranous beta-catenin are also characteristic of this neoplasm. However, there is also focal, weak positivity for synaptophysin, and raising the possibility that the resection material could show a mixed acinar/neuroendocrine carcinoma, hence, the final diagnosis of “pancreatic carcinoma with predominantly acinar differentiation.”

DISCUSSION

Acinar cell carcinoma (ACC) is a rare pancreatic malignancy that comprises about 1% of pancreatic neoplasia.[1,2] Due to the rarity of this neoplasm, its risk factors and prognosis have not been entirely characterized. Nevertheless, the prognosis appears to be somewhat better than pancreatic ductal adenocarcinoma (PDAC). The overall 5-year survival is approximately 22.6% compared to only 2.8% for PDAC; however, the 5-year survival increases to 47% in cases of localized disease.[3] Like other pancreatic neoplasms, pancreatic ACC may present with abdominal pain, weight loss, or diabetes. Jaundice is another possible presenting symptom but is less common than in pancreatic ductal carcinoma.[2]

Radiographically, pancreatic ACC presents as a solid and hypovascular mass. Fine-needle aspiration (FNA) is a fundamental step in achieving an accurate diagnosis with a sensitivity for detecting malignancy of up to 96%.[4] However, due to its rarity, pancreatic ACC may not be recognized in FNA material. Cytologic preparations will show variably sized loose clusters of cells with acinar formation. Cells show prominent cytoplasm with intracytoplasmic granules. The nuclei show mild anisonucleosis with prominent nucleoli. Of note, there is significant cytological overlap with the far more common pancreatic well-differentiated neuroendocrine tumors, in which pseudorosettes may mimic acini, and occasional prominent nucleoli may be present.[5] If cell block or core biopsy material is available (as in this case), the H&E stained slides show cells arranged in an acinar, glandular, or solid pattern. Nucleoli are usually present but may be inconspicuous. Immunohistochemical stains are also helpful in the diagnosis. Positivity for trypsin, chymotrypsin, BCL-10, and membranous beta-catenin are all characteristic of pancreatic ACC; however, one should also rule out other solid cellular neoplasms, including solid pseudopapillary neoplasm (CD10 positive with nuclear beta-catenin) and pancreatic neuroendocrine tumor/carcinoma (CD56, synaptophysin, and chromogranin positive). Focal positivity of neuroendocrine markers is present in 20–40% of pancreatic ACCs and should not lead to over-interpretation.[1,2] These markers on biopsy or cell block material do raise the possibility of a mixed acinar cell/neuroendocrine carcinoma, because the final resection specimen may show significantly more neuroendocrine differentiation. At present, this distinction is not critical, because it carries no prognostic or treatment implications. In this biopsy material, we favored a diagnosis of “pancreatic carcinoma with predominantly acinar differentiation” pending examination of the entire tumor.


References

La Rosa S Adsay V Albarello L Asioli S Casnedi S Franzi F et al Clinicopathologic study of 62 acinar cell carcinomas of the pancreas: insights into the morphology and immunophenotype and search for prognostic markers Am J Surg Pathol 201236178295 10.1097/PAS.0b013e318263209d 23026929
Klimstra DS Heffess CS Oertel JE Rosai J Acinar cell carcinoma of the pancreas. A clinicopathologic study of 28 cases Am J Surg Pathol 19921681537 10.1097/00000478-199209000-00001 1384374
Luo G Fan Z Gong Y Jin K Yang C Cheng H et al Characteristics and outcomes of pancreatic cancer by histological subtypes Pancreas 20194881722 10.1097/MPA.0000000000001338 31210663
Guarneri G Gasparini G Crippa S Andreasi V Falconi M Diagnostic strategy with a solid pancreatic mass Presse Med 201948e12545 10.1016/j.lpm.2019.02.026 30878333
Stelow EB Bardales RH Shami VM Woon C Presley A Mallery S et al Cytology of pancreatic acinar cell carcinoma Diagn Cytopathol 20063436772 10.1002/dc.20450 16604543


Cytopathologic evaluation of a subcarinal lesion presenting as mass in a smoker

A 72-year-old asymptomatic male with 25 pack-year smoking history underwent low dose computed tomography (CT) of chest for lung cancer screening. The CT imaging reported a subcarinal mass (5.5 × 3.9 × 2.0 cm). Endobronchial ultrasound confirmed a 2 cm subcarinal mass at Station 7. A transbronchial fine-needle aspiration (TB-FNA) of the lesion was performed [Figure 1] that aspirated thick milky and cloudy fluid.

Direct smears of TB-FNA were hypocellular with predominance of cellular debris (a and b) with a few apical fragments of lining cells. These cell fragments demonstrated diagnostic features as seen in zoomed images (arrows) (a1 through a8 and b1 through b8). (a and a1 through a8: Papanicolaou stain; b and b1 through b8: Diff-Quik stain).

QUESTION # 1

What is the most likely interpretation?

Metastatic carcinoma with cystic necrosis

Cystic hygroma (cavernous lymphangioma)

Esophageal duplication cyst

Ciliated lined cyst (bronchogenic cyst/ciliated foregut cyst)

Abscess

ANSWER TO QUESTION # 1

D. Ciliated lined cyst (bronchogenic cyst/ciliated foregut cyst)

The posterior mediastinal lesion was reported on CT scan as well demarcated smooth right subcarinal soft-tissue mass [Figure 2]. Aspirate from this lesion was thick and milky cloudy fluid with cytological features of ciliated lined cyst (bronchogenic cyst/ciliated foregut cyst) (Option d). It showed predominance of degenerated cellular debris admixed with scattered fragments of cells showing ciliary tufts [Figure 1]. These diagnostic cell fragments in this case were seen in direct smears but were few in number and required significant efforts to find them. Processing of the aspirate of such cystic lesion for cytopathological evaluation as liquid-based cytology (LBC) preparation (ThinPrep or SurePath or other similar method) or Cytospin would concentrate detached ciliary tufts (degenerated debris with ciliated cellular fragments of cell tops without nuclei similar to ciliocytophthoria) and occasional intact ciliated cells so that they could be detected with relative ease.[1] The cellular fragments with ciliary tufts could also be detected after diligent efforts in cell-block sections [Figure 3]. Cytomorphologically, these lesion cannot be distinguished from other ciliated lined cysts such as branchial cleft cyst and ciliated foregut cyst[1] without clinicopathological and imaging correlation.

Unenhanced CT scan of thorax showing a smooth margined right subcarinal soft tissue mass (arrows) measuring 3.9 x 5.4 cm in the posterior mediastinum with no airspace consolidation, pleural effusion or pneumothorax.

Cell-block of TBFNA aspirate showing degenerated cellular debris with occasional cell fragments showing cilia (arrows). These cell fragments demonstrate diagnostic features as seen in zoomed images (a through f).

Metastatic carcinoma (Option A) as metastasis of carcinoma of lung, breast, thyroid, and genitourinary tract is relatively common to the mediastinum. Although most of the metastatic carcinoma present as a solid lesion, they can have a cystic component with coagulative necrosis as tumor diathesis, some viable diagnostic malignant cells are expected be found in the sample. It can be challenging to sample such lesions adequately during rapid on-site evaluation due to a high percentage of non-diagnostic necrotic component. Sampling from the periphery of the lesions increases the diagnostic yield. If the cystic lesion does not resolve completely after the aspiration of cyst contents, the residual lesion should be sampled adequately.

Cystic hygroma (cavernous lymphangioma) (Option B) usually occurs in children. It may be located in the neck or axilla, often extending into mediastinum, and rarely present in the lymph node. Large mediastinal lesions may compress lungs, heart, and nerves, even though most lesions are asymptomatic. Microscopically large and irregular lymphovascular spaces are lined by flattened and bland endothelial cells with variable proportion of fibroblastic collagenous stroma. The aspirates are usually hypocellular with relatively non-specific findings including scant endothelial cells with bland morphology with a few scattered lymphocytes and histiocytes in the background of amorphous proteinaceous material.[2]

Esophageal duplication cysts (option C) are usually unilocular; however, they can be multiloculated with mucoid contents. Depending on the type of the cyst-lining, the aspirated cyst contents in addition to the debris show squamous, simple columnar, pseudostratified columnar, or mixed epithelial cells but are devoid of ciliated cells.

Abscess (Option E) shows purulent inflammation with predominance of neutrophils. Grocott-Gomori Methenamine Silver and Gram stain may show organisms such as fungi and bacteria. The organisms may also be seen with Diff-Quik stained direct smears and culture may grow causative organism(s).

QUESTION # 2

What are the diagnostic features of bronchogenic cyst?

Contains tissues derived from all germ cell layers

Contains cartilage and smooth muscle in the wall

Typically lined by respiratory, cuboidal, and/or squamous epithelium

Lined by stratified squamous or gastrointestinal epithelium.

None of the above.

Cystic teratoma is lined by variable mixtures of gastrointestinal, squamous, and respiratory epithelium. It typically contains tissues derived from multiple germ cell layers. Some cases may contain immature tissue with nuclear atypia.

Thyroglossal duct cysts may be lined by respiratory epithelium; however, their cyst walls contain thyroid follicles and lack smooth muscle and cartilage.

Esophageal duplication cysts are lined by stratified squamous or gastrointestinal epithelium. They are usually found to be attached to the esophageal wall and have two smooth muscle layers.

Dermoid cysts are lined by stratified squamous epithelium, contain hair, and other skin appendages with keratinaceous or sebaceous material.

Abscess may mimic bronchogenic cyst. However, an abscess neither will have a true lining nor cartilage and smooth muscle in their wall. They, however, may contain foci of squamous metaplasia. The aspirates predominantly show suppurative material as numerous acute inflammatory cells with variable degenerative changes.

Branchial cleft cysts resemble bronchogenic cyst with ciliated lining; however, they may show squamous metaplasia and lymphoid aggregates with reactive germinal centers. They may also be filled with keratinaceous debris. In contrast to this, the bronchogenic cyst wall shows smooth muscle with variable proportion of seromucinous glands and hyaline cartilage (Option B).

QUESTION # 3

Which of the following malignancies are reported in bronchogenic cyst?

Squamous cell carcinoma

Adenocarcinoma

Large cell carcinoma

Mucoepidermoid carcinoma

All of the above

Bronchogenic cysts are non-neoplastic hamartoma. However, similar to other anatomical tissues, rarely malignancy has been reported in bronchogenic cyst. The risk of malignancy in bronchogenic cyst is reportedly 0.7%.[3] So far, the reported cases include large cell carcinoma,[4] bronchoalveolar carcinoma, adenocarcinoma,[5] schwannoma.[6] mucoepidermoid carcinoma,[7] and squamous cell carcinoma.[8]

Pathophysiology of carcinogenesis in a bronchogenic cyst is not clear. One of the possibility is that the unstable epithelial cells in the cyst wall could have malignant potential leading to carcinoma. Whooley et al. reported squamous cell carcinoma of bronchogenic cyst in the paratracheal region.[8] A few researchers have reported diaphragmatic bronchogenic cyst with mucoepidermoid carcinoma.[7] Schwannomas are also reported in the wall of intrapulmonary bronchogenic cyst.[6]

QUESTION # 4

In the list mentioned below, which is the best diagnostic feature of a bronchogenic cyst?

Degenerated debris

Ciliated cell fragments

Intracystic proteinaceous material

Chronic inflammatory cells in a myxoid background

Numerous acute inflammatory cells

Being a cyst lined by ciliated epithelial lining, the aspirates from the cyst show debris of degenerated exfoliated lining cells with apical fragments of ciliated cells similar to ciliocytophthoria.

QUESTION # 5

Bronchogenic cyst can be seen in which of the following location(s)?

Midline superficial supra-sternal

Lateral to the thyroid

Around the hilum of the lung

Sub-carinal

All the above

Although the most common location of the bronchogenic cyst is superficial midline supra-sternal, it can also be found sub-carinal in the mediastinum, around the hilum of the lungs and lateral to the thyroid gland.

BRIEF REVIEW OF THE TOPIC

Bronchogenic cyst aka non-communicating bronchopulmonary foregut malformation is a relatively rare hamartomatous lesion with prevalence of approximately 1/42,000 admissions.[9] Although it is more commonly seen in pediatric population, it may be detected for the 1st time during adulthood without any sex predilection.

It usually originates as a late budding from the ventral side of embryonic lung or the tracheobronchial tree that occurs between the 26th and the 40th day of gestation. This abnormal bud then becomes a fluid-filled and blind-ending pouch termed bronchogenic cyst. Most of them are asymptomatic at birth and early life. Later in life, it may be symptomatic due to cyst enlargement leading to local compression and pressure on adjacent tissue/organ, occasionally secondary to infection or perforation.[10]

The site of bronchogenic cysts depends on the stage of development when the malformation occurs. The most common site is thorax, for example, mediastinum (early in development), and lung (later in development).[11] However, they have been reported in other sites such as neck, ileal mesentery, intradiafragmatic, intrapericardial, suprasellar, and laryngeal locations.[12] These lesions may remain asymptomatic until they are picked on imaging for screening or otherwise [Figure 2]. Neck lesions are reported to be more common in males than in females (3– 4:1).[13] Subcutaneous bronchogenic cysts are rare and are usually found in children as a small cystic lesion (<3 cm). Zheng et al. reported a 52-year-old woman presenting with a mass confirmed histologically as bronchogenic cyst in the left intergluteal cleft region.[14] Rarely bronchogenic cysts can be located in the pericardial space. Intrapericardial cysts consist of 27% of the bronchogenic cysts and often difficult to diagnose even with newer imaging modalities.[15] Intramural bronchogenic cysts of the esophagus are located in the upper mediastinum and are very rare.[16]

Bronchogenic cysts are lined by pseudostratified columnar or cuboidal ciliated (respiratory) epithelium. The cyst wall often contains smooth muscle fibers, elastic fibers, submucosal bronchial glands, and hyaline cartilage. The cyst wall helps to distinguish them from other ciliated lined cysts such as foregut cyst, duplication cyst, and branchial cleft cyst. The role of cytology is crucial to properly triage such patients. Endoscopic ultrasound-guided fine-needle aspiration (EUSFNA) in the current case was consistent with ciliated lined cystic lesion without malignant cells [Figure 1]. The definitive diagnosis of bronchogenic cysts depends on histological examination of the surgical specimen with hyaline cartilage, bronchial glands, and/or smooth muscle in the wall of the lesion.

Complete removal of the cyst, especially if symptomatic due to possibility of complications in the future, is the preferred recommendation. Approximately 50% of bronchogenic cysts present with pain.[17] Others may cause respiratory distress, cough, dyspnea, hemoptysis, dysphagia, nausea, vomiting, etc. Fever is a sign of infection and warrants a quick diagnosis with medical treatment and surgical removal as clinically indicated. Usually long-term prognosis with complete excision is assuring. However, incomplete excision may increase risk of recurrence.

The efficacy of EUS-FNA of cystic lesions partly depends on the site, size, and characteristics of the target tissue as well as on the expertise, training, and coordination between the procedure-performer and the cytology team.[18] Acquisition of diagnostic samples is approached in different ways depending on the site and type of the lesion. Cytological samples are ideal for cyst fluid analysis, IHC, and molecular testing as indicated. As observed in the present case, the direct smears are invaluable, both for immediate assessment and also for the final morphological interpretation. The needle rinses can be processed as LBC and cell-block. Cell-block increases the diagnostic accuracy of EUS-FNA in general.[1]

Bronchogenic cyst may be difficult to aspirate by TB-FNA, especially when the cyst content is thickly mucoid with debris. As observed in our case, intact ciliated cells could not be detected; however, there were many cell top fragments of the ciliated cells similar to ciliocytophthoria in Papanicolaou stained and Diff Quik stained direct smears [Figures 1 and 3]. These features would be relatively easily detectable in cytological preparations made with concentration methods such as LBC preparations including Surepath,[1] ThinPrep, and Cytospin. Malignant cells or granuloma were absent in our case. Cell-block also showed similar findings; however, relatively few ciliated cell fragments showed significantly inferior morphology as compared to that seen in cytology preparations [Figure 3].

SUMMARY

A rare presentation or an uncommon lesion can be a clinical challenge. FNA cytology is minimally invasive modality for evaluation of space occupying lesions and for exclusion of malignancy. Although non-specific, the cytomorphological features in correlation with clinical and imaging details were consistent with bronchogenic cyst by detecting clues for respiratory type epithelium in the present case reported initially as mass lesion on imaging [Figure 2]. The detection of cell fragments with cilia similar to ciliocytophthoria [Figures 1 and 3] as diagnostic clue would be facilitated by proper processing with inclusion of concentration methodology such as LBC (SurePath/ThinPrep) or Cytospin.[1]

Answers for Question 2 through 5:

2. B

3. E

4. B

5. E


References

Dua KS Vijayapal AS Kengis J Shidham VB Ciliated foregut cyst of the pancreas: Preoperative diagnosis using endoscopic ultrasound guided fine needle aspiration cytolog-a case report with a review of the literature Cytojournal 2009622 10.4103/1742-6413.56362 19876385
Abdelkader A Hunt B Hartley CP Panarelli NC Giorgadze T Cystic lesions of the pancreas: Differential diagnosis and cytologichistologic correlation Arch Pathol Lab Med 20201444761 10.5858/arpa.2019-0308-RA 31538798
Kirmani B Kirmani B Sogliani F Should asymptomatic bronchogenic cysts in adults be treated conservatively or with surgery? Interact Cardiovasc Thorac Surg 20101164959 10.1510/icvts.2010.233114 20709698
Jakopovic M Slobodnjak Z Krizanac S Samarzija M Large cell carcinoma arising in bronchogenic cyst J Thorac Cardiovasc Surg 20051306102 10.1016/j.jtcvs.2004.12.022 16077456
Gómez-Hernández T Novoa N Aranda JL Jiménez-López MF Enteric adenocarcinoma arising from a bronchogenic cyst Arch Bronconeumol 2017535234 10.1016/j.arbr.2017.02.018 28318614
Aydogdu K Findik G Kaya S Koksal D Demirag F Schwannoma arising in a bronchogenic cyst wall Asian Cardiovasc Thorac Ann 20152332831 10.1177/0218492314529292 24719160
Taira N Kawasaki H Atsumi E Ichi T Kawabata T Saio M et al Mucoepidermoid carcinoma of arising from a bronchogenic cyst of the diaphragm Ann Thorac Cardiovasc Surg 20182424750 10.5761/atcs.cr.17-00131 29367500
Whooley J White A, Soo A Bronchogenic cyst: A rare case of malignant transformation BMJ Case Rep 202215e248916 10.1136/bcr-2022-248916 35379683
Coselli MP de Ipolyi P Bloss RS Diaz RF Fitzgerald JB Bronchogenic cysts above and below the diaphragm: Report of eight cases Ann Thorac Surg 1987444914 10.1016/S0003-4975(10)62106-6 3675053
Yang X Guo K Bronchogenic cyst of stomach: Two cases report and review of the English literature Wien Klin Wochenschr 20131252837 10.1007/s00508-013-0352-0 23579880
Matsuoka T Sugi K Matsuda E Okabe K Hirazawa K Azuma T Intrapulmonary and mediastinal, double bronchogenic cysts; Report of a case Kyobu Geka 20086141922 Japanese
Zarogoulidis P Tsakiridis K Vagionas A Hohenforst-Schmidt W Zaric B Tryfon S et al Bronchogenic cyst or lung cancer. Only biopsy can tell Respir Med Case Rep 202032101328 10.1016/j.rmcr.2020.101328 33457198
Teissier N Elmaleh-Bergès M Ferkdadji L François M Van den Abbeele T Cervical bronchogenic cysts: Usual and unusual clinical presentations Arch Otolaryngol Head Neck Surg 200813411659 10.1001/archotol.134.11.1165 19015445
Zheng CY Su SY Huang RB Giant subcutaneous bronchogenic cyst in the intergluteal cleft region of an adult: A case report and literature review BMC Med Imaging 202222126 10.1186/s12880-022-00853-y 35842586
Gutiérrez GS Gutiérrez FG Bastianelli GA Vaccarino GN Bronchogenic cyst in an unusual location Asian Cardiovasc Thorac Ann 202129446 10.1177/0218492320960271 32938205
Erbenová A Placrová B Špurková Z Horák P Bronchogenic cyst of gastric cardia-case report and literature review Rozhl Chir 202110050711 10.33699/PIS.2021.100.10.507-511 35021843
Limaïem F Ayadi-Kaddour A Djilani H Kilani T El Mezni F Pulmonary and mediastinal bronchogenic cysts: A clinicopathologic study of 33 cases Lung 20081865561 10.1007/s00408-007-9056-4 18064522
Jenssen C Möller K Wagner S Sarbia M Endoscopic ultrasound-guided biopsy: Diagnostic yield, pitfalls, quality management Z Gastroenterol 200846590600 10.1055/s-2008-1027413 18537087


Dumbbell-shaped swelling of the ear lobe: Cytomorphological clues

10 cm nodule (growing in size, firm, non-tender, dumbbell-shaped) on the left ear lobe in a 22-year-old male after ear-piercing 6 months ago. Fine-needle aspiration showed scant paucicellular material with a few spindle-shaped cells with scant to absent cytoplasm. Eosinophilic collagen-like material was seen in myxoid background [Figure 1].

(a) Paucicellular smear with a few spindle cells (MGG, ×400). (b) with eosinophilic collagen-like material (MGG, ×400). (c) Gross: Skin-covered soft tissue with 10 cm nodule. (d) Hyperkeratotic stratified squamous epithelium with large dense bundles of glassy collagen in mid to deep dermis showed (H & E, ×100). Inset: Masson’s trichrome stain highlights the thick collagen bundles in the mid-dermis (×200).

Q1. What is your interpretation?

Keloid

Hypertrophic scar

Nodular fasciitis

Fibromatosis

Answer: Q1-A. Keloid.

Cytomorphological differential diagnosis of the present case includes keloid, hypertrophic scar, fibromatosis, and nodular fasciitis. On cytology, keloid shows sparse fibroblastic spindle to oval-shaped cells, either single or in clusters along with thick, hyalinized, glassy, and eosinophilic collagen bundles named “keloid collagen” in a mucinous ground substance.[1] Keloid should be differentiated from hypertrophic scar, which usually occurs within 4–8 weeks of injury, is limited to the site of injury, and can regress gradually over time.[2] Cytologically, the hypertrophic scar has similar features as that of keloid but tends to be more cellular than keloid.[3] Hypertrophic scar has fine fibrillary collagen and occasional foreign body type of giant cells.[3]

Nodular fasciitis occurs most commonly in young adults and is more common in the subcutaneous tissue of the upper extremities, trunk, head, and neck. It is a self-limiting fibrous neoplasm.[4] Nodular fasciitis is cellular and comprises dispersed cells mixed with a tight cluster of spindle-shaped cells showing marked pleomorphism with fusiform nuclei and cytoplasmic processes in a background of inflammatory cells and myxoid stroma.[1,5]

Fibromatosis occurs almost at every site of the body.[4] The exact cause is unknown.[4] Cytology of fibromatosis reveals variable cellular smear with the scattered spindle to fusiform cell clusters, stromal fragments, and collagen matrix material.[5]

Q2. Which of the following characteristic feature distinguishes keloid from the hypertrophic scar?

Spindle- or oval-shaped fibroblastic cells

Bland fibroblastic cells with bipolar cytoplasmic extension

Mucinous ground substance

Sparse chronic inflammatory background

Answer: Q2-C. Mucinous ground substance.

Both keloid and hypertrophic scars show similar cytomorphological features. Mucinous ground substance is scant or absent in hypertrophic scar as opposed to keloid.[3]

FURTHER FOLLOW-UP OF THE CASE

The mass was surgically excised. Grossly, it measured 10 cm in maximum dimension, and externally, it was skin-covered [Figure 1c]. On the cut section, it was solid and grayish-white. Histopathological sections showed hyperorthokeratotic stratified squamous epithelium. Superficial dermis showed fibroblastic cells arranged parallel to the epidermis. Mid to deep dermis showed large dense bundles of glassy collagen [Figure 1d]. Masson’s trichrome stain highlighted the thick collagen bundles in the mid-dermis (inset). A final diagnosis of keloid was made.

The patient was on regular follow-up. There was no recurrence after 6 months of follow-up.

Q3. Which type(s) of collagen is found in keloids?

Type I

Type II

Type III

Types I and III

Answer: Q3-D. Types I and III.

Keloid is characterized by haphazardly arranged large, thick, Types I and III hypocellular collagen bundles with no nodules, or excess myofibroblasts.[2] Hypertrophic scar has primarily fine, well-organized, and wavy Type III collagen bundles oriented parallel to epidermis surface with abundant nodules containing myofibroblasts.[2]

BRIEF REVIEW OF THE TOPIC

Keloid is a reactive condition resulting from excessive scar formation due to an aberrant healing process.[3] Various causes lead to the formation of excessive scars, including trauma, surgery, and burn.[3] Keloid usually occurs in the head-and-neck region, ear lobes, upper chest, and arms.[3] It presents as raised, well-circumscribed, firm nodule, often pruritic, and painful and most do not regress spontaneously.[3]

Keloids are most frequent among African population and less common in Caucasians.[6] Positive family history is not uncommon and probably reflects a genetic predisposition to keloid formation.[6] The genetic study for keloid showed susceptibility loci on chromosomes 2q23 and 7p11.[7] Different studies revealed that multiple genes are associated with keloid formation.[6,7] The role of Th2 inflammatory response with the production of interleukins (IL)-4, IL-5, IL-10, and IL-13 has been implicated in the pathogenesis of keloid.[8] Fibrogenic response to growth factors such as transforming growth factor-beta and platelet-derived growth factor (PDGF) also plays an integral role in the formation of keloids.[8]

Although histopathological features of keloid have been aptly described in the literature, cytomorphology of keloid is rarely reported in the literature.[1,5] As most of the cases of keloid are clinically evident, FNAC is rarely done in these lesions. FNAC may be done only in clinically uncertain cases. Hypertrophic scar, nodular fasciitis, and fibromatosis were the differential diagnoses considered in the present case. The cytomorphological features of keloid and its differential diagnoses are summarized in [Table 1].

Cytomorphology of keloid and its differential diagnoses.

Cytomorphological features Keloid Hypertrophic scar Nodular fasciitis Fibromatosis
Cellularity Sparsely cellular Cellular Highly cellular Depending on age and location, the lesion may be cellular to acellular
Cell morphology Fibroblastic spindle to oval-shaped cells, either single or in clusters with bipolar cytoplasmic extensions Same as keloid Polymorphic cells have spindle, round, and oval to triangular-shaped cells. Cells have cytoplasmic processes and round to ovoid nuclei Fibroblastic oval to spindle-shaped cells in clusters with tapering cytoplasmic processes
Collagen Thick, hyalinized, eosinophilic “keloidal collagen” Fine fibrillar collagen Absent Densely eosinophilic collagen
Mucinous ground substance Present Scant or absent Myxoid background Absent
Chronic inflammatory cells Sparse Sparse Present Sparse
Giant cells Absent Occasional foreign body type giant cells Few binucleate/trinucleate cells Occasional multinucleated giant cells

Although hypertrophic scar and nodular fasciitis have traumatic etiology, they can regress without any surgical management. Keloid and fibromatosis require surgical excision and can recur if incompletely excised.[2,4] In addition to surgery, intralesional steroid injection, cryotherapy, laser removal, radiotherapy, and silicone gel sheeting are some of the most popular treatment modalities for keloid.[8] Less widely utilized therapies include topical imiquimod and antimetabolites such as 5-fluorouracil and bleomycin.[8]

In the present case, the cytological clues favoring keloid were paucicellular smears with the presence of eosinophilic collagen. The mass was surgically excised and there was no recurrence after 6 months of follow-up.

SUMMARY

Keloid is a common lesion with distinct cytological features. The characteristic cytological features include paucicellular spindle cells with eosinophilic collagen. The common cytomorphological differential diagnoses include a hypertrophic scar, nodular fasciitis, and fibromatosis.

COMPETING INTEREST STATEMENT BY ALL AUTHORS

The authors have no conflicts of interest.

AUTHORSHIP STATEMENT BY ALL AUTHORS

Concept: Biswajit Dey and Jitendra Singh Nigam. Design: Jyotsna Naresh Bharti, Biswajit Dey, Jitendra Singh Nigam, and Pooja Garg. Definition of Intellectual Content: Jyotsna Naresh Bharti and Biswajit Dey. Literature Search: Jyotsna Naresh Bharti, Biswajit Dey, Jitendra Singh Nigam, and Pooja Garg. Data Acquisition: Biswajit Dey and Pooja Garg. Data Analysis: Jyotsna Naresh Bharti, Biswajit Dey, Jitendra Singh Nigam, and Pooja Garg. Manuscript Preparation: Jyotsna Naresh Bharti, Biswajit Dey, Jitendra Singh Nigam, and Pooja Garg. Manuscript editing and Review: Jitendra Singh Nigam, Jyotsna Naresh Bharti, and Biswajit Dey.

ETHICS STATEMENT BY ALL AUTHORS

The informed and written consent was obtained from the patient. The case was submitted without identifiers.


References

Soni PB Verma AK Chandoke RK Nigam JS A prospective study of soft tissue tumors histocytopathology correlation Patholog Res Int 20142014678628 10.1155/2014/678628 24876987
Gauglitz GG Korting CH Pavicic T Ruzicka T Jeschke GM Hypertrophic scarring and keloids: Pathomechanisms and current and emerging treatment strategies Mol Med 20111711325 10.2119/molmed.2009.00153 20927486
Poflee S Munshi M Lele V Bobhate S Fine needle aspiration cytology in keloid and hypertrophic scar J Cytol 200421111
Agaram NP Lin O Cytology of soft tissue, bone, and skin Bibbo M Wilbur DC Comprehensive Cytopathology 4th ed Philadelphia, PA Elsevier 2015363402
Tanwar P Gupta N Vashishta RK Singh G Fine needle aspiration cytology in fibromatosis J Cytol 201229668 10.4103/0970-9371.93221 22438623
Bran GM Goessler UR Hormann K Riedel F Sadick H Keloids: Current concepts of pathogenesis (review) Int J Mol Med 20092428393 10.3892/ijmm_00000231 19639219
Marneros AG Norris JE Watanabe S Reichenberger E Owen BR Genome scans provide evidence for keloid susceptibility loci on chromosomes 2q23 and 7p11 J Invest Dermatol 2004112112632 10.1111/j.0022-202X.2004.22327.x 15140214
Hunasgi S Koneru A Vanishree M Shamala R Keloid: A case report and review of pathophysiology and differences between keloid and hypertrophic scars J Oral Maxillofac Pathol 20131711620 10.4103/0973-029X.110701 23798844


Ulcerated scalp nodule in elderly female: Cytomorphological clues and pitfalls for diagnosis

A 63-year-old female with ulcerated scalp swelling (2.0 × 2.0 cm × 2.0 cm, not attached to the underlying bone, without regional lymphadenopathy) since 2 years with frequent bleeding on trivial trauma. Fine-needle aspirate showed features as shown in Figure 1.

FNA aspirate showing squamous and basaloid cells (a, PAP ×100; b, MGG ×100) with blotchy keratinous material (c, MGG ×100). Squamous cells without atypia showed moderate cytoplasm. (d, MGG ×100).

QUESTION

Q1: What is your interpretation?

Ulcerated pilar cyst

Pilomatrixoma

Trichoepithelioma

Proliferating trichilemmal tumor (PTT)

Squamous cell carcinoma d

ANSWER

d. Proliferating trichilemmal tumor.

Explanation: The fine-needle aspiration cytology (FNAC) smears were moderately cellular and showed anucleate and nucleated squamous cells, basaloid cells [Figure 1a], abrupt keratinization [Figure 1b], and blotchy keratinous material [Figure 1c]. Squamous cells did not show any significant cytological atypia [Figure 1d]. The basaloid cells and squamoid cells were almost equal in proportion. The basaloid cells did not show any peripheral palisading. Ghost cells, calcification, cholesterol clefts, and foreign body giant cells were absent.[1] A diagnosis of low-grade squamous neoplasm was rendered.

FOLLOW-UP OF PRESENT CASE

Wide local excision was advised for histopathological confirmation. On gross examination, the lesion was partly skin-covered; nodular, measuring 2.5 × 2.0 × 2.0 cm. The overlying skin was ulcerated. On the cut section, the tumor was solid and grayish-white. Microscopy revealed a lobulated intradermal mass of squamous epithelium [Figure 2]. Widespread trichilemmal keratinization was noted along with the absence of a granular cell layer [Figure 3]. The squamous cells did not show any atypia or mitotic figures. The diagnosis of a low-grade (benign) proliferating trichilemmal tumor was rendered. Post-surgery, 2 years follow-up did not show any recurrence.

Lobules of squamoid cells with pushing borders and cystic spaces filled with keratinous material. No evidence of infiltration was seen (H&E 40×).

Trichilemmal type keratinization showing extensive glassy keratinous material without granular layer. Squamous cells do not show any atypia (H&E 100×).

During fine-needle aspiration cytological evaluation of any skin nodule, the following differential diagnoses should be considered.

Pilar cyst: Pilar cyst is defined as a cyst containing keratin and its breakdown products. It arises preferentially in areas of high hair follicle concentrations; therefore, 90% of cases occur on the scalp. They are solitary in 30% of cases and multiple in 70% of cases. These are cystic nodular lesions with a smooth external surface. Young pilar cysts show abundant blotchy keratin with or without calcification and inflammation. Older cysts show necrotic debris with cholesterol crystals and inflammatory cells

Pilomatrixoma is a benign cutaneous adnexal tumor having differentiation toward the hair follicle matrix with a predilection for the head-and-neck region of children and young adults. However, a bimodal pattern with the first peak in the first decade and the second in the sixth decade of life, along with a female preponderance is observed.[2] Smears of pilomatrixomas are moderately cellular and comprised primarily of dense clusters of small- and medium-sized basaloid cells with overlapping nuclei, sheets of ghost cells, nucleate and anucleate squamous cells, and keratin fragments.[3] The background is dirty and rich in granular calcium and shows many multinucleated giant cells. PTT generally does not show many multinucleate giant cells. Ghost cells characteristic of pilomatrixoma is not seen in PTT

Conventional trichoepithelioma is usually seen in children and young adults as multiple, small, 2–4 mm papules. Giant solitary trichoepithelioma, however, occurs in elderly individuals and arises most commonly on the thigh and perianal region. On fine-needle aspiration cytology, it shows fronds of basaloid epithelial cells with abrupt keratinization, papillary mesenchymal body, and melanin pigmentation.[4] Although the present lesion also showed basaloid cells clusters with abrupt keratinization and melanin pigment, it did not show mesenchymal bodies

Proliferating trichilemmal tumor (PTT), also referred to as proliferating pilar tumor (PPT), is a tumor originating from the outer root sheath of a hair follicle. The histologic hallmark of PTT is the presence of trichilemmal keratinization.[5] It most commonly occurs on the scalp during the 4th–8th decades of life with a distinct predilection for women. Wilson-Jones first described PTT in 1966 as an entity that can clinically and histologically simulate squamous cell carcinoma (SCC).[6] A clinicopathological study of 76 cases divided PPT into three groups on the degree of stromal invasion and the level of cellular atypia.[7] Group 1 lesions (PPTs) behave benignly, Group 2 tumors (low-grade malignant PTT -LMPPTs) have a small risk of local recurrence, and Group 3 neoplasms (high-grade malignant PTT – HMPPTs) have the potential for regional recurrence and metastasis. Low- and high-grade malignant PTT shows additional marked nuclear anaplasia. The histological features are well recognized. However, characteristic fine-needle aspiration cytological findings of proliferating trichilemmal tumors are documented by a few authors only.[5,8] On cytology, diagnosis of PTT is sometimes difficult and misdiagnosed as adnexal neoplasm.[9]

The diagnosis of SCC was ruled out as squamous cells did not show any cellular atypia, pleomorphism, dyskeratotic cells, or tumor diathesis. Reactive epithelial atypia in inflamed pilar cysts may appear worrisome and raise the suspicion of SCC.

For cytological diagnosis of PTT, smears should show the presence of blotchy keratin, basaloid cells, squamous epithelial cells, and trichilemmal keratinization. However, these features may not be present in smears always and may lead to diagnostic dilemmas.

The presence of keratin material only may lead to misdiagnosis of epidermoid cysts or pilar cysts.

Reactive epithelial atypia in inflamed cysts can look worrisome. Smears with basaloid cells with abrupt keratinization may get erroneously diagnosed as trichoepithelioma or keratotic BCC.

It is not possible to differentiate on FNAC between benign and low-grade malignant PTT as smears in both have similar cytomorphological features. However, cytology of high-grade malignant PTT shows trichilemmal keratinization, blotchy keratin, basaloid cells, and atypical squamous epithelial cells.

SUMMARY

The cytological diagnosis of PTT can be made in the presence of blotchy keratin, basaloid cells, squamous epithelial cells, and trichilemmal keratinization. However, these features may not be present in cytological smears and may lead to diagnostic dilemmas. Furthermore, the distinction between benign PPT (Group I) and low malignant PTT (Group II) is not possible on FNAC alone. FNA plays an important role to exclude SCC and high malignant PTT and to decide surgical management. PTT without atypia has a benign behavior, wide local excision of the lesion is recommended to prevent a recurrence.


References

Folpe AL Reisenauer AK Mentzel T Rütten A Solomon AR Proliferating trichilemmal tumors: Clinicopathologic evaluation is a guide to biologic behavior J Cutan Pathol 2003304928 10.1034/j.1600-0560.2003.00041.x 12950500
Julian CG Bowers PW A clinical review of 209 pilomatricomas J Am Acad Dermatol 1998391915 10.1016/S0190-9622(98)70073-8
Bansal C Handa U Mohan H Fine needle aspiration cytology of pilomatrixoma J Cytol 20112816 10.4103/0970-9371.76940 21552399
Krishnamurthy J Divya KN The cytology of giant solitary trichoepitheliomas J Cytol 20102799101 10.4103/0970-9371.71874 21187885
Shet T Rege J Naik L Cytodiagnosis of simple and proliferating trichilemmal cysts Acta Cytol 2001455828 10.1159/000327868 11480722
Brownstein MH Arluk DJ Proliferating trichilemmal cyst: A simulant of squamous cell carcinoma Cancer 198148120714 10.1002/1097-0142(19810901)48:5<1207::AID-CNCR2820480526>3.0.CO;2-1
Ye J Nappi O Swanson PE Patterson JW Wick MR Proliferating pilar tumors: A clinicopathologic study of 76 cases with a proposal for a definition of benign and malignant variants Am J Clin Pathol 200412256674 10.1309/0XLEGFQ64XYJU4G6 15487455
Kini JR Kini H Fine-needle aspiration cytology in the diagnosis of malignant proliferating trichilemmal tumor: Report of a case and review of the literature Diagn Cytopathol 2009377447 10.1002/dc.21100 19405112
Uppada R Rout S Bora N Pullela RV An interesting case report of ulcerated proliferating pilar tumor (PPT) mimicking squamous cell carcinoma J NTR Univ Health Sci 201431113 10.4103/2277-8632.134858


Fine needle aspiration of hematolymphoid lesions of the thyroid: Onsite adequacy and ancillary testing

CASE REPORT

A 7-year-old girl presented with an enlargement of the thyroid gland. Thyroid hormones and serologic testing were normal. A thyroid ultrasound showed a right 0.8 cm thyroid nodule with microcalcifications and well-defined borders. Fine needle aspiration (FNA) revealed polymorphic lymphocytes without Hurthle cells [Figure 1a-d]. Flow cytometry showed a population of CD2+, CD5+, CD8+, CD7+, CD4+, and CD1a+ cells. Immunohistochemistry on cell block section showed immunoreactivity for TdT without immunoreactivity for CD34.

Polymorphic population of lymphocytes and histiocytes, with squamoid cells (yellow arrows in a and b). A: PAP stain ×10; (b) PAP stain ×100; (c) Higher magnification of histiocytes and polymorphic lymphocytes (red arrowhead- lymphoblast, green arrowhead- small lymphocytes) (Diff Quick stain ×100); (d) Polymorphic population of small lymphocytes (red arrowhead- lymphoblast, green arrowhead- lymphocytes) (PAP stain ×100).

QUESTION 1

What is your interpretation?

Colloid nodule

Primary thyroid extranodal NK/T-cell lymphoma

Extranodal mucosa-associated lymphoid tissue (MALT) lymphoma of the thyroid

Squamous cell carcinoma chronic inflammation

Ectopic intrathyroidal thymic tissue

Answer to question 1: Option E (ectopic intrathyroidal thymic tissue).

The aspirates were moderately cellular and showed predominantly polymorphic lymphocytes with occasional histiocytes. The lymphocytes were predominantly small to medium in size with mature clumped chromatin and a smaller subset featuring large nuclei with open chromatin (lymphoblasts). Furthermore, evaluation of the aspirate smear showed occasional aggregates of squamoid cells consistent with Hassall corpuscles [Figure 1].

Immunophenotyping by flow cytometry analysis [Table 1] revealed the presence of CD45/SS populations consistent with mature lymphocytes (46%), monocytes (1%), granulocytes (3%), and a CD45dim region (29%). The gated CD45dim area was comprised largely of immature/maturing T-cell subsets that were CD3 negative but expressed CD2, CD5, CD7 and variably expressed CD4+, CD8+, and CD4+/CD8+ subsets, along with partial CD1a expression. The gated CD45brt lymphocytes were predominantly mature CD3+ mixed T cells that displayed normal CD4/CD8 content (4:8 ratio = 1.9) without aberrant expression of associated markers. The gated minority granulocytes and monocytes appeared immunophenotypically normal. The flow cytometry interpretation illustrated a cellular makeup that was consistent with normal thymus tissue and likely represented ectopic intrathyroidal thymic tissue.[1] Flow cytometry was negative for any obvious abnormal or significant clonal populations [Table 1].

Flow cytometry report on needle rinse of FNA of the lesion.

Target cell population MoAb/CD# CD45dim Lymphs CD45brt Lymphs Gate type: CD45 versus SS-log
% of total cell population 29 46 Sample preparation: density-gradient; mononuclear isolation with RBC lysis
Commonly found on:
CD45 64 100 Pan-Leukocytes
T cell markers
CD1a 34 17 Cortical thymocytes
CD5+ 91 97 Pan T cells
CD7 95 90 Pan T cells
CD2 94 99 Pan T cells
CD3 4 98 Mature T cells
CD4 64 65 Helper/Inducer T cells
CD8 30 38 Suppressor/cytotoxic T cells
CD4+/CD8+ 27 6 T cell precursor subset
CD56 1 1 NK cells; T cell subset
CD57 0 2 NK cell subset; T cell subset
B cell markers
HLA-DR 10 10 B, NK, Myeloid, Activated T cells
CD10 3 0 Early B lineage, Early T cells
CD20 1 0 Late B lineage cells
CD22 2 0 Pan-B cells
CD19 2 1 Pan-B cells
Additional markers
CD38 95 76 Broad lineage; Activation marker
CD34 25 0 Early precursors, Stem cells

Markers for Myeloid (CD15, CD16, CD11b, CD14, CD13, CD33, CD34, CD64); Monocytic, Platelet/Megakaryocytic (CD41, 61+/CD14-, CD61, CD36); and Erythroid Markers (CD36, Gly A) negative. brt: Bright, Lymphs: Lymphocytes, MoAb/CD#: Monoclonal antibody/CD number

Cellblock sections showed polymorphic lymphocytes that were immunoreactive for TdT without immunoreactivity for CD34 [Figure 2].

The lymphocytes were immunoreactive for TdT and nonimmunoreactive for CD34 (a and b, ×40, immunohistochemistry on cell-block sections).

The aspirates did not show thyroid follicular cells and any colloid in the background (option A). Primary thyroid lymphomas are extremely rare and account for <1% of cases of extranodal lymphomas.[2] Most primary thyroid lymphomas are non-Hodgkin lymphomas of B-cell origin, and primary thyroid extranodal NK/T-cell lymphomas are exceedingly rare.[2] Cytologic examination would reveal a diffusely infiltrating small to medium-sized lymphoid cells, prominent mitotic figures, and areas of necrosis with apoptotic debris,[2] not observed in the current case (option B). Extranodal marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue (MALT) type is characterized by morphologically heterogenous B-cells and may be encountered in 0.5% of patients with Hashimoto thyroiditis.[3-5] It is postulated to occur as a response to autoimmune processes, such as in patients with Hashimoto thyroiditis and patients with Helicobacter pylori infection.[6] FNA would show thyroid follicular atrophy with Hurthle cell changes and follicular cells with lymphocytic infiltration as lymphoepithelial structures[7] which were not detected in the current case (option C). Primary squamous cell carcinoma, typically common in elderly patients, is a rare entity which carries poor prognosis (may be a component of anaplastic thyroid carcinoma).[8] FNA would reveal follicular cells infiltrated or replaced by morphologically malignant cells. The malignant cells may be cohesive and arranged in sheets, nests, cords, islands, or a trabeculae pattern.[9] The cells would exhibit large, pleomorphic, hyperchromatic to vesicular nuclei which may show eosinophilic nucleoli and abundant eosinophilic cytoplasm. Individual keratinization, mitotic figures (including the atypical forms), and tumor necrosis may also be noted.[9] The cell-block sections may show intercellular bridges. This morphology was not evident in the current case (option D).

QUESTION 2

The thymus is the primary organ of T lymphocyte development and partly develops from the endoderm of the third pharyngeal pouch. Which of the following organs have the same embryologic origin as the thymus?

Thyroid gland

Superior parathyroid glands

Inferior parathyroid glands

Ultimobranchial bodies

Answer to question 2: Option C (inferior parathyroid glands).

The inferior parathyroid gland originates from the endoderm of the third pharyngeal pouch just like the thymus.[10] The thyroid originates from the endodermal cells of the primitive pharynx between the first and second pharyngeal pouches (option A),[11] the superior parathyroid glands originate from the fourth pharyngeal pouch (option B),[10] and the ultimobranchial bodies which give rise to the calcitonin producing parafollicular cells of the thyroid originate from the fourth pharyngeal pouch (option D).[12]

QUESTION 3

According to the World Health Organization classification, the obligatory criteria for diagnosing type B1 thymoma includes:

Occurrence of bland spindle-shaped epithelial cells (at least focally) and paucity or absence of immature (TdT+) T cells throughout the tumor

Increased numbers of single or clustered polygonal or dendritic epithelial cells intermingled with abundant immature T cells

Thymus-like architecture and cytology with abundance of immature T cells, areas of medullary differentiation (medullary islands), and paucity of polygonal or dendritic epithelia cells without clustering (i.e.,<3 contiguous epithelial cells)

Occurrence of bland, spindle-shaped epithelial cells (at least focally), and abundance of immature (TdT+) T cells focally or throughout tumor

Sheets of polygonal slightly to moderately atypical epithelial cells; absent or rare intercellular bridges; paucity or absence of intermingled TdT+ T cells

Answer to question 3: Option C (thymus-like architecture and cytology with abundance of immature T cells, areas of medullary differentiation (medullary islands), and paucity of polygonal or dendritic epithelia cells without clustering (i.e.,<3 contiguous epithelial cells).[13]

Occurrence of bland spindle-shaped epithelial cells (at least focally) and paucity or absence of immature (TdT+) T cells throughout the tumor is classified as Type A thymoma (option A).[13]

Increased numbers of single or clustered polygonal or dendritic epithelial cells intermingled with abundant immature T cells are classified as type B2 thymoma (option B).[13] Occurrence of bland, spindle-shaped epithelial cells (at least focally); abundance of immature (TdT+) T cells focally or throughout tumor is classified as type AB thymoma (option D),[13] and sheets of polygonal slightly to moderately atypical epithelial cells; absent or rare intercellular bridges; paucity or absence of intermingled TdT+ T cells is classified as type B3 thymoma (option E).[13]

QUESTION 4

TdT immunostain is useful in delineating which of the following combinations?

Thymic carcinomas, thymomas, and epithelial cells of normal thymus

Immature T cells of normal thymus, >90% of thymomas, and neoplastic T cells of T lymphoblastic lymphoma

Epithelial cells of normal thymus, thymomas, thymic carcinomas, neuroendocrine tumors, many germ cell tumors, and dendritic cell tumors

Normal and neoplastic B cells and epithelial cells of Type A and AB thymoma

Answer to Question 4: Option B (immature T cells of normal thymus, >90% of thymomas, and neoplastic T cells of T lymphoblastic lymphoma).

Thymic carcinomas, thymomas, and epithelial cells of normal thymus may be identified with cytokeratins (option A).[13] Epithelial cells of normal thymus, thymomas, thymic carcinomas, neuroendocrine tumors, many germ cell tumors, and dendritic cell tumors may be identified with cytokeratins (option C),[13] and normal and neoplastic B cells and epithelial cells of type A and AB thymoma may be identified with CD20 (option D).[13]

BRIEF REVIEW OF THE TOPIC

Thyroid nodules are uncommon in the pediatric population as compared to adults and account for 0.2–2% of cases.[14] However, 20–73% of nodules found in children are malignant[15] and exhibit higher risks of regional and distant metastases than in the adult population.[16] Thyroid cancer is the eighth most common cancer in adolescents and the second most common cancer in girls.[16]

Advances in ultrasonographic examinations have resulted in increased numbers of incidental thyroid gland lesions than previously reported.[17] Ultrasound findings that suggest malignancy include microcalcifications, irregular borders, and hypoechogenicity.[18] Features which indicate malignancy may be determined using the thyroid imaging reporting and data system (TI-RADS). TI-RADS refers to any of several risk stratification systems for thyroid lesions usually based on ultrasound characteristics.[19] Points are assigned to the lesion based on its composition, echogenicity, shape, margin, and echogenic foci. A score of 0 is considered benign (TR1), 2 is considered not suspicious (TR2), 3 is considered mildly suspicious (TR3), 4 to 6 is considered moderately suspicious (TR4), and 7 points or more is considered highly suspicious (TR5).[1,20,21] Identification of some of these features on ultrasound prompts the provider to pursue testing to rule out malignancy.[18] Rarely, ectopic non-thyroidal tissue may be detected on ultrasound as a thyroid nodule with concerning features. Thus, in accordance with current guidelines, any thyroid lesion found in a child, except for pure cysts, requires thorough evaluation including FNA.[22]

The differential diagnosis for thyroid lesion in children typically includes nodular goiter, lymphocytic thyroiditis, colloid cysts, follicular adenomas, degenerating nodules, and malignant thyroid nodules.[23] However, other considerations for an intrathyroidal nodule, which occur less frequently, include ectopic thymic tissue, lymphomas, and even metastatic malignancies from other anatomic locations. Making the correct diagnosis through minimally invasive procedures, especially in instances of benign lesions, would prevent the performance of more invasive and complicated procedures, with their attendant consequences.

Thymus is a primary lymphoid organ that plays an important role in the differentiation of T-cells.[24] During embryogenesis, the thymus is formed from the ectoderm of the third branchial cleft and the endoderm of the third pharyngeal pouch.[22] The definitive thymus is formed by fusion of the right and left thymic primordial tissues before it descends to the upper anterior mediastinum.[22] Aberrant persistence of thymic tissue along migration may lead to ectopic thymus location, including intrathyroidal locus.[22] The prevalence of ectopic thymic tissue in children is very low and has been reported to range between 0.99%[25] and 1.8%.[26] Ectopic thymic tissue may manifest as a neck mass or an incidentally detected lesion.[27]

The two main differential diagnoses of ectopic thymic tissue (immature/maturing T-cells) include T-lymphoblastic leukemia (T-ALL) and thymoma. Since both the normal thymic tissue as well as the background lymphocytes in thymoma would have identical immunophenotypic profile, the distinction cannot be made solely based on immunophenotypic grounds, rather it is made based on morphological examination showing infiltrating “dispersed” epithelial cells, which is consistent with thymoma. Benign thymic maturing T-cells demonstrate a characteristic maturation expression pattern on flow cytometric immunophenotyping, which allows for reliable distinction from T-ALL.

The unique variable pattern of maturing T-cells mainly includes the following markers: CD3, CD1a, CD34, CD4, and CD8. The earliest maturing thymic T-cells are initially negative for CD3, CD1a, CD4, and CD8. Later, they acquire CD1a, CD4, and CD8, followed by expression of surface CD3. Finally, they will commit to either CD4 or CD8 and will lose CD1a with retention of surface CD3 (i.e., mature T cells). The normal progression of maturing, non-neoplastic thymocytes, is from CD4(-)/CD8(-), to CD4(dim+)/CD8(-), to CD4(+)/CD8(+), finally to a mature helper T-cell CD4(+)/CD8(-) or CD4(-)/CD8(+) cytotoxic T cells. Similarly, thymocytes progress from CD34(+)/CD1a (-)/CD3 (-), to CD34(-)/CD1a(+)/CD3(-), to CD34(-)/CD1a(+)/CD3(+), and eventually to mature T-cell immunophenotype CD1a(-)/CD3(+). In this case, the immunophenotypic analysis is consistent with a population of immature/maturing T-cells (CD3-/CD2+/CD5+/CD7+, with variably express CD4+, CD8+, and CD4+/CD8+ subsets, along with partial CD1a expression) which, in the given patient scenario, is most consistent with ectopic thymic tissue in the thyroid gland [Table 1].[28,29]

Rapid onsite evaluation (ROSE) is a critical component of FNA procedure by pathologists and cytotechnologists to increase the diagnostic yield of FNA procedures. Multiple studies have reported improved specimen adequacy with this technique[30-32] further emphasizing its unique role in patient care. In addition to improving diagnostic adequacy, ROSE also facilitates triaging of specimens that require ancillary diagnostic tests. In the current case, ROSE determined that the specimen was devoid of colloid and follicular cells and mainly consisted of singly scattered lymphocytes. A determination to triage a portion of the specimen for flow cytometric analysis was made during ROSE for evaluating lymphoproliferative processes. Flow cytometry was critical in making the right diagnosis and confirmed the nonneoplastic nature of the lesion, consistent with aberrant ectopic intrathyroidal thymic tissue with additional help from cell block.[33] FNA procedure without ROSE assessment may put the patient at risk for a more invasive procedure, especially if the cellblock contains insufficient diagnostic material. Therefore, the role of ROSE in evaluating and triaging cytopathology specimens cannot be overemphasized since it enhances diagnostic yield. One particular case illustrated here highlights ROSE’s critical role during the FNA procedure for effective patient care by submitting the needle rises for flow cytometry. The cases such as the one illustrated here highlight the critical role of ROSE during FNA procedure for effective patient care.

SUMMARY

Thyroid nodules are uncommon in the pediatric population and when they do occur, are more likely to be malignant and associated with local, regional, or distant metastasis.

Ectopic thymic tissue in the thyroid is a rare occurrence in children and may be mistaken for other B and T lymphocytic lesions on FNA procedures due to morphologically overlapping low-grade lymphoproliferative lesions. ROSE during an FNA procedure is critical because it can initiate appropriate triage of the specimen for ancillary testing and increase diagnostic accuracy. This would minimize the potential of invasive procedures and interventions.


References

Frates MC Benson CB Dorfman DM Cibas ES Huang SA Ectopic intrathyroidal thymic tissue mimicking thyroid nodules in children J Ultrasound Med 20183778391 10.1002/jum.14360 28850707
Li JH He HH Cheng Y He WJ Primary thyroid extranasal NK/T-cell lymphoma associated with good outcome: A case report and literature review: A care-compliant article Medicine (Baltimore) 201695e3460 10.1097/MD.0000000000003460 27196451
Karvounis E Kappas I Angelousi A Makris GM Kassi E Mucosa-associated lymphoid tissue lymphoma of the thyroid gland: A systematic review of the literature Eur Thyroid J 20209118 10.1159/000502204 32071897
Chandanwale SS Gore CR Bamanikar SA Gupta N Gupta K Cytomorphologic spectrum of Hashimoto's thyroiditis and its clinical correlation: A retrospective study of 52 patients CytoJournal 2014119 10.4103/1742-6413.131741 24987440
Shoyele O Bacus B Haddad L Li Y Shidham V Lymphoproliferative process with reactive follicular cells in thyroid fine-needle aspiration: A few simple but important diagnostic pearls CytoJournal 20191620 10.4103/cytojournal.cytojournal_5_19 31741667
Kuper-Hommel MJ Snijder S Jansen-Heijnen ML Vreugdenhil A Noordijk EM Kluin-Nelemans HC et al Treatment and survival of patients with thyroid lymphoma: A population-based study with clinical and pathologic reviews Clin Lymphoma Myeloma 200562407 10.3816/CLM.2005.n.052 16354330
Jeon EJ Shon HS Jung ED Primary mucosa-associated lymphoid tissue lymphoma of thyroid with the serial ultrasound findings Case Rep Endocrinol 201620165608518 10.1155/2016/5608518 27099797
Staerkel GA Bishop JA Shidham VB Zarka MA Undifferentiated (Anaplastic) Carcinoma and Squamous Cell Carcinoma of the Thyroid Cibas E Ali SZ The Bethesda System for Reporting Thyroid Cytopathology: Definitions, Criteria, and Explanatory Notes 2nd ed Ch. 11 United States Based on NCI Thyroid Fine Needle Aspiration State of the Science Conference 2009
Ibrahim MI Jusoh YR Adam NN Mohamad I Primary squamous cell carcinoma of the thyroid gland Iran J Otorhinolaryngol 201830658
Rosen RD Bordoni B Embryology, parathyroid StatPearls Treasure Island, FL StatPearls Publishing 2021 Available from: https://www.ncbi.nlm.nih.gov/books/NBK554580 [Last accessed on 2020 May 24]
Rosen RD Sapra A Embryology, thyroid StatPearls Treasure Island, FL StatPearls Publishing 2021 Available from: https://www.ncbi.nlm.nih.gov/books/NBK551611 [Last accessed on 2020 Jul 10]
Gordon J Manley NR Mechanisms of thymus organogenesis and morphogenesis Development 2011138386578 10.1242/dev.059998 21862553
Marx A Chan JK Coindre JM Detterbeck F Girard N Harris NL et al The 2015 World Health Organization classification of tumors of the thymus: Continuity and changes J Thorac Oncol 201510138395 10.1097/JTO.0000000000000654 26295375
Tracy ET Roman SA Current management of pediatric thyroid disease and differentiated thyroid cancer Curr Opin Oncol 2016283742 10.1097/CCO.0000000000000250 26575691
Hernandez-Cassis C Poniecka A Vogel CK McKenzie JM A six-year-old boy with a suspicious thyroid nodule: Intrathyroidal thymic tissue Thyroid 20081837780 10.1089/thy.2007.0262 18341382
Francis GL Waguespack SG Bauer AJ Angelos P Benvenga S Cerutti JM et al Management guidelines for children with thyroid nodules and differentiated thyroid cancer Thyroid 20152571659 10.1089/thy.2014.0460 25900731
Chang YW Kang HM Lee EJ Long-term follow-up ultrasonographic findings of intrathyroidal thymus in children Korean J Radiol 202021124855 10.3348/kjr.2019.0973 32729268
Eng L Underland L Lam L Intrathyroidal thymic tissue in an adolescent with Graves' disease: Case report and review of current literature Case Rep Pediatr 201920198089714 10.1155/2019/8089714 31065395
Thyroid Imaging Reporting and Data System (TI-RADS) Available from: https://www.radiopaedia.org/articles/thyroid-imaging-reporting-and-data-system-ti-rads?lang=us [Last accessed on 2021 Jun 09]
TI-RADS Calculator Description of Features Available from: http://www.tiradscalculator.com/description-of-features [Last accessed 9-62021]
TIRADS 2017 Flow Chart Available from: http://www.tiradscalculator.com/wp-content/uploads/2017/05/TIRADS-2017-Flow-Chart.jpg
Stasiak M Adamczewski Z Stawerska R Krawczyk T Tomaszewska M Lewinski A Sonographic and elastographic features of extra-and intrathyroidal ectopic thymus mimicking malignancy: Differential diagnosis in children Front Endocrinol (Lausanne) 201910223 10.3389/fendo.2019.00223 31110490
Patel MN Komlos M Racadio JM Intrathyroidal thymic tissue mimicking a thyroid nodule in a 4-year-old child J Clin Ultrasound 20134131920 10.1002/jcu.21930 22531870
Bang MH Shin J Lee KS Kang MJ Intrathyroidal ectopic thymus in children: A benign lesion Medicine (Baltimore) 201897e0282 10.1097/MD.0000000000010282 29620644
Fukushima T Suzuki S Ohira T Shimura H Midorikawa S Ohtsuru A et al Thyroid examination unit of the radiation medical center for the fukushima health management survey, prevalence of ectopic intrathyroidal thymus in japan: The fukushima health management survey Thyroid 2015255347 10.1089/thy.2014.0367 25778711
Escobar FA Pantanowitz L Picarsic JL Craig FE Simons JP Viswanathan PA et al Cytomorphology and sonographic features of ectopic thymic tissue diagnosed in paediatric FNA biopsies Cytopathology 2018292416 10.1111/cyt.12529 29577488
Park SH Ryu CW Kim GY Shim KS Intrathyroidal thymic tissue mimicking a malignant thyroid nodule in a 4-year-old child Ultrasonography 201433713 10.14366/usg.13001 24936498
van Dongen JJ Lhermitte L Böttcher S Almeida J van der Velden VH Flores-Montero J et al EuroFlow Consortium (EU-FP6, LSHB-CT-2006-018708) EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes Leukemia 201226190875 10.1038/leu.2012.120 22552007
Kroft SH Role of flow cytometry in pediatric hematopathology Am J Clin Pathol 2004122S1932 10.1309/1GLC0VY081X8HY2V 15690640
Witt BL Schmidt RL Rapid onsite evaluation improves the adequacy of fine-needle aspiration for thyroid lesions: A systematic review and meta-analysis Thyroid 20132342835 10.1089/thy.2012.0211 23043247
Pastorello RG Destefani C Pinto PH Credidio CH Reis RX de A Rodrigues T et al The impact of rapid on-site evaluation on thyroid fine-needle aspiration biopsy: A 2-year cancer center institutional experience Cancer Cytopathol 201812684652 10.1002/cncy.22051 30317695
Shield PW Cosier J Ellerby G Gartrell M Papadimos D Rapid on-site evaluation of fine needle aspiration specimens by cytology scientists: A review of 3032 specimens Cytopathology 2014253229 10.1111/cyt.12157 24844295
Alrajjal A Choudhury M Yang J Gabali A Cell-blocks and hematolymphoid lesions CytoJournal 2021187 10.25259/Cytojournal_10_2021 34221096


A subcutaneous firm nodule on scrotal skin: Cytological considerations

A 30-year-old patient presented with a solitary painless subcutaneous 3 × 3 cm nodule in the ventral aspect of the scrotum [Figure 1a] since 2 years without prior history of sexually transmitted disease, trauma, or inflammation of the scrotum. Fine-needle aspiration of the nodule showed an amorphous debris. The smears stained with May-Grunwald Giemsa and Papanicolaou stain showed findings shown in Figure 1b and c.

Solitary subcutaneous nodule (a) Cytology smear showed amorphous debris (b: May-Grunwald Giemsa, ×400); c: Papanicolaou, ×400; d: Zoomed area from c).

Q1. What is the most likely diagnosis?

Calcified epidermal cyst

Necrotic debris

Scrotal calcinosis

Scrotal pilomatrixoma.

Answer:

Q1-C. Scrotal calcinosis.

The presence of the amorphous basophilic substance, that is, calcific deposits without any epithelial cells in fine-needle aspiration (FNAC) smears, favored diagnosis of calcinosis.[1-4] The lack of epithelial cells in the FNAC smears helped in ruling out the possibility of a calcified epidermal cyst.[1] The calcified debris is also reported in pilomatrixoma.[5] FNAC of pilomatrixoma may show the presence of anucleated squames and basaloid cells, which were absent in the present case.[5] A possibility of necrotic debris was ruled due to the lack of inflammatory background.

Q2: Which one generally is not considered as the cytological features of scrotal calcinosis?

Amorphous basophilic granular material

Presence of foreign body giant cells

Lymphocytes surrounding basophilic amorphous material

Presence of numerous epithelial cells.

The FNAC of scrotal calcinosis generally shows the presence of amorphous basophilic granular material.[1-4] This material may be surrounded by lymphocytes and foreign body giant cells.[4,5] The epithelial cells are usually absent in the cytological smears of scrotal calcinosis.[1-4]

Q3. Which special stain is used to demonstrate calcium?

Periodic acid–Schiff (PAS)

von Kossa

Perl’s Prussian blue

Rhodanine stain.

Calcium tissue deposits can be identified by the presence of von Kossa-positive black masses.[6] With the hematoxylin and eosin stain, calcium appears deep blue purple. PAS is a commonly used stain to detect polysaccharides such as glycogen and mucosubstances, which appears deep red (magenta). Perl’s Prussian blue stain is used to demonstrate iron deposits in tissues. Copper deposits are demonstrated by rhodanine stain, which gives a red to orange-red cytoplasmic granular positivity to elemental copper.

A definitive diagnosis of scrotal calcinosis can be made only on the basis of histology.[6] In the present case, the subcutaneous nodule was excised, and histopathological examination showed lobules of amorphous calcified areas in the dermis. The lobules were surrounded by fibrosis; however, no surrounding foreign body reaction was observed [Figure 2]. The calcium deposits may be surrounded by lymphocytes and foreign body giant cell reactions. In some reports, foreign body reaction surrounding the calcific deposits was not seen.[1]

Histopathology showed calcific deposits in desmis (H and E, ×100).

Q4: Which of the following is not implicated in the pathogenesis of scrotal calcinosis?

Dystrophic calcification of scrotal epithelial cyst

Degeneration of the dartos muscle

Metastatic calcification secondary to metabolic derangement

Unknown.

The pathogenesis of scrotal calcinosis is still unknown.[7] Various authors have put forth various hypotheses. The prominent among them is that scrotal calcinosis arises due to dystrophic calcification of the ruptured scrotal epithelial cysts.[6] Authors have argued that there is some quality to the scrotal skin that leads to rapid resolution of epidermoid cysts with calcification.[6] Another hypothesis proposes that scrotal calcinosis is a degenerative phenomenon involving the scrotal dartos muscle, leading to calcium deposit.[2,6] The argument favoring the scrotal calcinosis idiopathic nature is that the basophilic calcified material within the dermis may or may not show foreign body granulomatous reaction, and none show evidence of epithelial lining.[6] Metastatic calcification occurs secondary to metabolic derangements such as hypercalcemia or hyperphosphatemia.[1] In the present case, they were normal.

There was no foreign body giant reaction around the calcific deposits or epithelial lining with normal levels of serum calcium, phosphorus, and parathormone in the present case. These findings favored an idiopathic etiopathogenesis in the present case suggesting a diagnosis of idiopathic scrotal calcinosis (ISC).

BRIEF REVIEW OF THE TOPIC

ISC is rare and has a benign course.[1] This condition of uncertain cause typically begins in adolescence or early adulthood and occurs in the absence of abnormalities in calcium and phosphate metabolism.[1] It is characterized by slow-growing subcutaneous nodules.[8] The nodules vary in number and can be solitary or grouped.[8] The color of the nodule may vary from yellowish-white to no color change depending on the age of the nodule. These lesions are usually firm and asymptomatic, although itching, pain, and episodes of infection may occur.[8] ISC may be complicated with itching, heaviness, or chalky material discharge.[1,2]

Pathogenesis still remains unknown and continues to be debated.[7] The dystrophic calcification of scrotal epithelial cysts and degenerative changes of dartos muscles leading to change in the microenvironment causing deposition of calcium and phosphate are the suggested pathophysiological modes for the development of ISC.[6]

Clinical differential diagnosis includes other scrotal lesions such as calcified epidermal inclusion cyst, pilomatrixoma, steatocystoma, calcified parasitic cyst, ancient schwannoma, lipoma, fibroma, and cutaneous horn.[1,8] Cytologically calcified epidermal cyst is differentiated from ISC by the presence of epithelial cells in the epidermal cyst.[1] Pilomatrixoma is cytologically characterized by the presence of basaloid cells, calcium deposits, naked nuclei, shadow (“ghost”) cells, giant cells, and inflammatory background.[5] Steatocystoma shows acellular, granular debris, rare cholesterol crystals, and anucleate squames.[9] Ancient schwannoma, which shows secondary changes including calcification, is usually hypocellular consisting of clusters of spindle cells with elongated nuclei.[10] In the case of a calcified parasitic cyst, parts of parasitic remnants in smears usually provide the clue. However, diagnosis is usually confirmed on histopathological features.[6]

The role of FNAC in the diagnosis of ISC remains limited; however, it can be a helpful as a preliminary diagnostic tool for this rare disorder.[1,2] A diagnosis by FNAC may be comforting for both the treating surgeon and the patient.[1] Surgical excision is the treatment of choice; however, cosmetic disfigurement is the major limitation of the surgical excision.[8]

Answers to Q2 through Q4:

Q2-D. Presence of numerous epithelial cells.

Q3-B. von Kossa.

Q4-C. Metastatic calcification secondary to metabolic derangement.

SUMMARY

We have described an uncommon case of ISC in scrotal skin that can be reliably diagnosed on FNAC. ISC occurs in the absence of any calcium and phosphate metabolism abnormalities; however, the pathogenesis remains elusive. Therefore, surgical excision is the treatment of choice.


References

Chakrabarti I Sharma SR Idiopathic scrotal calcinosis: Is cytological diagnosis enough? Indian Dermatol Online J 20134589 10.4103/2229-5178.105490 23437426
Sherwani RK Varshney BK Maheshwari V Rahman K Khan MA Idiopathic calcinosis of scrotum: Cytological diagnosis of a case J Cytol 200825234 10.4103/0970-9371.40654
Shivkumar VB Gangane N Kishore S Sharma S Cytological features of idiopathic scrotal calcinosis Acta Cytol 2003471101
Sahoo N Sethy M Mishra P Patra S Purkait S Giri SK Idiopathic scrotal calcinosis diagnosed by fine needle aspiration cytology-report of a rare case Diagn Cytopathol 2017459224 10.1002/dc.23757 28603937
Nigam JS Singh S Fine-needle aspiration cytology of pilomatrixoma: A short series of three cases Cytojournal 20141130 10.4103/1742-6413.144689 25506386
Dubey S Sharma R Maheshwari V Scrotal calcinosis: Idiopathic or dystrophic? Dermatol Online J 2010165 10.5070/D34QV0S2PG 20178701
Tela UM Ibrahim MB Scrotal calcinosis: A case report and review of pathogenesis and surgical management Case Rep Urol 20122012475246 10.1155/2012/475246 22888460
Pompeo A Molina WR Pohlman GD Sehrt D Kim FJ Idiopathic scrotal calcinosis: A rare entity and a review of literature Can Urol Assoc J 20137E43941 10.5489/cuaj.1387 23826060
Oertel YC Scott DM Cytologic-pathologic correlations: Fine needle aspiration of three cases of steatocystoma multiplex Ann Diagn Pathol 1998231820 10.1016/S1092-9134(98)80025-7
Nair V Dey B Deshpande AH Nigam JS Cystic schwannoma of tongue masquerading as mucocele: A cytological pitfall J Cytol 2018351256 10.4103/JOC.JOC_50_17 29643663


A diagnostic challenge in a rare variant of invasive breast carcinoma – How far one can go

CASE REPORT

Painless upper outer quadrant lump in left breast (single, hard, mobile, non-tender, irregular, measuring 4 × 3 cm) without axillary lymphadenopathy and without family history. Fine needle aspiration (FNA) showed findings shown in Figure 1.

(a) Tight clusters and morules of tumor cells against a clean background (Pap stain; ×10). (b) Cell clusters in papillary configurations, with many dissociated tumor cells (Pap stain; ×20). (c) Papillary structures with well-defined outline and without true fibrovascular core (Pap stain; ×40). (d) Round to oval tumor cells with distinct cell margins, moderate amount of pale cytoplasm, eccentrically placed round to pleomorphic nuclei with coarse chromatin and inconspicuous nucleoli (Pap stain; ×40) (Breast lump, FNA direct smear).

Q1: What is the interpretation?

Papillary neoplasm of breast

Papillary carcinoma of the breast

Invasive micropapillary carcinoma of the breast

Metastasis of papillary carcinoma to the breast.

Answer: (c) Invasive micropapillary carcinoma of the breast (IMPC)

The reports mentioning histopathological features are available in the reviewed literature; however, reports explaining the cytology of IMPC are scarce. The commonly observed features are richly cellular smears displaying angulated, three-dimensional cohesive clusters of ductal epithelial cells, and numerous dissociated cells with intact cytoplasm in a clean background [Figure 1a and b].[1-4] These aggregates show papilloid configuration meaning thereby that they lack a true fibrovascular core and possess smooth round to angulated contours [Figure 1c]. The tumor cells show a moderate amount of pale cytoplasm, high nucleocytoplasmic ratio, irregular nuclear membrane, fine to coarse chromatin, and inconspicuous nucleoli [Figure 1d]. Occasional mitosis is noted. Bare bipolar nuclei, cyst macrophages, and necrotic material were not observed in the smears examined. These distinctive cytomorphological features favor a cytodiagnosis of ductal carcinoma with the possibility of micropapillary subtype. Now, the certainty with which specific diagnosis of IMPC could be offered prospectively on cytology which is less commonly observed; and in most times, it is a retrospective diagnosis after correlating cytohistologic features.[1] This is because there exists a a list of differentials which can come to the mind of a cytopathologist that includes ranging from benign to malignant papillary lesions. In malignant category, it can either be primary or metastatic papillary carcinoma in of breast.[2-4]

The distinction of benign and malignant papillary neoplasms of the breast is essential from a management point of view; however, the cytologic diagnostic criteria are not sufficient to arrive at a definitive diagnosis in papillary lesions of the breast and show significant error due to overlapping features. The presence of increased cellularity, cellular atypia, long slender papillae, discohesive tumor cells with marked nuclear atypia, and absence of bare bipolar nuclei favor a malignant papillary lesion over the benign

Invasive papillary carcinoma reveals ramifying papillae having true fibrovascular core lined by columnar cells with hyperchromatic and pleomorphic nuclei. The background also shows many cyst macrophages, presence of necrosis, and hemorrhage[2]

In contrast to this, IMPC displays cohesive and angulated cell clusters, tumor morules, and papilloid aggregates lacking a true fibrovascular core and marked cell dissociation. These cells are round to cuboidal with pale cytoplasm and centrally placed pleomorphic nuclei against a clean background[1,3,4]

The metastatic papillary carcinoma to breast also needs to be ruled out as the line of management is different. Detailed clinical workup for ovarian, endometrial, or thyroid papillary carcinoma could help to exclude the possibility of metastatic disease. In situations of dilemma, an immunohistochemical such as gross cystic disease fluid protein-15 or mammaglobin a sensitive and specific marker for further confirming the diagnosis of primary breast carcinoma.[2,3]

Question 2: IMPC is seen to be commonly associated with which subtype of invasive breast carcinoma?

Medullary carcinoma breast

Tubular carcinoma

Secretory carcinoma

Mucinous carcinoma

Answer: (d) Mucinous carcinoma.[5]

Question 3: The smears from IMPC can also show features such as

Psammoma bodies

Apocrine cytology

Focal mucin deposition

All of the above

Answer: (d) All of the above.[5]

Question 4: IMPC an uncommon variant shows aggressive behavior because of

Hematogenous spread

Lymphatic spread

Pagetoid spread

None of the above

Answer: (b) Lymphatic spread.

FURTHER WORKUP OF THE CASE

The patient subsequently underwent a modified radical mastectomy of left breast with ipsilateral axillary lymph node dissection. The specimen received measured 50 × 30 × 30 cm, and the cut surface revealed an irregular, firm, yellowish, and gritty tumor of size 5 × 4 × 3 cm situated in the upper and outer quadrant. A total of 15 lymph nodes were dissected from the left axilla. Multiple sections examined from mass revealed an infiltrating tumor composed of small hollow and morula-like clusters of malignant epithelial cells surrounded by distinctly clear space and separated by thin fibrous septa [Figure 2]. The papilloid clusters were deficient in the central fibrovascular core and at places revealed tubules with the central lumina. The tumor cells displayed reverse polarity or inside out phenomenon characteristic feature of IMPC seen on histology only [Figure 3].[5] The lining epithelial cells were cuboidal with scant pale cytoplasm and round nuclei oriented toward the luminal surface. Nuclear pleomorphism was evident and mitotic figures were <5/10 hpf. The DCIS component, vascular invasion, nipple involvement, and skin infiltration were not evident in the surrounding breast parenchyma. Histopathology exhibited a pure IMPC (Grade I) without any associated component of conventional invasive breast carcinoma (IBC) or mucinous carcinoma and with evidence of metastasis in six out of 15 axillary lymph nodes.

Tumor aggregates without fibrovascular core surrounded by large empty spaces and separated by thin septa (HP: H&E stain; ×5).

Tumor cells in nests and tubules with central lumina displaying reverse polarity and separated by thin fibrous septa. The cells reveal nuclear pleomorphism and coarse chromatin with inconspicuous nucleoli (HP: H&E stain; ×40).

Detailed further workup also excluded primary ovarian, endometrial, thyroid, and renal malignancy. The patient underwent post-operative radiotherapy and chemotherapy. There is no evidence of relapse to date.

BRIEF REVIEW OF THE TOPIC

Pure IMPC is a rare variant of IBC constituting 0.9–2%.[5] This tumor type has received much attention in recent times not only because of its characteristic pathologic appearance, high Ki-67 proliferation index, but also more frequent HER-2/Neu overexpression, high incidence of lymph node metastasis, and aggressive biological behavior.[5,6] Positron emission tomography scan shows a high maximum standardized uptake value, which is an apparent feature of IMPC due to its frequent lymphovascular invasion and lymph node metastasis hence might be helpful in the early detection of IMPC.[7] The carcinomas with a micropapillary component reflect aggressive behavior, with a 5-year survival rate significantly lower than IBC -No special type (IBCNST) of the corresponding grade.[8,9] IMPC occurs as a pure form or maybe a component of IBC-NST or in combination with mucinous carcinoma. In IBC-NST, EMA is expressed on the luminal surface of the cell membrane; however, in IMPC, it is expressed in stroma-facing surface indicating reverse polarity. Furthermore, the expression of basolateral adhesive protein (E-cadherin) is mutually exclusive with EMA substantiating reverse polarity. In vitro studies revealed that increased expression of EMA on cell membrane facing toward stroma and simultaneous reduced expression of adhesion molecules might be responsible for the detachment of the cells from the stroma. Gene expression profiling reveals that IMPC is mostly luminal type breast carcinomas.[5]

As a consequence of increasing awareness, timely diagnoses, and appropriate management of this rare variant; the 5-year overall survival of these patients has recently been improved and the treatment of choice is radical mastectomy with postoperative radiotherapy and chemotherapy.[9]

Histopathology shows characteristic micropapillae lying within clear spaces and separated by a thin fibrocollagenous stroma, thus exhibiting inside-out phenomenon a diagnostic hallmark of IMPC.[10] Fine-needle aspiration cytology being simple and easy, office procedure is utilized as a first-line investigation in an initial assessment of almost every palpable breast lump. Hence, it is imperative to identify prototype cytological features for early detection of this rare variant, as specific diagnosis mandates identifying a group of patients with a poor outcome. The consistent cytologic appearance of IMPC is unique to reveal high cellularity, angulated clusters, morules of tumor cells and papilloid aggregates lacking a fibrovascular core, and marked cell dissociation exhibiting high-grade nuclear features against a clean background. These appear to be the most reliable cytological features as quoted in earlier reports that should raise suspicion of IMPC.[3,4]

SUMMARY

To sum up the precise, cytodiagnosis of invasive IMPC can be offered with certainty if one carefully looks for the indicators such as angulated papilloid cell clusters with anatomical borders and lacking a fibrovascular core, tumor morules, and plenty of dissociated malignant cells in a clean background.


References

Huq GE Canberk S Oznur M Yildiz P Bahadir B Toglu KB Cytological features of pure micropapillary carcinoma of various organs: A report of eight cases Oncol Lett 2014870510 10.3892/ol.2014.2198 25013488
Tse GM Ma TK Lui PC Ng DC Yu AM Vong JS et al Fine needle aspiration cytology of papillary lesions of the breast: How accurate is the diagnosis? J Clin Pathol 2008619459 10.1136/jcp.2008.057489 18552172
Kelten EC Akbulut M Duzcan SE Diagnostic dilemma in cytologic features of micropapillary carcinoma of the breast. A report of 2 cases Acta Cytol 20095346336 10.1159/000325355 19697739
Kumarshinghe MP Fernando MS Sheriffdeen AH Weerasekera K Cytohistologic features of invasive micropapillary carcinoma in a young female Diagn Cytopathol 2000231968 10.1002/1097-0339(200009)23:3<196::AID-DC11>3.0.CO;2-G
Marchio C Horlings HM Vincent-Salomom A Invasive Micropapillary Carcinoma in WHO Classification of Tumours of the Breast 5th ed Lyon IARC 209112830
Wu Y Zhang N Yang Q The prognosis of invasive micropapillary carcinoma compared with invasive ductal carcinoma in the breast: A meta-analysis BMC Cancer 201717839 10.1186/s12885-017-3855-7 29228910
Yun SU Choi BB Shu KS Kim SM Seo YD Lee JS Imaging findings of invasive micropapillary carcinoma of the breast J Breast Cancer 2012155764 10.4048/jbc.2012.15.1.57 22493629
Yoon GY Cha JH Kim HH Shin HJ Chae EY Choi WJ Comparison of invasive micropapillary and invasive ductal carcinoma of the breast: A matched cohort study Acta Radiol 201960140513 10.1177/0284185119834689 30897929
Liu F Yang M Li Z Guo X Lin Y Lang R et al Invasive micropapillary mucinous carcinoma of the breast is associated with poor prognosis Breast Cancer Res Treat 201515144351 10.1007/s10549-015-3413-4 25953688
Uddin Z Idrees R Aftab K Kayani N Invasive micropapillary carcinoma of the breast: an under-recognized entity. A series of eight cases Breast J 20121826771 10.1111/j.1524-4741.2012.01232.x 22583197


Epithelioid cell granulomas in urine cytology smears: A diagnostic approach

CLINICAL HISTORY

A 35-year-old female patient presented with painful gross hematuria associated with clots and burning micturition for a duration of 1 month. She also reported urgency, incontinence, and nocturia. She had a history of lower segment cesarean section done 8 months previously. There was no history of fever or other associated comorbidity. The general examination was unremarkable. Figure 1 shows the cytological features of three consecutive urine samples in Giemsa- and Pap-stained smears.

(a,b) Dense acute inflammation along with histiocytic aggregates (Urine smear, Giemsa, a ×100; b, ×400), (c) Epithelioid cell granuloma with inflammatory cells (Papanicolaou stain, ×400), (d) Positive for Acid fast bacilli (AFB) (arrow) (Ziehl–Nielsen stain, ×1000 oil immersion).

QUESTION

What is your interpretation?

Tuberculosis of urinary tract

Urothelial changes with treatment effect

Malignancy

Cystitis cystica glandularis.

Answer

The correct cytopathological interpretation is (a) Urinary tract tuberculosis.

Explanation

Three consecutive urine samples showed the presence of epithelioid cell granulomas and multinucleated giant cells along with reactive urothelial cells and inflammatory cells [Figure 1a-c]. Special stain for acid-fast bacilli (AFB) was positive [Figure 1d], thereby confirming the diagnosis of tuberculosis. The reverse transcription polymerase chain reaction and cartridge-based nucleic acid amplification test of the urine sample were also positive. The patient was immediately started on antitubercular therapy, to which she responded well.

Ultrasonography for kidney, ureter, and urinary bladder revealed a mid-pole, well-defined, cortical hyperechoic space-occupying lesion (SOL) measuring 7×8×4 mm suggestive of hemangioma. A focal concretion and right-sided mild-to-moderate hydroureteronephrosis were present. Urinary bladder showed circumferentially thickened wall. Contrast-enhanced computed tomography revealed similar findings [Figure 2a and b].

(a) Ultrasonography KUB and (b) computed tomography showing evidence of hydroureteronephrosis.

ADDITIONAL QUIZ QUESTIONS

Q2. What is the most common route of infection in renal tuberculosis?

Ascending spread

Hematogenous

Lymphatic spread

Direct invasion.

Q3. In genitourinary TB, which one of the following is true ?

Sterile pyuria is a consistent finding

AFB in early morning sample is always positive.

Most common site is pelvis

It is the commonest cause of pyelonephritis.

Q4. Golf hole ureter is seen in:-

Ureteric calculus

Ureteral polyp

Tuberculosis of ureter

Retroperitoneal fibrosis.

Q5. Which of the following considered as reliable diagnostic modalities for urinary tract tuberculosis?

ZN staining and cultures isolation for Mtb in urine,

PCR for Mtb,

Imaging studies,

Histopathological evidence for TB

All of the above.

ANSWERS TO THE ADDITIONAL QUIZ QUESTIONS

Q2. b; Q3. a; Q4. c; Q5. e

Q2.b Genitourinary tuberculosis (GUTB) is mostly secondary to pulmonary infection. Renal TB is a chronic process that can start many years after the initial lung infection.

Q3. a Sterile pyuria is the rule. Tubercle bacilli can be identified on AFB staining of 24 h urine specimen or the first morning urine sample collected on 3 successive days. AFB staining is positive in about 60% of the cases. The most common site of GUTB is kidney. The most common cause of pyelonephritis is E. coli.

Q4. c Fibrosis due to Mycobacterium tuberculosis (Mtb) usually starts around the ureter and causes pull at the ureter leading to a strictured, dilated, and rigid mouthed ureter called as golf hole ureter.

Q5. e The most common procedures used for diagnosis of urinary tract tuberculosis include: (1) Urine cytology smears examination, (2) Ziehl–Neelsen (ZN) staining and cultures isolation for Mtb in urine, (3) PCR for Mtb, (4) imaging studies, and (4) histopathological evidence for TB.

DISCUSSION

GUTB is a term coined by Wildbolz in 1937.[1] GUTB is the second most common form of extrapulmonary tuberculosis after lymph node involvement and it usually affects adults between the second and fourth decades of life.[1] Mtb bacilli are shed into the urine; they spread into the urinary tract, involving the renal pelvis, ureters, and bladder; the urinary tract mucosa may be ulcerated, thin, and without contractility.[2] In most patients, acquired cellular immunity develops which inhibits multiplication of bacilli and contains the disease by forming microscopic granulomas, leading caseous necrosis with local tissue destruction.[2] In addition, the tubercular granulomas are usually associated with Langhans’ type multinucleated giant cells.

Urinary tract tuberculosis diagnosis is a challenge due to the insidious onset of UTTB with few non-specific symptoms and atypical presentations, technical difficulties to isolate Mtb, and the long time required to confirm the diagnosis by culture, lack of awareness of physicians, and poor care seeking behavior which lead to difficulty and delay the diagnosis.[3]

The differential diagnosis of presence of granuloma in urine cytology includes tubercular, fungal, or protozoal infections, foreign body reactions after instrumentation, and bacillus of Calmette-Guerin (BCG)-induced cystitis.[4]

Presence of epithelioid cell granuloma in urine specimen is a rare finding on urine cytology smears. Kapila and Verma described dispersed or loose clusters of epithelioid histiocytes that often have spindle or carrot shaped nuclei in urinary tuberculosis cases. It is necessary to differentiate between a case of tuberculosis and granulomatous reaction as a result of BCG therapy.[5] In our case, there was no history of instrumentation or BCG therapy. There was no fungal or parasitic profile in the routine smears.

Multinucleated giant cells can display similar morphological features as umbrella cells, so must be differentiated. Umbrella cells are commonly seen in spontaneously voided urine of patients with renal colic, pronounced distention of the bladder, viral infections, previous radiation, or topical chemotherapy for superficial bladder cancers.[6] Diamond shape of the cells, cytoplasmic vacuolation, and numerous nuclei (50 or more) usually indicate that the cells are of epithelial origin rather than histiocytic.[7]

The presence of AFB in ZN stain or positive urine culture indicates a definitive diagnosis of urinary tuberculosis, however, the same is not excluded based on negativity of ZN staining.

A few studies have reported AFB in urine cytologic specimen previously. In one large series, the presence of bacilluria has been demonstrated in 5.2% of cases. In other studies, AFB positivity by ZN stain on urine smear has been reported variably from 25 to 42%.[8] Urinary PCR is considered highly sensitive as it can detect 94.29% of cases. It is especially useful when AFB smears are false negative still suspected of having tuberculosis.[9,10]

SUMMARY

The presence of epithelioid cell granulomas in urine along with Langhans’ type giant cells is highly suggestive of tuberculosis. The presence of AFB on ZN stain on the urine smears confirms the diagnosis, thereby obviating the need for invasive techniques such as cystoscopy and biopsy and allows immediate initiation of antitubercular therapy. Hence, AFB staining in urine smears is mandatory in all cases where tuberculosis is suspected clinically or cytomorphologically.

Tuberculosis continues to be a worldwide disease with predilection for immunocompromised patients and the lower socioeconomic populations. The kidney remains the primary target for disseminated disease. Untreated urinary tuberculosis can cause severe urinary symptoms, renal failure, and death. Effective treatment is dependent on awareness, early recognition, and prompt treatment.

COMPETING INTEREST STATEMENT BY ALL AUTHORS

The authors declare that they have no competing interest.

AUTHORSHIP STATEMENT BY ALL AUTHORS

Each author has participated sufficiently in the work and takes public responsibility for appropriate portions of the content of this article.

All authors read and approved the final manuscript.

Each author acknowledges that this final version was read and approved.

ETHICS STATEMENT BY ALL AUTHORS

As this is case without identifiers, our institution does not require approval from Institutional Review Board (or its equivalent).

LIST OF ABBREVIATIONS (In alphabetic order)

AFB – Acid-fast bacilli

BCG – Bacillus of Calmette-Guerin

CBNAAT – Cartridge-based nucleic acid amplification test

CECT – Contrast-enhanced computed tomography

GUTB – Genitourinary tuberculosis

Mtb – Mycobacterium tuberculosis

RT PCR – Reverse transcription polymerase chain reaction

SOL – Space-occupying lesion

USG KUB – Ultrasonography for kidney, ureter, and urinary bladder

ZN – Ziehl–Neelsen.

EDITORIAL/PEER-REVIEW STATEMENT

To ensure the integrity and highest quality of CytoJournal publications, the review process of this manuscript was conducted under a double-blind model (authors are blinded for reviewers and vice versa) through automatic online system.


References

Gupta NP Genitourinary tuberculosis Indian J Urol 200824355 10.4103/0970-1591.42617 19468468
Rieder HL Snider DE Jr Cauthen GM Extrapulmonary tuberculosis in the United States Am Rev Respir Dis 199014134751 10.1164/ajrccm/141.2.347 2301852
Wise GJ Urinary tuberculosis: Modern issues Curr Urol Rep 2009103138 10.1007/s11934-009-0051-6 19570494
Peto HM Pratt RH Harrington TA LoBue PA Lori R Armstrong LR Epidemiology of extrapulmonary tuberculosis in the United States, 1993-2006 Clin Infect Dis 20094913507 10.1086/605559 19793000
Ghaleb K Afifi M Gohary ME Assessment of diagnostic techniques of urinary tuberculosis Mediterr J Hematol Infect Dis 20135e2013034 10.4084/mjhid.2013.034 23795272
Kapila K Verma K Cytologic detection of tuberculosis of the urinary bladder Acta Cytol 198428901
Figueiredo AA Lucon AM Urogenital tuberculosis: Update and review of 8961 cases from the world literature Rev Urol 20081020717
Arora SK Gupta N Nijhawan R Mandal AK Epithelioid cell granulomas in urine cytology smears: Same cause, different implications Diagn Cytopathol 20103876567 10.1002/dc.21327 20187110
Adhya AK Dey P Cytologic detection of urinary tract tuberculosis Acta Cytol 2010546534 10.1159/000325195 20715673
Altintepe L Tonbul HZ Ozbey I Guney I Odabas AR Cetinkaya R et al Urinary tuberculosis: Ten years' experience Ren Fail 20052765761 10.1080/08860220500234857 16350814


Pancreatic cyst endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA): Benign or malignant. Clues to cytological diagnosis with major consequences

EUS-FNA of a 2.3 cm cystic lesion in tail of pancreas of 50 year old American woman showed cytopathologic findings seen in Figure 1. The Cyst fluid amylase was high (>20,000 units/L). Carcinoembryonic Antigen (CEA) was not raised.

Relatively hypocellular aspirates showed poorly preserved cells with myxoid debris and focal yellow refractile pigment (in both Pap and Diff Quik stained smears) without epithelial cells, except scant gastric mucosal contamination. [a (x 20)- Pap stain; b (x 20), c and d (x 100)- Diff-Quik stain].

QUESTION # 1

What is your interpretation?

Mucinous cystic lesion (mucinous cystic neoplasm [MCN]/intraductal papillary mucinous neoplasm [IPMN])

Pseudocyst

Carcinoma with cystic and necrotic changes

Cystic neuroendocrine tumor.

ANSWER

b. Pseudocyst.

The differential diagnosis of pancreatic cyst includes neoplastic and non-neoplastic categories. Although the majority of pancreatic cysts are benign, the survival after invasive carcinomas in potentially malignant mucinous cystic lesions either MCN or IPMN is often dismal. Distinction between a true cyst versus a pseudocyst can be challenging. The crucial role of cytology is to properly triage such patients.

EUS-FNA in the current case was relatively hypocellular with abundant thick myxoid material in the background without unequivocal epithelial cells (cyst lining cells). The polyhedral cells showed vacuolated/foamy cytoplasm with small, occasional folded nuclei consistent with reactive foamy histiocytes. Many spindle cells with repair-like pattern were also noted [Figure 2]. Cyst debris with occasional non-specific crystalline yellow pigment was noted in the background. The yellow pigment retained its yellow crystalline feature in Diff-Quik smear also. The cell-block (CB) showed stroma with reactive pancreatic ducts with mild chronic inflammation without pancreatic acini and malignancy [Figure 3]. The findings were consistent with pseudocyst in the background of chronic pancreatitis.

Cell-block of transgastric EUS-FNA cytology of cystic lesion in tail of pancreas. The sections showed reactive pancreatic ducts in ill-defined lobular architecture with sclerotic stroma without pancreatic acini. (H and E stained cell-block sections; (a) ×10, (b) ×20, (c) ×40, (d) magnification of cropped area).

Transgastric EUS-FNA cytology of cystic lesion in the tail of pancreas. Focal collection of polyhedral stromal histiocytes should not be confused with epithelial cells with myxoid debris in the background. This may lead to misinterpretation as mucinous cystic lesion (MCN or IPMN). (Diff-Quik stained direct smear, ×40).

The patient had medical history of alcohol abuse, pancreatitis, and ovarian borderline mucinous neoplasm status post-resection presented with upper abdominal pain. Based on the history of borderline mucinous neoplasm of the ovary, the differential interpretation included MCN and IPMN.[1,2] However, the absence of epithelial lining and lack of thick viscid mucin ruled out MCN or IPMN.

Cystic neuroendocrine tumor would have shown singly scattered plasmacytoid neuroendocrine cells with focal cohesive pattern without significant proportion of myxoid material which was not seen in this case.

QUESTION # 2

Which are the diagnostic features of pancreatic pseudocyst?

Chemical analysis of cyst fluid with high amylase

Absence of epithelial cells with repair-like stromal cells

Foam cells and cyst debris with myxoid background

Presence of yellow crystalline pigment

All of the above.

The proper diagnosis of a pancreatic cyst should be based on the combination of pre-procedural findings (clinical and imaging) in conjunction with post-procedural findings (chemical analysis of cyst fluid and cytologic evaluation). The ultimate goal of such diagnostic tests is to distinguish benign cyst with malignant potential (IPMN and MCN) and to evaluate the morphological features of malignancy. The helpful features include the presence or absence of yellow pigment, the proportion and nature of background myxoid contents, and the presence or lack of epithelial lining. The cytologic findings that distinguish pseudocyst from neoplastic cyst with malignant potential are summarized in Table 1. In addition to the cytologic features, cyst fluid analysis along with the clinical presentation and imaging is critical to make correct interpretation. Cytopathology of mucinous cystic lesions also allows evaluation and follow-up for neoplastic progression towards high grade dysplasia and invasive carcinoma.

Comparison of cytological features of pancreatic pseudocyst and neoplastic mucinous cysts (IPMN, MCN).

Cytological features Pseudocyst Neoplastic mucinous cyst (IPMN, MCN)
Epithelial lining cells (possibility of GI mucosal contamination especially gastric foveolar mucosa may be a pitfall) Absent Present in benign, borderline or malignant
Proportion (quantity) of/nature (quality) of mucin in the background Usually absent or low amount (however, myxoid background even with positive histochemical mucin staining such as Alcian blue or Mucicarmine may be a pitfall)[3] Usually abundant as viscid mucin
Extracellular pigmented material Amorphous and crystalline yellow pigment as surrogate marker in a number of cases Consistently absent
Presence of cyst debris in the background Foam cells and fat necrosis debris often present with spindle cells and stromal fragments with repair-like pattern Foam cells can be present

Myxoid background in pancreatic pseudocysts may be difficult to distinguish from viscid mucin in mucinous cystic lesions [Figure 4]. In our case, it showed focal myxoid areas admixed with spindle cells and stromal fragments with refractile yellow pigment. However, the aspirates lacked atypical epithelial cells. There were many histiocytes including foam cells with neutrophils.

Transgastric endoscopic EUS-FNA cytology of cystic lesion in the tail of pancreas. The direct smears showed spindle cells (a and b) and some polyhedral cells (c) with foam cells (d) in the background with myxoid debris (Diff-Quik stain; (a and b) ×20; (c and d) ×40).

QUESTION # 3

What clinical/imaging scenario is typical for pseudocyst compared to neoplastic cyst?

New, non-specific GI complaints with multilocular complex cyst in pancreatic head

New, non-specific GI complaints with cystically dilated pancreatic duct

History of recurrent pancreatitis with unilocular simple cyst

History of pancreatic neuroendocrine tumor with a newly identified partially cystic mass.

Pancreatic pseudocyst results from reparative changes secondary to the pancreatic parenchymal injury. Typically, pseudocysts are the result of multiple episodes of acute and chronic pancreatitis. The pseudocyst contents are rich in amylase and/or lipase and lack epithelial lining. History of recurrent pancreatitis with unilocular simple cyst is the classic clinical scenario to trigger the clinical suspicion of a pancreatic pseudocyst. MCN and IPMN, on the other hand, usually present with a multilocular cystic lesion. IPMN communicates with pancreatic ductal system and usually causes pancreatic duct obstruction with dilated duct on imaging (endoscopic retrograde cholangiopancreatography [ERCP] or magnetic resonance cholangiopancreatography [MRCP]). Thus, obstructive jaundice is the usual initial presentation of IPMN.

Serous cystic neoplasm composed numerous small cysts in a honeycomb-like formation of locules ranging from 1 to 20 mm size are typically seen on CT scan as a multicystic, lobulated lesion described as “bunch of grapes” with tendency for central scar and calcification. There is usually lack of the history of pancreatitis. Some pancreatic primary neoplasms can be associated with cystic degeneration, including neuroendocrine tumors of the pancreas. However, the presence of a previous neoplasm makes the possibility of a pseudocyst less likely.

QUESTION # 4

What chemical markers are most helpful in distinguishing pseudocyst from neoplastic mucinous cyst?

CEA and amylase

Amylase and lipase

CA 19-9 and CA-125

Glucose.

Chemical analysis can be an useful ancillary test to support cytopathologic evaluation, The panel of chemical tests to be performed on cyst fluid should be directed based on the clinical suspicion and the amount of fluid aspirated. In general, the most useful markers to distinguish pseudocyst versus neoplastic mucinous cyst are CEA and Amylase. A pseudocyst is, by definition, a collection of amylase-rich fluid. CEA, on the other hand, is a marker of glandular epithelial cells. As a result, a typical pseudocyst would show increased amylase and low level of CEA. A reversed result is expected in neoplastic mucinous cysts. The combination of amylase and lipase is not helpful as both markers are expected to be elevated in most pseudocysts but can also be elevated in neoplastic a number of neoplastic cysts as well. Tumor markers CA 19-9 and CA-125 are typically elevated in neoplastic cysts while they are not elevated in most pseudocysts thus are not helpful in distinguishing between the two.

BRIEF REVIEW OF THE TOPIC Role of cytology

Pancreatic cystic lesions encompass a wide range of neoplastic and non-neoplastic entities. The recent advances in imaging technologies allowed for an increased identification of pancreatic cysts with up to 19% prevalence in some reports.[4] It is crucial to discern non-neoplastic cysts from neoplastic lesions because the latter though considered benign, have malignant potential. International Consensus Algorithm for the management of mucinous cysts proposed management algorithm recommends that patients with cysts <1 cm should be re-evaluated after 1 year with MRI or thin-slice CT studies and should undergo resection if the cyst increases in size.[5,6]

About 80% of pancreatic cystic lesions identified on imaging are non-neoplastic, including pseudocysts.[7] Pancreatic pseudocyst is an encapsulated collection of homogenous amylase-rich fluid with little or without necrotic debris within it. Often, it is well-circumscribed and located outside of the pancreas, usually in the lesser sac. However, pseudocysts have been located in many areas in and around the pancreas.[2] Pancreatic pseudocysts form as a complication of chronic pancreatitis and less commonly from acute pancreatitis. They are found in 0.5–1/100,000 adults per year with significant association with alcohol consumption.

Pancreatic cancer is arguably the deadliest cancer. Because of the malignant potential of neoplastic pancreatic cysts, familiarity of the cytomorphologic features of pancreatic pseudocysts are of momentous importance for practicing cytopathologists to make an accurate diagnosis that would affect the trajectory of the patient care. A comprehensive multimodal approach including a combination of clinical, imaging, chemical, cytomorphologic, and molecular findings needs to be applied to reach the final diagnosis.[8]

The efficacy of EUS-FNA of cystic lesions of pancreas partly depends on the site, size, and characteristics of the target tissue as well as on the expertise, training, and interaction between the endosonographer and the cytopathologist.[9] Acquisition of diagnostic samples is approached in different ways depending on the site and type of the lesion.[10] Cytological samples are ideal for cyst fluid analysis, immunohistochemistry, and molecular testing. Direct smears are invaluable, both for immediate assessment but also for the final morphological diagnosis. The needle rinses can be processed as liquid-based cytology or CB. CB increases the diagnostic accuracy of EUS-FNA.[11] It facilitates immunocytochemistry. A mixture of inflammatory cells, islet cells, and fragments of fibrous stroma is usually present in the aspirate from patients with acute pancreatitis. Cytology helps in distinguishing cyst types and also allows evaluation for dysplasia and invasive carcinoma in mucinous cystic lesions with the higher malignant potential. Cytology can be helpful in finding atypical epithelial cell clusters with drunken honey-comb pattern that has high nuclear-tocytoplasmic ratio, nuclear membrane irregularity, nuclear size, nuclear crowding, and hyperchromasia to clinch a diagnosis of malignancy.

Pseudocyst usually contains inflammatory cells, for example, histiocytes, neutrophils, or both on myxoid background focally in Diff-Quik stained preparation. On a Papanicolaou stain, abundant extracellular mucin with epithelial cells is a finding that strongly suggests a diagnosis of neoplastic mucinous cyst. However, gastrointestinal mucosal contamination (both epithelial cells and mucin) is very common and could be a significant diagnostic pitfall leading to atypical interpretations.[12] Pseudocyst shows yellow crystalline material in both Pap stained and DiffQuik stained preparations. This pigment is not hemosiderin (which stains blue-gray in Diff-Quik stain). The features suggest that it may be related to hematoidin which also retain its yellow crystalline nature in Diff-Quik stained smear.[13]

It is important to consider preparation of CBs from all FNA specimens whenever possible. Specimens that have tissue material and/or blood are suitable for CBs following smear preparation. Recently described method, with ready to use kits allows quantitatively and qualitatively optimum CBs from most of the cytology specimens including EUS-FNA aspirates of pancreatic cystic lesions.[14,15] In the present case, CB was performed on the cyst fluid tissue material. Sclerotic stroma with reactive pancreatic ducts was noted. The duct lumens were irregular and lined by epithelial lining with polymorphic cells with dense nuclear chromatin. Spindle cells and foamy histiocytes are present with a repair like pattern.

Not all cysts could be characterized by FNA and cytopathology alone. An elevated CEA level (>192 ng/mL) and v-Ki-ras2 mutation in the aspirate are confirmatory of a mucinous cyst. Overexpression of DNA oncogenes (e.g., GNAS, KRAS) and/ or loss of heterozygosity of tumor suppressor genes (e.g., tumor protein [p53], cyclin-dependent kinase inhibitor 2A [p16], ring finger protein 43) detected in cyst fluid also aid to the diagnosis and determination of malignant potential.[4]

SUMMARY

The primary role of cytology is the exclusion of cyst with malignant potential (IPMN and MCN) or malignant cystic lesion. Pseudocysts usually show cytologic features which are frequently nonspecific and are primarily interpreted in correlation with clinical (alcoholism and pancreatitis) and imaging (gland atrophy with calcification and a unilocular cyst without a mural nodule) features with chemical analysis of cyst fluid.

Yellow pigment which continues to be yellow crystalline in Diff-Quik stained smears is important clue as surrogate marker of a pseudocyst. Myxoid background focally should not be confused for viscid and abundant mucin in cysts with malignant potential (IPMN and MCN).

Answers for Question 2 through 4:

2. e

3. c

4. a.


References

Lowery M Klimstra D Hedvat C Jarnagin W Chi D O'Reilly EM A case of synchronous pancreatic ductal adenocarcinoma and ovarian mucinous cystic neoplasm: Use of kras mutation molecular phenotyping to demonstrate independent primary origin Gastrointest Cancer Res 201256770
Gagne LJ Colacchio T Longnecker DS Simultaneous mucinous cystadenoma of ovary and mucinous cystadenocarcinoma of pancreas Int J Pancreatol 200028913 10.1385/IJGC:28:1:09
Gonzalez Obeso E Murphy E Brugge W Deshpande V Pseudocyst of the pancreas: The role of cytology and special stains for mucin Cancer 20091171017 10.1002/cncy.20000 19365837
Yoon WJ Brugge WR Pancreatic cystic neoplasms: Diagnosis and management Gastroenterol Clin 20124110318 10.1016/j.gtc.2011.12.016 22341252
Shen J Brugge WR DiMaio CJ Pitman MB Molecular analysis of pancreatic cyst fluid Cancer Cytopathol 200911721727 10.1002/cncy.20027 19415731
Brugge WR Diagnosis and management of cystic lesions of the pancreas J Gastrointest Oncol 2015637588
Misra D Sood T Pancreatic pseudocyst Stat Pearls Treasure Island, FL Stat Pearls Publishing 2020
Bellizzi AM Stelow EB Pancreatic cytopathology: A practical approach and review Arch Pathol Lab Med 2009133388404 10.5858/133.3.388 19260745
Ieni A Barresi V Todaro P Caruso RA Tuccari G Cell-block procedure in endoscopic ultrasound-guided-fine-needle-aspiration of gastrointestinal solid neoplastic lesions World J Gastrointest Endosc 20157101422 10.4253/wjge.v7.i11.1014 26322154
Jenssen C Möller K Wagner S Sarbia M Endoscopic ultrasound-guided biopsy: Diagnostic yield, pitfalls, quality management Z Gastroenterol 200846897908 10.1055/s-2008-1027419 18810676
Ieni A Todaro P Crino SF Barresi V Tuccari G Endoscopic ultrasound-guided fine-needle aspiration cytology in pancreaticobiliary carcinomas: Diagnostic efficacy of cell-block immunocytochemistry Hepatobiliary Pancreat Dis Int 20151430512 10.1016/S1499-3872(15)60367-8
Obeso EG Murphy E Brugge W Deshpande V Pseudocyst of the pancreas Cancer Cytopathol 20091171017 10.1002/cncy.20000 19365837
Scooby V Macrophage with Hematoidin Crystal Available from: https://www.youtube.com/watch?v=p5rmty-n5ui [Last accessed on 2021 May 21]
Shidham VB CellBlockistry: Chemistry and art of cell-block making-a detailed review of various historical options with recent advances Cytojournal 20191612 10.4103/cytojournal.cytojournal_20_19 31367220
AV BioInnovation, Processing of a Single Specimen of any Cellularity to Make a Cell Block with Nano Unit Available from: https://www.youtube.com/watch?v=y29ss1nwo_8&feature=youtu.be [Last accessed on 2021 May 21]


Plasmacytoid cells in a thyroid aspirate – Look before you leap

CASE HISTORY

A 60-year-old female with hypothyroidism under treatment presented with neck swelling for 2 months. Ultrasonography showed bulky right thyroid lobe (6.4 × 4.2 × 3.1 cm) with multiple heterogeneous nodules and microcalcification and with multiple necrotic cervical lymph nodes. Fine-needle aspiration (FNA) of the largest thyroid nodule (3 × 3 × 2 cm) is shown in Figure 1.

(a) Right thyroid FNA. Cellular smear showing predominantly discohesive plasmacytoid cells of variable sizes (Giemsa stain, ×400). (b) Few binucleate and multinucleate cells present. Background shows a few scattered lymphoglandular bodies (Giemsa stain, ×600). (c) Cells have eccentric nuclei with finely stippled chromatin and moderate amount of cytoplasm (Papanicolaou stain, ×400). (d) Discohesive plasmacytoid cells. (Giemsa stain, ×1000).

What is your interpretation?

Hurthle cell neoplasm

Non-Hodgkin’s lymphoma

Medullary carcinoma

Metastatic carcinoma

Answer

The correct answer is b. Non-Hodgkin’s lymphoma.

Thyroid FNA smears were highly cellular, showing predominantly discohesive plasmacytoid cells of variable sizes [Figure 1a]. Also seen were binucleate and multinucleate cells [Figure 1b]. The cells had eccentric nuclei with finely stippled chromatin and moderate amount of cytoplasm [Figure 1c]. Lymph node aspirate also showed similar cytomorpholological features. The location, that is, mass in the thyroid, along with the above cytologic features led to a high suspicion of diagnosis of medullary thyroid carcinoma (MTC, plasmacytoid type) with lymph node metastasis. Serum calcitonin was advised. The calcitonin level was found to be less than 2 pg/ml. On review of the slides, it was observed that the background had scattered lymphoglandular bodies (LGBs). One Pap stained slide was then destained for immunocytochemistry (ICC) and the cells showed strong immunoreactivity for leukocyte common antigen (LCA) (CD45) [Figure 2]. The possibility of non-Hodgkin lymphoma (NHL) with plasmacytic differentiation was highly considered. In the meanwhile, a lymph node biopsy was also done for further workup. The cells showed immunoreactivity for LCA, cluster designation (CD)79a, CD10, BCL2, and EMA and non-immunoreactivity for calcitonin, CD138, CD56, CD3, CD20, CD30, CD23, and anaplastic lymphoma kinase (ALK1). The proliferative index (MIB1) was 40%. The final diagnosis of high-grade non-Hodgkin B-cell lymphoma with plasmacytic differentiation was rendered.

Cells expressing LCA immunoreactivity (ICC, ×400).

ADDITIONAL QUIZ QUESTIONS

Q1. Which immunochemistry panel will best help in the diagnosis?

Calcitonin, synaptophysin, chromogranin

Calcitonin, LCA, CD138

LCA, synaptophysin, CK

Thyroglobulin, chromogranin, calcitonin.

Q2. All the following are features of medullary thyroid carcinoma except

Dispersed tumor cells

Eccentric nuclei

Stippled chromatin

Blue cytoplasmic granules on MGG.

Q3. Lymphoglandular bodies are derived from:

Nuclear remnants

Stain precipitation

Cytoplasmic fragments

Immunoglobulin.

Answers of additional Quiz Questions

Q1.b; Q2.d; Q3.c.

BRIEF REPORT

Clinically, a mass in the thyroid and the presence of discohesive plasmacytoid cells in a background containing amyloid/amyloid-like material on cytology smears leads to a diagnosis MTC as it is the most common carcinoma associated with the above two features among thyroid malignancies.[1] These features can, however, also be seen in other rare thyroid neoplasms such as plasma cell neoplasm and NHLs with plasma cell differentiation.[2] It is important to look for other supporting cytomorphological features that are often reported in MTC, like red cytoplasmic granules (88%), neuroendocrine type salt and pepper or stippled nuclear chromatin (65%), binucleated and multinucleated cells (70%), cytoplasmic “triangular” tails (58%), and amyloid.[3] Amyloid should be confirmed by Congo-red stain and serum calcitonin levels must be advised.

The presence of LGBs, cartwheel chromatin pattern, and Dutcher bodies can indicate malignant lymphoma and plasmacytoma.[4] The monomorphic appearance, granular cytoplasm, intranuclear inclusions, and eccentric nucleus of MTC cells can mimic Hürthle cell neoplasm and Hurthle cell variant of papillary thyroid carcinoma (PTC).[5] The finely textured chromatin, prominent macronucleoli, and blue cytoplasmic granules on Romanowsky stains correctly identify Hürthle cell neoplasm.[5] The presence of characteristic nuclear features of PTC, dense cytoplasm, and positive immunoreactivity for thyroglobulin distinguishes it from MTC. Rarely metastatic tumor, particularly melanoma warrants consideration but the presence of pigment and macronucleoli helps in the diagnosis. Serum calcitonin and immunochemistry for calcitonin, thyroglobulin, LCA, CD 138, and HMB 45 are of great help for confirmation and in avoiding the diagnostic dilemmas.

SUMMARY

The diagnosis of lymphomas with plasmacytic morphology in the thyroid can be overlooked and misinterpreted as MTC due to rarity and overlapping cytomorphologic features. Clinical correlation, precise study of the cytological features, and carefully screening the thyroid smears for LGBs in the background are crucial to avoid misdiagnosis, before instituting a definitive therapy.


References

Zeppa P Vetrani A Marino M Fulciniti F Boschi R De Rosa G et al Fine needle aspiration cytology of medullary thyroid carcinoma: A review of 18 cases Cytopathology 199013544 10.1111/j.1365-2303.1990.tb00324.x 2130998
Al-Marzooq YM Chopra R Younis M Al-Mulhim AS AlMommatten MI Al-Omran SH Thyroid low-grade B-cell lymphoma (MALT type) with extreme plasmacytic differentiation: Report of a case diagnosed by fine-needle aspiration and flow cytometric study Diagn Cytopathol 200431526 10.1002/dc.20086 15236266
Forrest CH Frost FA de Boer WB Spagnolo DV Whitaker D Sterrett GF Medullary carcinoma of the thyroid. Accuracy of diagnosis by fine-needle aspiration cytology Cancer (Cancer Cytopathol) 199884295302 10.1002/(SICI)1097-0142(19981025)84:5<295::AID-CNCR5>3.0.CO;2-J
Kaba S Hirokawa M Kuma S Maekawa M Yanase Y Kojima M et al Cytologic findings of primary thyroid MALT lymphoma with extreme plasma cell differentiation: FNA cytology of two cases Diagn Cytopathol 2009378159 10.1002/dc.21106 19526579
Papaparaskeva K Nagel H Droese M Cytologic diagnosis of medullary carcinoma of the thyroid gland Diagn Cytopathol 2000223518 10.1002/(SICI)1097-0339(200006)22:6<351::AID-DC5>3.0.CO;2-T


Cytologic evaluation of solitary thyroid nodule in a child

CLINICAL HISTORY

A 8-year-old boy with right sided thyroid lesion (1.5 × 1.5 cm firm, mobile, non-tender which moved with swallowing) since 1 year in addition to 1 × 0.5 cm lesion. Computed tomography scan lesion was heterogeneously enhancing with a few enlarged lymph nodes. Fine-needle aspiration (FNA) showed findings seen in Figure 1.

a through e: Aspirates revealed dispersed cells with finely granular cytoplasm showing some pink granules in MGG stained smears and with eccentric nuclei, inconspicuous nucleoli, and few binucleate and multinucleated cells (arrow) with loose aggregates showing nuclear pleomorphism. (a: MGG×200, b: PAP×400, c: MGG×400, d: MGGx400, e: PAP×400), with intranuclear cytoplasmic pseudoinclusions (arrow in ‘c’). f and g: The H and E stained smear (×100) showed homogenous, eosinophilic material which demonstrated yellowish to apple green birefringence under polarizing microscopy. h and i: The cells were immunoreactive for calcitonin (h) and chromogranin (i).

A 8-year-old boy presenting with the right sided thyroid swelling and a separate swelling in the neck (arrows).

QUESTION

What is your interpretation?

Adenomatous nodule with oncocytic features

Hurthle cell neoplasm

Papillary thyroid carcinoma

Medullary thyroid carcinoma.

Answer

The correct cytological interpretation is

d. Medullary thyroid carcinoma (MTC).

EXPLANATION

Cytology smears were cellular, comprising predominantly of dispersed cells and few loose cellular aggregates [Figure 1a]. Cells were oval to polygonal in shape and showed pleomorphism, with presence of anisokaryosis and some binucleate to rare multinucleate cells [Figures 1d and e]. Nuclei were round and placed eccentrically and the simultaneous presence of moderately abundant, and fairly dense cytoplasm with well-defined borders imparted a distinct plasmacytoid aspect [Figures 1b-e]. Intranuclear cytoplasmic inclusions were also noted [Figure 1c]. Nucleoli were inconspicuous. Few of the cells showed pink cytoplasmic granules [Figure 1d]. These appearances were fairly characteristic of MTC. Additional confirmation was forthcoming by way of few dense amorphous clumps of eosinophilic material evident on hematoxylin and eosin staining [Figure 1f], which was verified to be amyloid by virtue of yellow birefringence under the polarizing microscope subsequent to Congo red staining [Figure 1g]. Finally, intense cytoplasmic positivity for calcitonin [Figure 1h] and chromogranin [Figure 1i] was demonstrable on immunocytochemistry.

ADDITIONAL QUESTIONS

Regarding MTC which of the following features is not true

It can occur in sporadic or hereditary forms

In hereditary form, pattern of inheritance is autosomal dominant

It is generally unilateral

Lymph node metastasis may be seen at the time of diagnosis.

Which of the following statements is true regarding MTC –

It is a tumor of parafollicular C-cells

It secretes calcitonin

Amyloid is found in 80–85% of cases

All of the above.

Which of the following is/are cytological feature of MTC?

Dispersed cellular aspirate with anisocytosis

Eccentric nuclei with binucleate and multinucleate forms

Cytoplasmic granularity

All of the above.

Which of the following immunocytochemical stains yield negative results in MTC

Thyroglobulin

Calcitonin

Chromogranin

Synaptophysin.

ANSWERS TO ADDITIONAL QUIZ QUESTIONS

Answers: Q1 – (c), Q2 – (d), Q3 – (d), Q4 – (a).

Q1 (c) – It is generally unilateral. The correct answer is

C. Different clinical forms of MTC are hereditary and sporadic. In hereditary MTC, pattern of inheritance is autosomal dominant. In hereditary MTC, tumor is bilateral in 92–98% of cases and in sporadic MTC, bilaterality is seen in 0–32% of cases. Sporadic MTC shows lymph node metastasis at the time of diagnosis in 40–50% of cases, while in hereditary MTC, it varies from 10% to 38%, in various forms.[1]

Q2 (d) – All of the above. The correct answer is D. MTC is a malignant tumor showing parafollicular C-cell differentiation. It characteristically secretes calcitonin, but can also produce a variety of other peptide products. Histologically, it shows amyloid in 80–85% of cases, which appears as pink staining amorphous material in the form of globules or massive deposits.[1]

Q3 (d) – All of the above. The correct answer is D. Cytologically, in MTC, the aspirates are moderately to highly cellular, comprising dispersed cell population with variability in size and shape. The nuclei are often eccentrically placed with in the cytoplasm, giving a plasmacytoid appearance. Multinucleate cells and nuclear pleomorphism may be present with occasional bizzare giant cells. With MayGrunwald-Giemsa stain, a proportion of cells show typically pink, cytoplasmic granularity.[2]

Q4 (d) – All of the above. The correct answer is A. Immunocytochemically, MTC shows immunoreactivity for calcitonin, chromogranin, and synaptophysin with negativity for thyroglobulin.[2]

BRIEF REVIEW OF THE TOPIC

Thyroid swellings are uncommon in the pediatric age group and the reported incidence, in the age group of 8–18 years, is estimated to be 0.05–1.8%, whereas, in adults, the reported incidence ranges between 3.2% and 8%.[3] Most thyroid nodules developing in childhood are benign and are examples of either follicular adenoma or nodular hyperplasia.[4] However, the proportion of carcinomas is much higher (5–50%) than that seen in adults (5–15%),[5] to the point of surpassing the benign processes in some series. Papillary thyroid carcinoma accounts for 90% of all pediatric thyroid cancers followed by medullary carcinoma (about 5%) and Hurthle cell neoplasms. Follicular carcinoma, poorly differentiated carcinoma, and anaplastic thyroid carcinoma are extremely rare.[4,6] The most frequent cause of goiter in children is autoimmune thyroid disease; although a benign colloid goiter is also a common cause.[7] MTC is a rare tumor arising from parafollicular C-cells.[8] It accounts for <5% of all thyroid cancers.[9] The incidence of pediatric MTC is estimated to be 0.03 cases/100,000 population/year.[10] Due to extremely low incidence of pediatric MTC, we should have awareness of this entity. MTC occurs either sporadically or in an inherited autosomal dominant manner. In adults, sporadic MTC accounts for 65–75% of MTC, but in children, sporadic MTC is very rare; the vast majority of MTC diagnosed in the childhood are hereditary.[10] Hereditary, MTC occurs as familial MTC or as a part of multiple endocrine neoplasia Type 2A and 2B syndromes. MTC diagnosed during childhood almost always results from activating mutations in the RET protooncogene, which encodes the RET receptor tyrosine kinase.[11] The prognosis of pediatric MTC is extremely satisfactory, with a 5-year overall survival (OS) of 95% and 15 year OS of 86%.[10]

The aspirates of MTC are cellular, comprising dispersed cells to loosely cohesive groups of plasmacytoid, spindle, and/or epithelioid cells.[12,13] The characteristic nuclear features include variable nuclear pleomorphism, bi or multinucleated tumor cells, and granular chromatin pattern.[14] The presence of intranuclear inclusions in MTC has been described.[12] These are not diagnostic of MTC, as they are also observed in papillary thyroid carcinoma as well as some benign lesions of the thyroid. The cells have variable amount of cytoplasm ranging from scant to abundant. The cytoplasm is gray or gray-reddish with fine granules, which represents neurosecretory granules. Bose et al. have reported the presence of cytoplasmic granules in 85% of their cases.[12] Amyloid has been demonstrated in 43–81% of MTC cases.[12,13] Congo red staining has been used to highlight the presence of amyloid.[13] The use of immunocytochemistry for calcitonin is sufficient for the diagnosis of MTC, since it is expressed in 80% of the tumors.[15] There is also coexpression of keratin, vimentin, and CEA. Thyroglobulin expression is negative in MTC and is used as negative control marker. If calcitonin expression is negative in suspected MTC, then positive immunostaining using antibodies against chromogranin A or synaptophysin should be sought.[16] The common cytological differential diagnoses of MTC include Hurthle cell neoplasm, papillary thyroid carcinoma and metastatic tumor, particularly melanoma. Hurthle cell neoplasm is a frequent consideration, as it also features dispersed to loosely cohesive cells with moderate to abundant cytoplasm. However, in MTC, chromatin is coarse and macronucleoli are rarely seen. Furthermore, red cytoplasmic granules are observed in MTC with Romanowsky stain.[17] The extremely eccentrically placed nucleus and presence of amyloid can also assist in proper identification of MTC.[16] In both papillary thyroid carcinoma and MTC, intranuclear inclusions are seen, but other nuclear and architectural features help in distinguishing the two.[17] MTC and metastatic melanoma are difficult to distinguish by cytomorphology alone. Immunocytochemistry is helpful in these cases.[17] Furthermore, serum calcitonin levels are elevated in virtually all patients with MTC. In our patient, serum calcitonin level was elevated. He was further investigated and operated at some other center. Hence, the results of genetic studies, and thus, the type of MTC (sporadic vs. familial) is not known. However, none of his family (parents and one elder sibling) gave a prior history of the disease. The patient’s relatives were contacted and the child was well and disease free at 16 months after the surgery.

The cytomorphological features of MTC are quite distinctive and in most of the cases, definitive diagnosis on FNA can be achieved, which can be confirmed with immunocytochemistry.


References

Chan JK Tumors of the thyroid and parathyroid glands Fletcher CD Diagnostic Histopathology of Tumors 4th ed Philadelphia, PA Elsevier 20131177272
Buley ID Thyroid gland Gray W Kocjan G Diagnostic Cytopathology 3rd ed London, England Elsevier 2010487512 10.1016/B978-0-7020-3154-0.00017-X
Huang SA Thyromegaly Lifshitz F Pediatric Endocrinology 5th ed New York Informa Healthcare 200744353
Rosai J Tallini G Thyroid gland Rosai J Rosai and Ackerman's Surgical Pathology 10th ed London, England Elsevier 2011487564 10.1016/B978-0-323-06969-4.00015-5
Niedziela M Pathogenesis, diagnosis and management of thyroid nodules in children Endocr Relat Cancer 20061342753 10.1677/erc.1.00882 16728572
Rossi ED Martini M Cenci T Capodimonti S Larocca LM The role of thyroid FNA cytology in pediatric malignant lesions: An overview of the literature Cancer Cytopathol 2017125594603 10.1002/cncy.21884 28581673
Muirhead S Diagnostic approach to goitre in children Paediatr Child Health 200161959 10.1093/pch/6.4.195 20084235
Vinciguerra GL Noccioli N Cippitelli C Minucci A Capoluongo E Bartolazzi A Oncocytic variant of medullary thyroid carcinoma: A rare case of sporadic multifocal and bilateral RET wild-type neoplasm with revision of the literature Rare Tumors 201686537 10.4081/rt.2016.6536
Watkinson JC The British thyroid association guidelines for the management of thyroid cancer in adults Nucl Med Commun 200425897900 10.1097/00006231-200409000-00006 15319594
Hogan AR Zhuge Y Perez EA Koniaris LG Lew JI Sola JE Pediatric thyroid carcinoma: Incidence and outcomes in 1753 patients J Surg Res 200915616772 10.1016/j.jss.2009.03.098 19631341
Mulligan LM Kwok JB Healey CS Elsdon MJ Eng C Gardner E et al Germ-line mutations of the RET proto-oncogene in multiple endocrine neoplasia type 2A Nature 199336345860 10.1038/363458a0 8099202
Bose S Kapila K Verma K Medullary carcinoma of the thyroid: A cytological, immunocytochemical, and ultrastructural study Diagn Cytopathol 199282832 10.1002/dc.2840080107 1551364
Zeppa P Vetrani A Marino M Fulciniti F Boschi R DeRosa G et al Fine needle aspiration cytology of medullary thyroid carcinoma: A review of 18 cases Cytopathology 199013544 10.1111/j.1365-2303.1990.tb00324.x 2130998
Bhanot P Yang J Schnadig VJ Logrono R Role of FNA cytology and immunochemistry in the diagnosis and management of medullary thyroid carcinoma: Report of six cases and review of literature Diagn Cytopathol 20073528592 10.1002/dc.20502 17427218
Saad MF Ordonez NG Guido JJ Samaan NA The prognostic value of calcitonin immunostaining in medullary carcinoma of the thyroid J Clin Endocrinol Metab 1984598506 10.1210/jcem-59-5-850 6207198
Papaparaskeva K Nagel H Droese M Cytologic diagnosis of medullary carcinoma of the thyroid gland Diagn Cytopathol 2000223518 10.1002/(SICI)1097-0339(200006)22:6<351::AID-DC5>3.0.CO;2-T
Cibas ES Thyroid Cibas ES Ducatman BS Cytology 4th ed Philadelphia, PA Elsevier 201426797


A case of neck swelling with an unusual presentation

A 25-year-old female with headache for 8 months with nasal obstruction, diplopia, and diffuse mildly tender swelling in the left periorbital and frontal region for 2 months. There was a left cervical, firm, slightly mobile, non-tender 2 × 2 cm swelling. Patient had left lateral rectus palsy with decreased temporal field vision. Fine needle aspiration (FNA) of left cervical swelling was performed [Figures 1].

(a) Aspirates were high cellularity with loosely cohesive clusters and dispersed population of atypical cells (Giemsa; x100), (b) Markedly pleomorphic, oval to spindled cells with plump elongated nuclei, vesicular chromatin, prominent nucleoli and ill- defined cytoplasm (Giemsa; x400), (c) Few reactive lymphoid cells in background (Papanicolaou; x400), (d) The biopsy showed sheets of large atypical cells with brisk mitoses (H and E; x400), inset: nuclear p63 in atypical cells (DAB; x400).

QUESTION

What is you interpretation?

Granulomatous lesion

Olfactory neuroblastoma

Metastatic nasopharyngeal carcinoma (NPC), undifferentiated type

Metastasis from CNS tumor

Malignant melanoma.

ANSWER

The correct cytopathological interpretation is:

C. Metastatic nasopharyngeal carcinoma (NPC), undifferentiated type.

EXPLANATION

NPC is a high-grade epithelial malignancy which is usually seen in the elderly age group. Propensity for lymph node metastases particularly jugulodigastric lymph nodes has been commonly reported in the advanced stages.[1] NPC arising from fossa of Rosenmuller has a tendency to invade parapharyngeal space with frequent spread to trigeminal perineural space.[2] Hematogenous dissemination to bone marrow, liver, lung, etc., has been observed consistently, however rarer. The usual symptoms of NPC include cervical lymphadenopathy, nasal obstruction, epistaxis, hearing loss, and tinnitus.[3]

The fine-needle aspiration smears were highly cellular comprising of loosely cohesive clusters and dispersed population of markedly pleomorphic oval to spindled cells with plump elongated nuclei, vesicular chromatin, prominent nucleoli, and ill-defined cytoplasm. Few bizarre giant cells with binucleate and multinucleate forms including occasional atypical mitotic figures were noted. Background revealed presence of few reactive lymphoid cells. Based on cytological findings, a possibility of metastatic epithelial malignancy was considered. The differential diagnoses of masses which need to be excluded are summarized in Table 1.

Cytomorphological differentials of masses in NPC.

Diagnosis Age Cytomorphology Anaplasia Mitoses Immunohistochemistry
Squamous cell carcinoma 55–65 years Malignant cells with vesicular nuclei, large nucleoli, with evidence of keratinization Common Variable EMA, CK5/6, CK14, p63+
NPC 40–60 years Spindly cells with vesicular nuclei, prominent central nucleoli, Background shows lymphoid cells and plasma cells Common Frequent Pan CK, p63, EMA, CK5/6, CK14, CK 19+
Sinonasal undifferentiated carcinoma 50–60 years Medium to large sized cells with scant cytoplasm and well defined borders Little Frequent PanCK, CK 7, CK8, CK 18+, (CK5/6, p63 variable, p40)
Olfactory neuroblastoma 2–90 years Round cells with fragile cytoplasm and delicate chromatin, rosetting present Variable Frequent NSE, synaptophysin, CD 56, chromogranin+, (CD 99 and FLI1-)
Ewing sarcoma/PNET 2–20 years Loosely dispersed small to medium sized cells, scant cytoplasm, fine chromatin, rosetting present Uncommon Common Synaptophysin, CD99, FLI1, Vimentin+
Malignant melanoma 40–70 years Large plasmacytoid cells with inclusion like nucleoli, melanotic cytoplasm Common Frequent HMB 45, S 100, Vimentin, Melan A+

EMA: Epithelial membrane antigen, CK: Cytokeratin, NSE: Neuron-specific enolase, PNET: Primitive neuroectodermal tumor, FLI1: Friend leukemia integration-1, HMB: Human melanoma black, CD: Cluster of differentiation, NPC: Nasopharyngeal carcinoma

Contrast-enhanced computed tomography and magnetic resonance imaging (MRI) of brain showed an extra-axial isointense rounded solid mass lesion measuring 2.5 × 2.2 × 1.6 cm in the left temporal lobe invading cavernous sinus and encasing internal carotid artery with mild erosion of medial aspect of petrous temporal bone and extension into nasopharynx [Figure 2]. MRI of neck revealed a discrete well-defined homogenously enhancing soft-tissue lesion in posterior aspect of the left carotid sheath measuring 3.5 × 3 × 2.5 cm, along with few subcentimetric lymph nodes in the cervical region at Level II, III, IV, and V of the left side, largest measuring 5 mm.

Contrast-enhanced computed tomography showing a heterogeneously enhancing rounded solid mass lesion involving left temporal lobe, extending into cavernous sinus.

An immediate endoscopic biopsy was done from the nasopharyngeal mass as this was easily accessible in comparison to the intracranial lesion. Paraffin-embedded sections were studied which comprised of multiple fragments partly lined by respiratory epithelium with an underlying tumor composed of sheets of monomorphic round to oval cells with high nucleocytoplasmic ratio, round nuclei, prominent eosinophilic nucleoli, and scant cytoplasm with the presence of brisk mitoses and focal areas of fascicles of spindle cells [Figure 1d]. At places, infiltration by lymphocytes and plasma cells was seen in the stroma. Immunohistochemistry (IHC) revealed p63, cytokeratin (CK) 19, and 5/6 positivity with negative CK7 and 14 [Figure 3]. Henceforth, a final histopathological diagnosis of NPC, undifferentiated type was rendered in conjunction with IHC findings, showing extension into brain, and presenting as a huge neck mass. The prospective plan of action of operating the brain tumor could not be executed because of the widespread extension of the tumor and the patient was referred to oncology department. The patient has received 3 cycles of cisplatin and 5_FU chemotherapy with a favorable treatment response.

(a) p63 positivity in atypical cells (DAB; x400), (b) some cells reveal CK5/6 positivity (DAB; x400), (c) many cells reveal CK 19 positivity (DAB; x400), (d) cells are negative for CK 7 (DAB; x400).

ADDITIONAL QUIZ QUESTIONS

Q1. What is the incidence of NPC, undifferentiated type?

<1 case/100,000 population

5–10 cases/100,000 population

15–20 cases/100,000 population

20–25 cases/100000 population.

Q2. Which of the following is true regarding NPC, undifferentiated type?

Epstein–Barr virus (EBV) association is less frequently seen in NPC, undifferentiated type as compared to keratinizing NPC.

Subclassification into undifferentiated and differentiated types has no prognostic significance.

Undifferentiated NPC has a high propensity for locally advanced tumor growth and a lower propensity for lymph node spread.

HPV infection is not associated with undifferentiated NPC.

Q3. Which IHC marker is not expressed in NPC?

CK 5/6

CK 19

Pan CK

CK 7.

ANSWERS TO ADDITIONAL QUIZ QUESTIONS

Answers: Q1-a, Q2-b, Q3-d.

Q1– NPC is an uncommon tumor with annual incidence of <1 case/100,000 population.[4]

Q2– It has been shown in many studies that non-keratinizing NPC (NK-NPC), especially undifferentiated type has a strong association with EBV infection.[5] Moreover, EBV is generally absent in keratinizing NPC (K-NPC). Subclassification of NPC into differentiated and undifferentiated types carries no clinical or prognostic significance.[6] Undifferentiated NPC has a high propensity for locally advanced tumor growth along with higher propensity for LN spread.[7] Conversely, K-NPC has a lower propensity for LN metastasis, however, local advancement is high. HPV-related NPC is likely to be undifferentiated type, especially in non-endemic region.[7]

Q3– NPC stains strongly for p63, panCK, and CK19. Immunohistochemical staining for CK 7 and 14 is negative.[8]

BRIEF REVIEW OF THE TOPIC

NPC is a rare malignant disease with complex and unique etiology. It usually presents as nasal obstruction or cervical lymphadenopathy as an early manifestation. In advanced cases, tumor can invade the orbital fissure or the orbital apex and can result in compressive optic neuropathy or direct infiltration of the optic nerve.[9] Wong et al. described a series of 354 patients who had been diagnosed with NPC and found that 27 out of these 354 had ophthalmic involvement but only eight of these 27 had ocular signs without orbital invasion.[9] Most of the patients had infiltration of cavernous sinus and sphenoid sinus with associated third and sixth cranial nerve palsy. This is dissimilar to the present case, as our patient had involvement of the second and sixth cranial nerves, sparing all the other cranial nerves without any intraorbital spread.

This case is interesting from pathologist’s point of view because of the broad differential in the head and neck area. Histopathological findings and IHC played a key role here to help us reach a definite diagnosis.

SUMMARY

The present case highlights the importance of ocular symptoms as the rare and first manifestation of NPC. We should be aware that NPC should be added to the list of differential diagnosis in patients with intracranial disease where the exact location of primary tumor is unclear.

COMPETING INTEREST STATEMENT BY ALL AUTHORS

The authors declare that they have no competing interest.

AUTHORSHIP STATEMENT BY ALL AUTHORS

Each author has participated sufficiently in the work and takes public responsibility for the appropriate portions of the content of this article. SJ: Conceptualization, drafting of the manuscript, literature review. MK: Data acquisition, revising it critically for important intellectual content. MB: Critical review, finalization of the manuscript. Each author acknowledges that this final version was read and approved.

ETHICS STATEMENT BY ALL AUTHORS

As this is a quiz case without identifiers, our institution does not require approval from Institutional Review Board (IRB).

LIST OF ABBREVIATIONS (IN ALPHABETIC ORDER)

CK – Cytokeratin

EBV – Epstein–Barr virus

FNA – Fine-needle aspiration

FNAC – Fine-needle aspiration cytology

IHC – Immunohistochemistry

K-NPC – Keratinizing nasopharyngeal carcinoma

NK – Non-keratinizing

NPC – Nasopharyngeal carcinoma.

EDITORIAL/PEER-REVIEW STATEMENT

To ensure the integrity and highest quality of CytoJournal publications, the review process of this manuscript was conducted under a double-blind model (authors are blinded for reviewers and vice versa) through automatic online system.


References

Shen C Ying H Lu X Hu C Nasopharyngeal carcinoma with central nervous system metastases Medicine 20179649 10.1097/MD.0000000000009175 29245363
Su CY Lui CC Perineural invasion of the trigeminal nerve in patients with nasopharyngeal carcinoma. Imaging and clinical correlations Cancer 19967820639 10.1002/(SICI)1097-0142(19961115)78:10<2063::AID-CNCR5>3.0.CO;2-P
Wani SQ Khan T Wani SY Najmi AM Malik TR Lone MM et al Nasopharyngeal carcinoma with atypical ophthalmic presentation-a rare case report World J Surg Med Radiat Oncol 201543640
Chang ET Adami HO The Enigmatic epidemiology of nasopharyngeal carcinoma Cancer Epidemiol Biomarkers Prev 200615176577 10.1158/1055-9965.EPI-06-0353 17035381
Young LS Dawson CW Epstein-Barr virus and nasopharyngeal carcinoma Chin J Cancer 20143358190 10.5732/cjc.014.10197
Thompson LD Update on nasopharyngeal carcinoma Head Neck Pathol 20071816 10.1007/s12105-007-0012-7 20614287
Lo EJ Bell D Woo JS Li G Hanna EY El-Naggar AK et al Human papillomavirus and WHO type I nasopharyngeal carcinoma Laryngoscope 201012019907 10.1002/lary.21089 20824783
Franchi A Moroni M Massi D Paglierani M Santucci M Sinonasal undifferentiated carcinoma, nasopharyngeal-type undifferentiated carcinoma, and keratinizing and nonkeratinizing squamous cell carcinoma express different cytokeratin patterns Am J Surg Pathol 2002261597604 10.1097/00000478-200212000-00007 12459626
Wong WM Young SM Amrith S Ophthalmic involvement in nasopharyngeal carcinoma Orbit 2017368490 10.1080/01676830.2017.1279658 28388349


An unusual breast malignancy with central cystic lesion: Important related pitfall

A 43-year-old female was referred for fine-needle aspiration (FNA) of two lumps in the left breast noticed 4 months back. On examination, there was a lump in the upper outer quadrant, 3 cm in diameter while another lump was felt in the central quadrant, 6 × 5 cm in size. The larger lump was smooth, soft to firm in consistency. It was not fixed to the overlying skin or underlying muscle. Mammogram was reported as BIRADS 3. An USG-guided FNA was performed from both the lumps. Cytologic features of the FNA from the larger lump are shown in Figures 1a and 1b. FNA findings from the smaller lump are shown in Figures 1c and 1d.

Aspiration cytology smear from larger lump shows occasional cluster of benign keratinized squamous cells (a, Giemsa x100). Papanicolaou-stained smear of the same shows benign-appearing squamous cells (b, x100).

QUESTION

What is the cytological interpretation on FNA smears of the larger lump?

Cystic lesion with squamous metaplasia

Squamous metaplasia secondary to infarcted fibroadenoma

Infarcted phyllodes tumor with squamous metaplasia

Squamous cell carcinoma of the breast

The correct cytological interpretation is:

a. Cystic lesion with squamous metaplasia.

EXPLANATION

USG-guided FNA from the smaller lump (upper outer quadrant) showed features of proliferative breast disease with the presence of stromal fragments and clusters of ductal epithelial cells showing focal mild anisonucleosis in a background of bipolar myoepithelial cells [Figure 2a]. FNA smears from the larger lump (central quadrant) showed a few inflammatory cells, an occasional cluster of benign squamous cells and histiocytes. Diligent search failed to reveal any features suggestive of malignancy. Hence, a cytological impression of proliferative breast disease with cystic change and squamous differentiation was rendered. In view of the patient’s age, size of the lump and mild anisonucleosis in aspiration smear, a core biopsy was performed. The core biopsy of the larger lump showed benign breast tissue and a tiny island of neoplastic squamous cells that were negative for CK7. In view of the suspicious features on core biopsy, a modified radical mastectomy was performed. The excised specimen showed a large mass with central cystic zone occupying almost the entire breast parenchyma with unremarkable skin, nipple, and areola.

Histopathological examination revealed features of a well-differentiated squamous cell carcinoma (SCC) comprising almost 100% of the tumor area [Figure 2b]. One of the ducts in this area had metaplastic squamous lining displaying transition to frank SCC [Figure 2c]. The peripheral areas of the mass showed fibroadenomatoid change with foci of atypical ductal hyperplasia (ADH) and focal ductal carcinoma in situ (DCIS) adjoining the areas of SCC. On immunohistochemistry, the tumor was triple-negative (estrogen receptor [ER], progesterone receptor [PR], and Her2neu) as well as negative for CK7 while being positive for pan-CK and CK5/6 [Figure 2d]. A thorough clinical examination and follow-up positron emission tomography computed tomography (PET-CT) scan failed to detect any other primary site; hence, the final diagnosis was rendered as primary squamous cell carcinoma of breast (SCCB) with coexisting spectrum of proliferative breast disease encompassing ADH and DCIS. The patient was referred for chemotherapy. She was negative for relapse or distant metastasis during 12 months follow-up.

Histopathological section shows the invasive squamous cell carcinoma component (a, H&E x100). A central cystic zone with squamous metaplasia (sq meta) and transformation to SCC and infiltration denoted as I is also seen (b, H&E x 40). The focus shown in ‘b’ is highlighted by immunohistochemistry for CK5/6 (c, Labeled Streptavidin-Biotin method x 40).

ADDITIONAL QUIZ QUESTIONS

Q1. The cytomorphologic features on FNA of smaller breast lump are suggestive of:

Infiltrating duct carcinoma of breast

Fibrocystic disease of the breast

Proliferative breast lesion

Ductal carcinoma in situ.

Q2. On immunohistochemistry of a breast lesion, which of the following suggest a diagnosis of SCC?

Negative for CK 7 expression

Negative for ER PR and HER2 Neu

Positive for Pan-CK and CK5/6 expression

All of the above.

Q3. Which of the following possibilities should be considered in a FNA of breast mass showing atypical squamous cells

Infarcted fibroadenoma

Epidermoid cyst

Phyllodes tumor with metaplasia of canalicular lining cells

SCC

All of the above.

Answers to the quiz questions:

Q1. The correct answer is c (proliferative breast lesion)

Q2. The correct answer is d (all of the above)

Q3. The correct answer is e (all of the above).

The explanations of these questions follow in the next section.

DISCUSSION

Primary SCCB is very rare, accounting for <1% of all invasive breast carcinomas. There have been only sporadic case reports and a few mini-case series reported in the literature.[1-5] SCCB most commonly presents in elderly patients with a palpable breast lump, which is sometimes bigger than 5 cm and may mimic a breast abscess.[5] Similar to the basal-type breast cancer, majority of the cases of SCCB are triple negative with a cumulative 5-year survival rate of 51–68%.[6] SCCB has been defined by the WHO as a subtype of metaplastic carcinoma showing atypical squamous cells.[7] Rosen recommends that carcinomas with more than 90% of the component as squamous should be categorized as SCCB.[8] However, few reports in literature have classified tumors with squamous component of 50–100% or spindle cell variant in this category leading to confusion in the true incidence of primary SCCB.[2] We have utilized the Rosen’s criteria for defining our case as SCCB.

Macia et al. stated that a primary SCCB should meet all of the three criteria: (1) Absence of other neoplastic components like ductal or mesenchymal, in the tumor; (2) tumor independent of the adjacent cutaneous structures; and (3) no other primary epidermoid tumors in the patient.[9] Our case met these three criteria and hence was considered as primary SCCB. The proposed hypotheses for the histogenesis of SCCB include origin from keratinous cyst or chronic abscesses or metaplastic glandular tissue.[10] The tumor in our report demonstrated a focus of metaplastic lining transiting into frank SCC. We had another case of an elderly woman with initial mammographic findings of a cystic lesion and FNA showing benign squamous cells and inflammatory cells, similar to the present case. A repeat mammogram was reported as BIRADS3/4 and a core biopsy showed features of SCC. Histopathological examination of the mastectomy specimen and PET-CT scan confirmed a diagnosis of primary SCCB, possibly having originated from a dermoid cyst.

Cytologic diagnosis of SCCB has been cited in the literature as rare case reports only.[6,11-13] The characteristic cytologic findings include malignant squamous cells in loose cohesive clusters or lying singly, tadpole cells, keratin pearls, and cellular keratinization, similar to SCC of other body sites.[12] An admixture of malignant squamous cells with glandular or mesenchymal component should prompt a cytologic diagnosis of mixed carcinoma or metaplastic carcinoma.[14] Rarely, SCCB has also been reported as an intracystic tumor, postulated to originate through metaplasia of the cyst wall.[15] SCCB usually presents as a large tumor with central cystic degeneration in over half of the cases.[10]

In the current case, the tumor had cystic component. However, the cyst fluid revealed benign-appearing squamous cells with inflammatory cells, leading to a diagnostic pitfall on cytology. This case highlights the problem of sampling artifact, if enough sampling is not performed from all suspicious areas of the cystic lesions. At least three adequately cellular aspirates should be sampled from the periphery of the cystic lesions with ill-defined periphery. The sensitivity and specificity of FNA in diagnosis of breast masses have been variably reported as 94–99% and 99–100%, respectively.[16]

FNA forms an integral component of the triple test used widely in the evaluation of breast lesions. The diagnostic accuracy of triple test has been reported to be 100% if all the three tests are concordant for the benign or malignant diagnosis.[17] A study utilizing a triple test score showed that only 8% of the masses received a score of 5 (equivocal) and required a confirmatory biopsy.[18] Our patient had benign clinical, mammographic, and FNA findings, suggesting that the triple test was false negative.

Mere presence of atypical squamous cells in the FNA smears is not sufficient to confer a diagnosis of SCCB since similar cells may be seen in epidermoid cysts, metaplasia secondary to infarction of fibroadenoma or papilloma, phyllodes tumor with intracanalicular metaplastic lining cells, and subareolar abscess (Zuska’s disease).[11] Subareolar abscess, a rare benign lesion of the breast, usually presents with recurrent nipple discharge, mastalgia, and breast masses with nipple retraction. Radiologic differentiation from a malignant lesion of the breast may be difficult in such cases and histopathologic examination of the excised sinus or fistula tract provides the correct diagnosis.[19] Cases showing malignant squamous cells in breast FNA need to be differentiated from ductal carcinoma with squamous component and carcinosarcomas.[10] Considering the criteria defined by Marcia et al. for primary SCCB, FNA can only suggest a diagnosis of SCCB, which needs to be confirmed after adequate histopathologic examination of the excised tumor.[10] Another differential diagnosis to be considered is apocrine carcinoma of the breast, which demonstrates granular cytoplasm, low nuclear-cytoplasmic ratio, and enlarged nucleolus. Streaming arrangement of cells in a cluster, less conspicuous nucleoli, cannibalism, and necrotic background has been shown to be characteristic cytologic features of SCCB assisting in its differentiation from apocrine carcinoma.[6]

The optimal therapy for SCCB is surgery (breast conserving or mastectomy) with or without axillary lymphadenectomy. The role of post-operative adjuvant chemo or radiation therapy is as yet not known.[20]

SUMMARY

Primary SCC of the breast is a rare subtype of breast cancer. The clinical, mammographic, and aspiration cytologic features of this tumor may be misleading, especially in cases presenting with a cystic mass. Presence of benign-appearing squamous cells in cystic lesions should suggest better sampling to detect associated invasive carcinoma. This case highlights the problem of sampling artifact, if enough sampling is not performed from all suspicious areas of the cystic breast lesions with squamous cells. At least three adequately cellular aspirates should be sampled from the periphery of the cystic lesions with ill-defined periphery.


References

Hennessy BT Krishnamurthy S Giordano S Buchholz TA Kau SW Duan Z et al Squamous cell carcinoma of the breast J Clin Oncol 200523782735 10.1200/JCO.2004.00.9589 16258085
Nayak A Wu Y Gilcrease MZ Primary squamous cell carcinoma of the breast: Predictors of locoregional recurrence and overall survival Am J Surg Pathol 20133786773 10.1097/PAS.0b013e3182877569 23629447
Chen XL Luo J Xu FL Squamous cell carcinoma of the breast: Particularity and clinical management Int J Clin Exp Med 201691416774
Lim GH Acosta HA Gudi MA Natural history of metaplastic squamous cell breast cancer: A case report and literature review on surgical management Gland Surg 2017673841 10.21037/gs.2017.05.10 29302494
Punzo C Fortarezza F de Ruvo V Minafra M Laforgia R Casamassima G et al Primitive squamous cell carcinoma of the breast (SCCB): Case report of an uncommon variant of metaplastic carcinoma G Chir 20173813942 10.11138/gchir/2017.38.3.139 29205144
Kinoshita M Matsuda Y Arai T Soejima Y Sawabe M Honma N Cytological diagnostic clues in poorly differentiated squamous cell carcinomas of the breast: Streaming arrangement, necrotic background, nucleolar enlargement and cannibalism of cancer cells Cytopathology 201829227 10.1111/cyt.12461 28868787
Ellis IO Cornelisse CJ Schnitt SJ Sasco AJ Sastre-Garau X Kaaks R et al Invasive breast carcinoma Lakhani SR Ellis IO, Schnitt SJ, Tan PH, van de Vijver MJ, editors. WHO Classification of Tumours of the Breast. Lyon IARC Press 2012163208
Rosen PP Rosen's Breast Pathology Philadelphia, PA Lippincott Williams & Wilkins 200145561
Macia M Ces JA Becerra E Novo A Pure squamous carcinoma of the breast, Report of a case diagnosed by aspiration cytology Acta Cytol 1989332014
Vera-Alvarez J García-Prats MD Marigil-Gómez M Abascal-Agorreta M López-López JI Ramón-Cajal JM Primary pure squamous cell carcinoma of the breast diagnosed by fine-needle aspiration cytology: A case study using liquid-based cytology Diagn Cytopathol 20073542932 10.1002/dc.20658 17580346
Ng WK Kong JH Significance of squamous cells in fine needle aspiration cytology of the breast. A review of cases in a seven-year period Acta Cytol 2003472735 10.1159/000326471 12585027
Annam V Giriyan SS Kulkarni MH Diagnosis of pure squamous cell carcinoma of the breast by fine needle aspiration cytology Acta Cytol 2009537223 10.1159/000325422 20014569
Teerthanath S Hariprasad S Krishna US Primary intracystic squamous cell carcinoma of the breast: A case report and review of the literature J Cytol 20092615860 10.4103/0970-9371.62189 21938184
Cha N Wang S Lv M Wang DW Zhang XJ Zheng M et al Breast metaplastic squamous cell carcinoma diagnosed with fine needle and core biopsy: A case study Am J Case Rep 2018192036 10.12659/AJCR.907254 29472521
Shigekawa T Tsuda H Sato K Ueda S Asakawa H Shigenaga R et al Squamous cell carcinoma of the breast in the form of an intracystic tumor Breast Cancer 20071410912 10.2325/jbcs.14.109 17245005
Aker F Gümrükçü G Onomay Erkan M Gürleyik G Kiliçoglu G et al Accuracy of fine-needle aspiration cytology in the diagnosis of breast cancer a single-center retrospective study from Turkey with cytohistological correlation in 733 cases Diagn Cytopathol 20154397886 10.1002/dc.23380 26466750
Baykara M Özkan Z Gül Y Aslan Ö Güngör L Effectiveness of the triple test and its alternatives for breast mass evaluation J Breast Health 201391959 10.5152/tjbh.2013.32
Morris KT Pommier RF Morris A Schmidt WA Beagle G Alexander PW et al Usefulness of the triple test score for palpable breast masses Arch Surg 2001136100813 10.1001/archsurg.136.9.1008 11529822
Silverman JF Raso DS Elsheikh TM Lannin D Fine-needle aspiration cytology of a subareolar abscess of the male breast Diagn Cytopathol 1998184414 10.1002/(SICI)1097-0339(199806)18:6<441::AID-DC11>3.0.CO;2-H
Behranwala KA Nasiri N Abdullah N Trott PA Gui GP Squamous cell carcinoma of the breast: Clinico-pathologic implications and outcome Eur J Surg Oncol 2003293869 10.1053/ejso.2002.1422 12711295


An uncommon diagnosis in pleural effusion cytology

A 50-year-old man presented with complaints of breathlessness of 3-month duration. Computed tomogram (CT) scan of thorax showed a large 4 × 3 cm, predominantly cystic mass in his left anterosuperior hemithorax with necrotic lymph nodes and left-sided severe pleural effusion. Five hundred milliliters of pleural fluid were tapped and cytology examination was performed for the workup. The cytological picture is shown in Figure 1.

(a) Aggregates of tumor cells with round to oval morphology, moderate pleomorphism, and prominent nucleoli (Papanicolaou × 200) (b) Groups of tumor cels embedded in myxoid stroma highlighted by metachromatic magenta (May Grunwald Giemsa × 200) (c) Ultrasonography scan of thorax shows moderate-to-severe left pleural effusion with internal echoes (d) Pleural fluid cell block shows groups of tumor cells within amphophilic hyaline stroma (H and E × 200).

QUESTION

What is your diagnosis?

Reactive mesothelial cells

Metastatic adenocarcinoma

Positive for malignancy, possibly myxoid chondrosarcoma

Positive for malignancy, cannot characterize further.

ANSWER

The correct cytological interpretation is C. Positive for malignancy, possibly myxoid chondrosarcoma.

EXPLANATION

Grossly, the fluid sample was straw in color. Centrifugation was done at 2000 rpm for 10 min. The supernatant was discarded and the sediment was picked up with the help of a cotton-tipped applicator stick and smears were prepared by rolling the swab stick on glass slides. The smears were fixed in 100% methanol and stained by the routine Papanicolaou (Pap) and May Grunwald Giemsa (MGG) methods.

The smears were cellular and showed atypical cells dispersed singly, in cords and small aggregates with varying amounts of granular, myxoid, and chondromyxoid stroma. The atypical cells were predominantly round to oval in morphology, displaying mild-to-moderate pleomorphism with prominent nucleoli. The cytoplasm was finely vacuolated. Occasionally, the cells showed “rhabdoid” morphology. Atypical cells did not show any specific features of epithelial differentiation. Myxoid stroma was further highlighted by MGG stain as bright magenta (metachromasia). Few reactive mesothelial cells, many red blood cells, neutrophils, and lymphocytes were also noted.

The patient had a history of myxoid chondrosarcoma of his left thigh, which was excised 5 years ago, following which, he received six cycles of adjuvant chemotherapy and radiation therapy.

The cytomorphology of the abnormal cells led to the suspicion of extraskeletal myxoid chondrosarcoma (EMC) and was supported by the clinicoradiological findings. In the given clinical context of known history of EMC of the left thigh and lung mass, pleural fluid cytology was suggestive of metastatic EMC and biopsy/cell block confirmation was advised.

Cell block was prepared from pleural fluid which showed tumor cells arranged in cords within an amphophilic hyaline stroma, consistent with metastatic EMC from a known primary in the left thigh.

Following cytology, a CT-guided biopsy of the lung mass was performed and histopathological examination showed features consistent with metastatic EMC, in a known case. By immunohistochemistry (IHC), the tumor cells were diffusely and strongly positive for neuron-specific enolase (NSE).

Considering the advanced stage of the disease and dismal prognosis, the patient was advised symptomatic treatment.

DISCUSSION

Cytologic examination of pleural fluid is a simple, noninvasive, cost-effective, diagnostic modality used to detect abnormal cells that exfoliate in the fluid. In most of the cases, cytology can recognize the origin of a neoplasm presenting in the fluid on the basis of its morphologic features so as to exclude a second possible primary tumor. When malignant cells are present in the fluid, a definite diagnosis can be made in approximately 90% of the cases.[1] Mostly tumors of the epithelial and non-epithelial origin that metastasizes to the serous membrane are diagnosed in the pleural fluid cytology. Non-epithelial tumor like sarcomas account for only 3–6% of malignant effusions, however, their diagnosis is made in the setting of a known primary tumor.[2] The commonly encountered primary pulmonary malignancies are of epithelial origin. Primary tumors with cartilaginous or osteoid features originating in the lung are extremely rare, and most likely indicate metastatic disease.

Cytological findings of EMC obtained from fine-needle aspiration or imprint smears have been reported previously. However, the cytopathologic features of this rare sarcoma in effusion have not been frequently reported. To the best of our knowledge, only one case report on EMC diagnosed in pleural fluid in a patient presenting with a thigh mass has been published.[3] We report a (rare) case of EMC detected in pleural fluid in a patient presenting with a known history of EMC of the right thigh.

EMC is a rare soft-tissue sarcoma, first described by Stout and Vernar,[4] and later coined as a distinct clinicopathologic entity by Enzinger and Shiraki.[5] It is usually seen in proximal extremities of middle-aged adult males.[6] The median age of patients with this sarcoma is 48.7 years.[7] Lung is the most frequent metastatic and extremely rare primary site of myxoid chondrosarcoma.[8] It is an intermediate to high-grade sarcoma, depending on the degree of nuclear atypia and other histopathologic features, and is associated with late recurrences and lung metastasis. EMC of lung may present as an asymptomatic mass, with clinical symptoms such as hemoptysis, dyspnea, or anemia.[8-10] The myxoid stroma with varied morphology in EMC can be mistaken for other myxoid neoplasms including chordoma, myxoid liposarcoma, myxomas, low-grade fibromyxoid sarcoma, and myxofibrosarcoma. However, unlike other myxoid sarcomas, EMC is known to be associated with a specific translocation, namely, t(9;22) (q22;q12), resulting in EWSCHN (TEC) fusion gene product, which can be tested using fluorescent in situ hybridization[11,12] and is tumor-type specific or pathognomotic for this entity and assumed to play an important role in the development of EMC. Metastasis and the rate of recurrence are related to degree of cellularity and the relative amount of myxoid material. Tumors with the least cellularity and the most myxomatous changes carry the best prognosis.[13]

Detection of tumor cells in the pleural fluid is not possible unless the cells exfoliate in the fluid in large numbers.

In effusion cytology, at times, there is considerable challenge in differentiating reactive mesothelial cells from adenocarcinoma and other metastatic tumors. It is a critical situation, indicative of a high-stage disease. The reported literature on the cytologic features of sarcomas in serous effusions is very scanty. Sarcomas tend to lose the tissue arrangement or the stromal patterns as observed in fine-needle aspiration specimen.[2] They usually tend to round up in the fluids and hence they can be misinterpreted as carcinoma cells, especially in the absence of characteristic myxoid stroma.[3] Massoni and Hajdu pointed out that variable chromatin pattern in nuclei, prominent nucleoli, indistinct cytoplasmic borders, and bipolar cytoplasmic processes have been observed not only in uterine sarcomas but also they appear to be useful in distinguishing these tumors from carcinomas.[14] Accuracy improves when the cytologic findings are correlated with the clinical information and prior history.

Differential diagnoses include reactive mesothelial cells and metastatic adenocarcinoma cells. Reactive mesothelial cells show slightly enlarged nuclei with smooth and regular nuclear membrane, fine chromatin pattern, and moderate amount of cytoplasm. In this case, the atypical cells displayed unequivocal features of malignancy, including enlarged hyperchromatic nuclei, irregular nuclear border, and anisonucleosis. Therefore, the possibility of these cells being reactive mesothelial cells was not considered.

Metastatic adenocarcinoma is the most common diagnosis in effusion cytology. In most cases of adenocarcinoma, tight clusters of tumor cells are observed, including overlapping three-dimensional clusters, papillaroid formations, glandular differentiation, or “signet ring” cell morphology. Extracellular or intracellular mucinous material may be seen in certain cases. Furthermore, the tumor cells exhibit enlarged hyperchromatic nuclei with irregular nuclear membrane, coarse and clumped chromatin pattern, and anisonucleosis. However, the present case did not reveal any of the above features.

Mucinous material in Pap staining appears usually as pale purple amorphous material, while in MGG, it may appear either colorless or pink. The present case showed abundant extracellular ground substance (myxochondroid matrix). This might resemble mucinous material and can be mistaken for a diagnosis of adenocarcinoma. The metachromatic (magenta) effect of the matrix on MGG stain was a useful clue toward the diagnosis.

In the present case, the characteristic features included hypercellular smears with variable tumor cell arrangement and the presence of abundant metachromatic myxoid/ chondromyxoid stroma in the background. Small clusters and sheets of markedly pleomorphic cells with vacuolated cytoplasm, enlarged round to oval nuclei, and small nucleoli were noted. In view of previous history of myxoid chondrosarcoma, and presence of malignant cells amidst abundant myxochondroid matrix, a diagnosis of metastatic EMC was rendered. Kumar et al.[12] observed that tumor cells showing “rhabdoid” morphology are more commonly associated with aggressive clinical outcomes, including lung metastasis. The present case did show tumor cells with rhabdoid morphology and also presented with lung metastasis.

Sarcomas do not exhibit a specific immunophenotype. Application of immunocytochemical marker for EMC is not helpful. As per review of literature, diffuse positivity for vimentin and S-100 is of limited value in identifying an EMC; the expression for NSE supports the hypothesis of a possible neural or neuroendocrine differentiation recently reported in a subset of EMC, providing a new insight into their histogenetic nature. Hence, the diagnosis of these tumors in serous effusions relies mainly on morphologic evaluation[2] and clinical correlation.

ADDITIONAL CME QUESTIONS

The stromal matrix in EMC in MGG stained smears appear as

Magenta

Purple

Colorless

Pale pink.

For exact tumor subtyping in effusion, the requirements include

Detailed clinical history

Pap and MGG stained smears

Cell block and IHC

All of the above.

Chromosomal rearrangements seen in EMC include.

t(9;22)

t(10;11)

t(8;14)

t(11;22).

Answers to additional questions

Q1: A; Q2: D; Q3: A.

SUMMARY

During the metastatic workup, effusion cytology can provide a valuable and accurate means of diagnosis. The presence of sarcoma in effusions, including EMC, is extremely rare. Tumor cells with characteristic stroma, cytologic, and immunomorphologic features, in combination with clinical history, are useful in arriving at a correct diagnosis.


References

Ehya H Effusion cytology Clin Lab Med 19911144367 10.1016/S0272-2712(18)30563-8
Abadi AM Zakowski MF Cytologic features of sarcomas in fluids Cancer 199784716 10.1002/(SICI)1097-0142(19980425)84:2<71::AID-CNCR1>3.0.CO;2-G
Chen KT Effusion cytology of metastatic extraskeletal myxoid chondrosarcoma Diagn Cytopathol 2003282223 10.1002/dc.10271 12672100
Stout AP Vernar EW Chondrosarcoma of the extraskeletal soft tissues Cancer 1953658190 10.1002/1097-0142(195305)6:3<581::AID-CNCR2820060315>3.0.CO;2-T
Enzinger FM Shiraki M Extraskeletal myxoid chondrosarcoma. An analysis of 34 cases Hum Pathol 1972342135 10.1016/S0046-8177(72)80042-X
Jambkekar NA Baraniya J Baruah R Joshi U Badhwar R Extraskeletal myxoid chondrosarcoma: Clinicopathologic, histochemical, and immunohistochemical study of 10 cases Int J Surg Pathol 199757782 10.1177/106689699700500302
Tsang S Nguyen GK Aspiration biopsy cytology of metastatic extraskeletal myxoid chondrosarcoma and its cytologic differential diagnosis Acta Cytol 1985295669
Zhou Q Lu G Liu A Extraskeletal myxoid chondrosarcoma in the lung: Asymptomatic lung mass with severe anemia Diagn Pathol 20127112 10.1186/1746-1596-7-112 22925697
Goetz S Robinson R Landas S Extraskeletal myxoid chondrosarcoma of the pleura: Report of a case clinically simulating mesothelioma Am J Clin Pathol 199297498502 10.1093/ajcp/97.4.498 1553915
Ichimura H Endo K Ishikawa S Primary chondrosarcoma of the lung recognized as a long-standing solitary nodule prior to resection Jpn J Thorac Cardiovasc Surg 2005531068 10.1007/s11748-005-0011-0 15782574
Hisaoka M Hashimoto H Extraskeletal myxoid chondrosarcoma: Updated clinicopathological and molecular genetic characteristics Pathol Int 20055545363 10.1111/j.1440-1827.2005.01853.x 15998372
Kumar R Rekhi B Shirazi N Pais A Amare P Gawde D et al Spectrum of cytomorphological features, including literature review, of an extraskeletal myxoid chondrosarcoma with t(9; 22) (q22; q12) (TEC/EWS) results in one case Diagn Cytopathol 20083686875 10.1002/dc.20931 18925568
Enzinger FM Weiss SW Soft Tissue Tumours St Louis CV Mosby 1983698719
Massoni EA Hajdu SI Cytology of primary and metastatic uterine sarcomas Acta Cytol 19822651720


Detection of parasite by fine-needle aspiration cytology on unstained smear

A 38-year-old male patient presented with a swelling on the left upper arm, measuring 4 × 3 cm from past 7 years. FNAC was performed using a 22-G needle. On aspiration, drop of fluid was aspirated and air-dried smears were prepared. Before submitting the smears for staining, as a routine protocol unstained smears were evaluated microscopically for cellular adequacy [Figure 1a]. The smears were then stained with May-Grunwald-Giemsa (MGG) and evaluated [Figure 1b and c].

(a) Unstained FNA smears showing fibrillar and granular parenchyma partially obscured by uneven tegument and subtegument thrown into folds (multiple arrowheads) 100×. (b) FNA smears showing blue nuclei concentrated in the areas where the parenchyma is covered by tegument and subtegument. MGG stain 100×. (c) Other areas reveal bladder wall fragment with blue nuclei. MGG stain 400×.

QUESTION

Identify the parasite

Echinococcus

Cysticercus cellulosae

Wuchereria bancrofti

Enterobius vermicularis.

Answer

The correct cytological interpretation is

b. Cysticercus cellulosae.

EXPLANATION

Microscopic examination revealed the bladder wall fragment of cysticercus appearing as a granular, loose fibrillary sheet-like structure, partially obscured by a knobby undulating layer comprising tegument, and subtegument, which could be very well-identified in the unstained smears [Figure 1a]. Thereafter, smears were stained with MGG, which verified the presence of bladder wall of cysticercus comprising parenchyma partly covered by tegument and subtegument, as attested to by the presence of numerous blue nuclei. The folds were better visualized in the unstained preparation, being largely obscured by the numerous nuclei present in the subtegument [Figure 1b]. Other areas of the smear also revealed bladder wall fragment with blue nuclei [Figure b and c].

DISCUSSION

Cysticercosis is a parasitic disease caused by the larval stage of Taenia solium (Cysticercus cellulosae). The disease is more common in endemic areas of Central and South America, India, China, Southeast Asia, and subSaharan Africa.[1] Humans acquire this parasite by drinking contaminated water or by eating uncooked vegetables infected with eggs.[2,3] Consumption of raw vegetables such as cabbage, radish, and carrots has also been known to cause the disease.[3] Subcutaneous or intramuscular palpable parasitic nodules are commonly due to cysticercosis and it is difficult to differentiate them from the more commonly encountered benign mesenchymal tumors and lymphadenitis on physical examination alone.[4] Hence, cytologic diagnosis of cysticercosis is often a minor surprise for the referring clinician. FNAC is a simple, sensitive, cost effective, and rapid diagnostic tool for evaluation of any superficial palpable lump at various sites in the body. Demonstration of bladder wall fragment, hooklets, or calcareous corpuscles confirms the diagnosis of cysticercosis.[5] The bladder wall has three layers; from outside inward they are tegument, subtegument, and parenchyma. When present separately, the parenchyma is characterized by fibrils, and when in continuity with that of entrance canal, few calcareous corpuscles may be seen.[6] In our case, fluid was aspirated and cytology revealed a flat fragment with a somewhat fibrillary and granular part (parenchyma) in association with an undulating/folded (tegument and subtegument) area which evoked strong suspicion of cysticercosis in the unstained air dried smear. It is speculated that hooklets would also lend themselves to easy detection in unstained smears.

On cytology, cysticercosis can be differentiated from other parasite. In Echinococcus, the bladder wall is thick and laminated, while, it is thin and membranous in cysticercosis. Multiple small scolices are seen in Echinococcus, whereas single scolex is seen in cysticercus. In coenurus, multiple protoscolices are seen, which can be distinguished from the cysticercus, which have a single scolex.[7] Microfilaria or of Wucheria bancrofti can be easily distinguished from cysticercus, based on the characteristic cytomorphology, that is, sheathed larvae with tail-tip free from nuclei.[8]

Recognition of cysticercus on unstained smears is of some importance as numerous smears may be generated from the aspirated fluid, only a minority of which may harbor the diagnostic parasite fragments. Selection of appropriate smears for staining is crucial, if non-diagnostic reports are to be curtailed.

ADDITIONAL CONTINUING MEDICAL EDUCATION QUESTIONS

Following statement is true regarding cysticercosis

Is caused by larvae of Taenia solium

“Embryonated eggs” are consumed by human host in drinking water or with vegetables as accidental intermediate host

Disease most commonly involves subcutaneous and muscle tissue, followed by brain and eye

All of the above.

Answer is d.

For the life cycle of taenia solium, the intermediate host is

Dog

Sheep

Pig

Horse.

Answer is c.

The aspirate of cysticercosis can reveal which of the following

Fragments of bladder wall

Scolex and hooklet

Calcareous spherules

All of the above.

Answer is d.

Which of the following statement is true regarding the cytomorphological feature of cysticercosis

Size of the hooklet is 15–40 microns

Bladder wall is thin and membranous

Multiple small scolices are observed

No inflammatory response is seen.

Answer is b.

SUMMARY

Fine-needle aspiration has a pivotal role in evaluating subcutaneous nodules caused by cysticercosis. Although stained FNA smears reliably demonstrate cysticercosis, rapid on site evaluation of unstained smears can also permit confident cytodiagnosis, if the cytologist is familiar with it.


References

Garcia HH Gonzalez AE Evans CA Gilman RH Cysticercosis Working Group in Peru Taenia solium cysticercosis Lancet 200336254756 10.1016/S0140-6736(03)14117-7
Gill M Dua S Gill P Gupta V Sen R Cytomorphological spectrum of subcutaneous and intramuscular cysticercosis: A study of 22 cases J Cytol 20102712326 10.4103/0970-9371.73294 21157562
Rajwanshi A Radhika S Das A Jayram N Banerjee CK Fine-needle aspiration cytology in the diagnosis of cysticercosis presenting as palpable nodules Diagn Cytopathol 199175179 10.1002/dc.2840070515 1954832
Kung IT Lee D Yu HC Soft tissue cysticercosis. Diagnosis by fine needle aspiration Am J Clin Pathol 1989928345 10.1093/ajcp/92.6.834 2589251
Handa U Garg S Mohan H Fine needle aspiration in the diagnosis of subcutaneous cysticercosis Diagn Cytopathol 2008361837 10.1002/dc.20792 18232008
Arora VK Gupta K Singh N Bhatia A Cytomorphologic panorama of cysticercosis on fine needle aspiration. A review of 298 cases Acta Cytol 19943837780
de Souza PE Barreto DC Fonseca LM de Paula AM Silva EC Gomez RS Cysticercosis of the oral cavity: Report of seven cases Oral Dis 200662535 10.1111/j.1601-0825.2000.tb00122.x 10918564
Jha A Shrestha R Aryal G Pant AD Adhikari RC Sayami G Cytological diagnosis of bancroftian filariasis in lesions clinically anticipated as neoplastic Nepal Med Coll J 20081010814


Subcutaneous nodule in the chest – Uncommon presentation of a common disease

A 19-year-old female presented to us with a midline swelling in the upper chest for 6 months. Physical examination revealed a reddish-brown nodular midline swelling in the upper chest measuring 0.8 × 0.8 cm. Ultrasound findings suggested a well-defined subcutaneous swelling measuring 10 × 0.8 mm in size. Underlying bony cortex was intact with no erosion. X-ray chest revealed no abnormality. Fine-needle aspiration from the subcutaneous swelling showed moderately cellular smears, with the presence of myeloid cells, erythroid cells, histiocytes, and megakaryocytes. Erythroid and myeloid series cells were seen in different stages of maturation [Figure 1a-d]. Few adipocytic fragments and red blood cells were noted in the background.

(a) Fine-needle aspiration smear showing moderate cellularity (40×, Giemsa stain), (b and c) Smears showing myeloid cells, erythroid cells in different stages of maturation (200×, Giemsa stain), (d) Smear showing hematopoietic cells with a megakaryocyte (400×, Giemsa stain).

Q1. What is your interpretation?

Leukemia cutis

Chronic myeloproliferative neoplasm

Extramedullary hematopoiesis

Primary myelofibrosis.

Please see the next page for answer and additional discussion on the topic

Answer

Q1: c. Extramedullary hematopoiesis.

Extramedullary hematopoiesis is defined as the production of blood outside the normal limits of the bone marrow.[1] In EMH, the cytological pattern is identical as in other sites such as spleen and liver. On FNAC reveals mixture of myeloid cells, erythroid cells, and megakaryocytes in various stages of maturation.

Leukemia cutis is the infiltration of neoplastic leukocytes or their precursors into the epidermis, the dermis, or the subcutis, resulting in clinically identifiable cutaneous lesions. Leukemia cutis may follow, precede, or occur concomitantly with the diagnosis of systemic leukemia. No blast cells were noted in the present case.

Diagnosis of myeloproliferative neoplasms needs hematological correlation.

Q2. Which of the following is true about EMH in case of thalassemia?

Skin is the most common site

Treatment is surgical removal of the mass

Usually occurs in liver, spleen, and lymph node

Presence of megakaryocytes is must for the diagnosis.

Q3. Which of the following is false about EMH?

Cutaneous lesions manifest as macules, papules, or nodules

Divided as para osseous and extraosseous

Cutaneous EMH is not seen in congenital infections

May represent a compensatory response to longstanding hypoxia.

Q4. Which of the following is not a microscopic feature of cutaneous EMH?

May resemble a fibrohistiocytic tumor

Dermal infiltrate predominantly composed of erythroid and myeloid elements

Chloroacetate esterase stain highlights the myeloid elements

Megakaryocytes are positive for glycophorin A immunostain.

FOLLOW-UP OF CASE

On further evaluation, patient was a diagnosed case of Beta-thalassemia major 8 years back. She has been treated with monthly blood transfusion and iron chelation therapy. She had a history of pulmonary tuberculosis, 9 years back for which she took category-1 anti-tubercular therapy. Per abdomen examination revealed palpable spleen crossing the umbilicus. Hematologic studies disclosed the following values: hemoglobin, 8.8 g/dl; WBC count, 11,200/mm3; with 64% polymorphs, 31% lymphocytes, 3% eosinophils and 2% monocytes; 6 nRBCs/100 WBCs; and platelet count, 150,000/mm3. Peripheral blood smears showed marked anisopoikilocytosis with microcytes, tear drop cells, target cells, and hypochromia [Figure 2a and b]. Repeat FNAC was done which showed similar morphology [Figure 3a-c].

(a) A reddish-brown nodular midline subcutaneous swelling in the upper chest measuring 0.8 × 0.8 cm, (b) Peripheral smear showing marked anisopoikilocytosis with microcytes, tear drop cells, target cells, and hypochromia (200×, Leishman stain).

(a) Megakaryocyte (arrow) (1000×, Giemsa stain), (b) Erythroid Island with normoblast (arrow) (1000×, Giemsa stain), (c) myeloid cell (arrow) (1000×, Giemsa stain).

Answers

Q2 – c. Usually occurs in liver, spleen, and lymph node

Q3 – c. Cutaneous EMH is not seen in congenital infections

Q4 – d. Megakaryocytes are positive for glycophorin A immunostain.

SUMMARY

During embryonic life, erythropoiesis normally takes place in this fashion, with blood production occurring in the liver, lymph nodes, and spleen. At birth, extramedullary hematopoiesis ceases and blood production shifts to the bone marrow. However extramedullary blood-producing connective tissue cells are still present in the child and adult, which, under certain circumstances, can once again become active. In this condition, foci of extramedullary hematopoiesis may arise within soft tissues.[1]

EMH in Thalassemia usually occurs at liver, spleen, kidney, lymph nodes, and paravertebral region.[2] Skin has not been described in literature so far. Skin as site for EMH has been described in myelofibrosis that too in an older patient with a mean age of 50 years.[3] Skin involvement by EMH is uncommon and has an estimated prevalence of 0.4% of cases in myelofibrosis.[4]

The cutaneous EMH lesions can manifest in a variety of ways: as macules, papules, nodules, and even ulcers. Several cases have been described of angioma-like cutaneous lesions and others with blisters and bleeding.[4]

Cytologically EMH composed of a combination of myeloid, erythroid, and megakaryocyte precursors. The erythroid precursors predominate in children, whereas megakaryocytes predominate in adults.[5]

EMH can be divided into two type: The first show para osseous foci resulting from herniation of medullary tissue from the adjacent bone as it is seen in hemolytic anemia where the marrow is hyperactive and a second type which shows extra osseous extramedullary hematopoiesis with foci in soft tissues. If the “mass” is in a para osseous location, CT or plain radiographs should be obtained to look for areas of adjacent bone destruction and for evidence of marrow expansion.[1] In our case, ultrasound findings showed a well-defined subcutaneous mass with no underlying bony erosion.

It has been described in many types of severe anemia, polycythemia, leukemia, lymphoma, hyperparathyroidism, rickets, chronic infections, and following radiation exposure, poisoning, or neoplastic replacement of the bone marrow.[1] To compensate for chronically low hemoglobin level in conditions such as b thalassemia major, myeloproliferative disorders, and other blood dyscrasias, hematopoiesis is upregulated and organs outside the bone marrow get involved in the production of red blood cells.[6] The incidence of EMH in patients with thalassemia intermedia may reach up to 20% compared to TM patients where the incidence is less than 1%.[7]

However, it has been reported nearly in every organ, but is frequently seen in hepatosplenic areas which can potentially produce fetal hemoglobin. Non-hepatosplenic EMH has been reported in numerous sites, such as lymph nodes, pleura, thyroid, maxillary antrum, the falx cerebri, pericardium, skin, synovium of joints, lungs, thymus, breast, and the dura of the brain and spinal cord.[1,8]

Specific treatment may not be required unless EMH is accompanied by symptoms. Treatment options for thalassemia patients with EMH depend on the location and mass effect symptom and include surgery, radiation, blood transfusion, and hydroxyurea or various combinations thereof.[8]

In the present case, the patient is a young female presenting with subcutaneous swelling without underlying bony connection. FNAC revealed extramedullary hematopoiesis containing hemopoietic elements of all the three lineages. So far in the literature, cutaneous EMH had not been described in thalassemia; our case is the first case with cutaneous EMH in thalassemia patient.

COMPETING INTERESTS STATEMENT BY ALL AUTHORS

The authors declare that they have no competing interests.

AUTHORSHIP STATEMENT BY ALL AUTHORS

Each author has participated sufficiently in the work and takes public responsibility for appropriate portions of the content of this article. All authors read and approved the final manuscript. Each author acknowledges that this final version was read and approved.

ETHICS STATEMENT BY ALL AUTHORS

As this is case without identifiers, our institution does not require approval from institutional review board (IRB) (or its equivalent).

LIST OF ABBREVIATIONS (In alphabetic order)

CT – Computed tomography

EMH – Extramedullary hematopoiesis

FNAC – Fine needle aspiration cytology

nRBC – Nucleated red blood cells

TM – Thalassemia Major

WBC – White blood cells.

EDITORIAL/PEER-REVIEW STATEMENT

To ensure the integrity and highest quality of CytoJournal publications, the review process of this manuscript was conducted under a double--blind model (the authors are blinded for reviewers and vice versa) through automatic online system.


References

Shawker TH Hill M Hill S Garra B Ultrasound appearance of extramedullary hematopoiesis J Ultrasound Med 1987628390 10.7863/jum.1987.6.6.283 3302293
Salehi SA Koski T Ondra SL Spinal cord compression in beta-thalassemia: Case report and review of the literature Spinal Cord 20044211723 10.1038/sj.sc.3101544 14765145
Sarma DP Extramedullary hemopoiesis of the skin Arch Dermatol 1981117589 10.1001/archderm.117.1.58 7458387
Corella F Barnadas MA Bordes R Curell R Espinosa I Vergara C et al A case of cutaneous extramedullary hematopoiesis associated with idiopathic myelofibrosis Actas Dermosifiliogr 200899297300 10.1016/S0001-7310(08)74679-9
Mizoguchi M Kawa Y Minami T Nakayama H Mizoguchi H Cutaneous extramedullary hematopoiesis in myelofibrosis J Am Acad Dermatol 1990223515 10.1016/0190-9622(90)70046-K
Kaitoukov Y Zdanovich E Moser T Chartrand-Lefebvre C Extramedullary hematopoiesis in beta thalassemia major: Multisystem involvement OMICS J Radiol 20165234
Keikhaei B Shirazi AS Pour MM Adrenal extramedullary hematopoiesis associated with b-thalassemia major Hematol Rep 20124e7 10.4081/hr.2012.e7 22826797
Vlahos L Trakadas S Gouliamos A Plataniotis G Papavasiliou C Retrocrural masses of extramedullary hemopoiesis in beta-thalassemia Magn Reson Imaging 19931112279 10.1016/0730-725X(93)90252-9


A small round blue cell tumor in urine: cytomorphology and differential diagnosis

CLINICAL HISTORY

This was a male patient in his late 70s presented with hematuria. He had Merkel cell carcinoma (MCC) of the right flank skin 5 years ago; diffuse large B-cell lymphoma (DLBCL) involving lymph nodes in the neck, retroperitoneum, and groin 2 years ago; and conventional prostatic adenocarcinoma 1 year ago. Cystoscopy showed a mass at the right side of the bladder wall. Bladder washing and biopsy were performed. The urine cytology and cell block sections showed scattered atypical cells [Figure 1]. The atypical cells were positive for CK20, MCPyv, and SATB2 immunostains and negative for CD20, PSA, P63, and GATA3.

Papanicolaou stained ThinPrep slide shows isolated MCC cells (arrows and inset) with high N/C ratios, irregular nuclear contours, stippled chromatin and scant basophilic cytoplasm; in a background of abundant neutrophils and reactive urothelial cells (×400).

QUESTION

Q1. What is your interpretation?

Diffuse large B-cell lymphoma

Metastatic prostatic adenocarcinoma

Squamous cell carcinoma

High-grade urothelial carcinoma

Metastatic Marked Cell Carcinoma (MCC).

ANSWER

The correct interpretation is E: Metastatic MCC.

EXPLANATION

Cytopathologic examination of the urine revealed scattered discohesive atypical cells of a small round blue cell tumor, in a background of abundant neutrophils and reactive urothelial cells [Figure 2]. The tumor cells were small to medium sized (about 2–4 times of a lymphocyte), with high N/C ratios and scant basophilic cytoplasm. The nuclei were round, centrally located, with mild nuclear irregularities, stippled chromatin and inconspicuous nucleoli.

Cell block sections showing the MCC cells (arrows) mixed with inflammatory cells (×400).

Due to the clinical history of multiple malignancies, immunohistochemical studies were performed on both the cell block and the bladder biopsy. Tumor cells are negative for CD20, which makes DLBCL unlikely. Tumor cells are negative for PSA, which does not support metastatic prostatic adenocarcinoma. Tumor cells are also negative for P63 and GATA3, excluding primary bladder squamous cell carcinoma and high-grade urothelial carcinoma, respectively. The atypical cells were positive for CK20, MCPyv, and SATB2 [Figure 3] and focally weakly positive for Synaptophysin, which support the diagnosis of metastatic MCC.

The tumor cells show immunostain positivity for CK20 (a, cell block, ×400), MCPyv CM2B4 clone (b, biopsy, ×200), and SATB2 (c, biopsy, ×200), confirming the diagnosis of MCC.

ADDITIONAL QUIZ QUESTIONS

Q2. Which virus is associated with MCC?

BK virus

JC virus

MCPyv

SV40

HPV.

Q3. Which of the following immunostain results are most likely seen in a metastatic MCC?

Synaptophysin+, Chromogranin+, TTF-1-, SATB2+, CK20+

Synaptophysin+, Chromogranin+, TTF-1+, SATB2- , CK20-

Synaptophysin-, Chromogranin-, GATA3+, SATB2- , CK20+

Synaptophysin-, Chromogranin-, TTF-1-, SATB2-, CD20+

Synaptophysin+, Chromogranin+, FLI-1+, SATB2-, CK20-.

Q4. When a malignant small round blue cell tumor is seen in urine cytology, the differential diagnosis should include:

Small cell carcinoma

Metastatic MCC

Lymphoma

High-grade urothelial carcinoma

All of above.

Answers to additional quiz questions

Q2: C

Q3: A

Q4: E.

EXPLANATION

Q2: More than 80% of the MCCs are associated with MCPyv infection. MCPyv is a non-enveloped double-stranded DNA virus. Although it belongs to polyomavirus family, it is distantly related to SV40 virus and has less cross-reaction with SV40 antibody (clone PAb416) than BK and JC viruses. A more specific antibody (clone CM2B4) has better detection rate of MCPyv.[1] Our case showed MCPyv CM2B4 clone positivity and SV40 antibody negativity, supporting the published results. Therefore, the use of SV40 immunostain in MCC is not recommended.

Q3: Metastatic MCC in urine shows typical neuroendocrine tumor cytomorphology, such as high N/C ratios and stippled chromatin. Immunohistochemical studies can aid the diagnosis. Most MCCs are positive for CK20 with membranous and/or perinuclear dot-like staining pattern. MCC is usually positive for one or multiple neuroendocrine markers, such as synaptophysin, chromogranin, CD56, and neuron-specific enolase. SATB2 is a nuclear matrix-associated protein expressing in Merkel cells and has been recently recognized as a highly specific marker for MCC[2] and is positive in our case.

Q4: In urine cytology, when atypical small round blue cells are seen, the differential diagnosis includes but not limited to small cell carcinoma (SmCC), lymphoma, and high-grade urothelial carcinoma.

Primary SmCC of the urinary bladder is rare and is morphologically indistinguishable from metastatic SmCC or metastatic MCC. In addition, primary SmCC, metastatic SmCC, and metastatic MCC all express neuroendocrine markers, which make differentiation extremely difficult.

Coexistence of urothelial carcinoma will favor primary SmCC in the urinary bladder. TTF-1 immunostain positivity favors SmCC over metastatic MCC. On the other hand, SATB2 positivity will support metastatic MCC. Of course, correlation with clinical history and imaging studies is very important.

Lymphoma in urine shows single atypical cells with high N/C ratios, hyperchromatic nuclei, and scant cytoplasm. Depending on the subtypes, the sizes of lymphoma cells could vary markedly. Lymphoma cells tend to have more clumped chromatin than MCC cells. When cytomorphological differences are subtle, immunohistochemical study and/ or flow cytometry are helpful in differentiation. However, some MCCs show positivity for TdT and CD10. Therefore, a multimarker panel is recommended.[3]

High-grade urothelial carcinoma can have variable cytomorphological appearances, so it should always be considered in the differential diagnosis. About half of the MCCs show P63 positivity, mimicking urothelial carcinoma immunohistochemically.[4] Rare MCCs even show focal weak staining for GATA3.[5] However, strong and diffuse immunohistochemical positivity of GATA 3 and uroplakin and negativity for neuroendocrine markers favor the diagnosis of urothelial carcinoma.

In our case, in addition to the scattered tumor cells, the urine sample also showed marked mixed inflammation. For patients with bladder tumors, it is not uncommon to have concurrent infections. Effort should be made to avoid neglecting the rare tumor cells in the background of numerous inflammatory cells and reactive urothelial cells.

BRIEF REVIEW OF THE TOPIC

MCC, previously known as trabecular carcinoma/sweat gland carcinoma/Toker’s tumor, was first described in 1972 by Toker.[6] Later, Tang and Toker suggested that this tumor may develop from Merkel cells and therefore renamed it as MCC.[7] Although its exact origin is still unclear, MCC is currently considered as a highly aggressive cutaneous neuroendocrine tumor, with greater metastatic potential than malignant melanoma.[8]

MCC is more common in males, with the most common primary site being head and neck. Heath et al.[9,10] suggested the mnemonic AEIOU for the clinical features of MCC. They are asymptomatic; expanding rapidly (doubling in <3 months); immune suppressed; older than 50 years; and UV exposed skin site, especially fair skin. The incidence of MCC in US was about 0.7 cases/100,000 people in 2013, and it is still rising.[11] The possible reasons for the rising incidence include identification of the association with Merkel cell polyomavirus (MCPyv); introduction of CK20 immunostain leading to increased detection rate; and increased aging population.[11] Although distant metastasis of MCC is common, urinary bladder is a very unusual site of MCC involvement and literature is limited to few case reports.[12-14]

The treatment for primary cutaneous MCC is surgery with optional sentinel lymph node biopsy and radiotherapy. Metastatic MCC has a bad prognosis. Recently, immunotherapy with PD-1 or PD-L1 inhibitors has been used to treat metastatic MCC. Avelumab, a PD-L1 inhibitor, is approved by FDA in 2017 for the treatment of metastatic MCC in adult and pediatric patients 12 years and older.[15]

SUMMARY

The presence of small round blue cell tumors in urine has a broad differential diagnosis, including but not limited to small cell carcinoma, lymphoma, and high-grade urothelial carcinoma. Metastatic MCC, although rare, should be considered in the differentials, especially if the patient is elderly with a history of skin MCC.


References

Pelletier DJ Czeczok TW Bellizzi AM A monoclonal antibody against SV40 large T antigen (PAb416) does not label Merkel cell carcinoma Histopathology 2018731626 10.1111/his.13483 29430700
Kervarrec T Tallet A Miquelestorena-Standley E Houben R Schrama D Gambichler T et al Diagnostic accuracy of a panel of immunohistochemical and molecular markers to distinguish Merkel cell carcinoma from other neuroendocrine carcinomas Mod Pathol 201932499510 10.1038/s41379-018-0155-y 30349028
Wong HH Wang J Merkel cell carcinoma Arch Pathol Lab Med 201013417116 10.5858/2009-0165-RSR2.1 21043828
Asioli S Righi A Volante M Eusebi V Bussolati G p63 expression as a new prognostic marker in Merkel cell carcinoma Cancer 20071106407 10.1002/cncr.22828 17599745
Mertens RB de Peralta-Venturina MN Balzer BL Frishberg DP GATA3 expression in normal skin and in benign and malignant epidermal and cutaneous adnexal neoplasms Am J Dermatopathol 20153788591 10.1097/DAD.0000000000000306 26595821
Toker C Trabecular carcinoma of the skin Arch Dermatol 197210510710 10.1001/archderm.105.1.107 5009611
Sun CC Toker C Masi JD Elias EG Primary low grade adenocarcinoma occurring in the inguinal region Cancer 1979443405 10.1002/1097-0142(197907)44:1<340::AID-CNCR2820440158>3.0.CO;2-E
Grabowski J Saltzstein SL Sadler GR Tahir Z Blair S A comparison of Merkel cell carcinoma and melanoma: Results from the California cancer registry Clin Med Oncol 2008232733 10.4137/CMO.S423 21892294
Schrama D Becker JC Merkel cell carcinoma--pathogenesis, clinical aspects and treatment J Eur Acad Dermatol Venereol 20112511219 10.1111/j.1468-3083.2011.04032.x 21923810
Heath M Jaimes N Lemos B Mostaghimi A Wang LC Peñas PF et al Clinical characteristics of Merkel cell carcinoma at diagnosis in 195 patients: The AEIOU features J Am Acad Dermatol 20085837581 10.1016/j.jaad.2007.11.020 18280333
Paulson KG Park SY Vandeven NA Lachance K Thomas H Chapuis AG et al Merkel cell carcinoma: Current US incidence and projected increases based on changing demographics J Am Acad Dermatol 20187845763 10.1016/j.jaad.2017.10.028 29102486
Santis WF Billings EJ DeWolf WC Metastatic Merkel cell tumor to bladder presenting as an encroachment tumor with gross hematuria Urology 199954163 10.1016/S0090-4295(99)00034-5
Strasser H Amann K Schrott KM Krause FS Solitary metastasis of a Merkel cell tumor to the urinary bladder Anticancer Res 20082813614
Mack DP Moussa M Cook A Izawa JI Metastatic Merkel cell tumor to the prostate and bladder Urology 2004641568 10.1016/j.urology.2004.03.027 15245960
Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/avelumab-bavencio [Last accessed on 2020 Jun 25]


A huge vulval cyst with iliac lymph node enlargement – A unique presentation of a rare tumor

A 32-year-old female presented with abdominal pain, right labial mass, and right iliac lymph node enlargement for 4 years, to the gynecology department. On examination, a fluctuant, polypoid mass involving the right labium majus was noted and clinical diagnosis of vulval cyst was made. Her MRI findings revealed a well-defined 10.0 × 10.0 × 5.0 cm right labial lesion with heterogeneous signal intensity. It revealed hyperintense signal with areas of hypointensity on T2 and predominantly hypointense on T1, without any calcifications. Another altered signal intensity lesion measuring 5 × 5 cm was seen along the right iliac vessels, suggestive of the right iliac lymphadenopathy. USG-guided fine-needle aspiration (FNA) of the right iliac lymph node was performed and slides were stained with PAP, H&E, and MGG [Figure 1].

FNAC of the right iliac lymph node (Arrowhead-plexiform vasculature, Arrow- lipoblast), PAP, ×100 (original).

Q1 – WHAT IS YOUR INTERPRETATION?

Angiomyxoma

Myxofibrosarcoma

Myxoid dermatofibrosarcoma protuberans

Myxoid liposarcoma.

Question 1 answer – d-Myxoid liposarcoma.

BRIEF DISCUSSION

Smears made from FNA of the right iliac lymph node showed tissue fragments with myxoid matrix and plexiform vascular network [Figure 1: Arrowhead] surrounded by round to ovoid cells with mild atypia admixed with univacuolated to multivacuolated lipoblasts with scalloped nuclei [Figure 1: Arrow]. A diagnosis of low-grade myxoid liposarcoma (MLS) was made on FNAC. FNAC of the iliac lymph node was followed by removal of the right labial mass which was sent to pathology department for histopathological evaluation. The lesion was a well encapsulated, grayish-white to grayish-brown, lobular mass measuring 8.5 × 5.0 × 4.0 cm [Figure 2].

MRI well-defined 10.0 × 10.0 × 5.0 cm right vulval mass showing heterogeneous signal intensity, hypointense on T1 and hyperintense on T2.

Gross evaluation revealed solid, multilobulated cut surface with grayish-white, fleshy and gelatinous areas with foci of hemorrhage [Figure 3]. Representative sections were submitted for microscopic evaluation. H&E stained sections from labial mass showed nodular lesion with hypocellular and hypercellular areas. Hypocellular areas showed uniform round to oval-shaped cells admixed with variable number of univacuolated to multivacuolated lipoblasts embedded in prominent myxoid stroma, rich in arborizing chicken wire capillary vasculature [Figure 4].

Solid, grayish-brown multilobulated mass with grayish-white fleshy and gelatinous areas (original).

Hypocellular areas showing round cells admixed with lipoblasts (arrowhead) embedded in prominent myxoid stroma, rich in arborizing, plexiform capillary vasculature (arrow). (H and E, ×400) (original) Inset=lipoblast).

Extensive grossing was done and then slides were stained with H&E. Sections from labial mass showed nodular lesion with hypocellular and hypercellular areas. Hypocellular areas show uniform round to oval-shaped cells admixed with variable number of univacuolated to multivacuolated lipoblasts embedded in prominent myxoid stroma, rich in arborizing chicken wire capillary vasculature [Figure 4]. Hypercellular areas showed predominantly sheets of round to ovoid cells (approximately 25%) with high nuclear cytoplasmic ratio and absent to scant amount of cytoplasm, favoring a diagnosis of high-grade MLS [Figure 5].

Hypercellular areas showing predominantly sheets of round to ovoid cells. (H and E, ×100) (original).

ADDITIONAL QUIZ QUESTIONS

Q2 – Plexiform blood vessels are frequently seen in all except

MLS

Superficial angiomyxoma

Lipoblastoma

Myxofibrosarcoma.

Q3 – Unimultivacuolated cells with scalloping of nuclei are the feature of

Foamy macrophage

Pseudolipoblast

Lipoblast

Adipocyte.

Q4 – Most common translocation involving MLS is?

t(12;16)(q13;p11)

t(7;16)(q33;p11)

t(17;22)(q21.3;q13.1)

t(11;12)(q23;q15).

ANSWERS TO ADDITIONAL QUESTIONS

Q2 (d); Q3 (c); Q4 (a).

Q2 (d) – Plexiform vessels are thin branching vessels usually seen in MLS, superficial angiomyxoma, and lipoblastoma. Myxofibrosarcomas, meanwhile, show thick walled, curvilinear vessels with perivascular alignment of tumor cells.

Q3 (c) – Criteria for diagnostic lipoblast include – hyperchromatic, indented or sharply scalloped nucleus with lipid-rich droplets in cytoplasm, and an appropriate histological background. Multivacuolated cells distended with hyaluronic acid (pseudolipoblast) can be seen in myxofibrosarcoma.

Q4 (a) – The entire range of myxoid/round cell liposarcoma is genetically tied to recurrent rearrangement of DDIT3 that partners with FUS in >95% of cases with a resulting FUSDDIT3 fusion [t(12;16) (q13;p11)] or partners with EWSR1 in the remaining cases with a resulting EWSR1-DDIT3 fusion [t(12;22) (q13;q12)]. Identification of either the FUS-DDIT3 or EWSR1-DDIT3 transcript is considered both highly sensitive and specific for myxoid/round cell liposarcoma, allowing its distinction from morphologically similar neoplasms.

BRIEF REVIEW OF TOPIC

Vulvar sarcomas account for only 1–3% of all vulvar malignancies and the most frequent primary vulvar sarcoma is leiomyosarcoma.[1,2] MLS of vulva is very rare and only seven cases are reported in literature till date, to the best of our knowledge [Table 1].[3-8] MLS is the second most common subtype of liposarcoma harboring translocation (12;16) (q13;p11) which creates a chimeric gene FUS-DDIT3 and it encompasses a spectrum of tumors defined by their degree of lipoblastic differentiation. It is divided into low-grade and high-grade MLS according to the WHO classification proposed in 2013. At one end of the spectrum is low-grade MLS associated with favorable prognosis and, on the other end, is, high-grade MLS, defined as having ≥5% round cell component, considered more aggressive tumor which tends to metastasize.

Clinical profile of seven cases of MLS of vulva (original).

Study Age (years) Site Duration Clinical diagnosis Maximum size (cm) Management Outcome
Brooks and LiVolsi[3] 15 Vulvar perineum, recurred on the left posterior medial thigh 20 months Soft-tissue sarcoma 18 Wide excision; local recurrence as round cell/high-grade myxoid liposarcoma 20 months later treated by chemotherapy DOD*
Donnellan and Moodley[4] 26 Left labium majus et minor 4 years Bartholin cyst 10 Local excision followed by reexcision NED+ 9 m
Wu and Tarn[5] 45 Right labium majus 72 months Lipoma 7 Local excision; reexcision of 6 cm recurrence 16 months later NED, 28 m
Schoolmeester et al.[6] 34 Left vulval mass 11 months Unknown 11.7 Local excision NED, 28 m
Baek et al.[7] 33 Bilateral perineum 4 months Unknown Wide excision NED, 2 yr
Kwak et al.[8] 37 Bilateral vulval mass 3 weeks Unknown 20 and 15 Local excision with radiotherapy NED, 44 m
Present case 32 Right labium majus 4 years Cyst 8.5 Wide excision NED, 6 m

DOD: Died of disease, +NED: No evidence of disease

MLS of vulva can be mistaken clinically as benign because of their rare location and presentation, which can lead to delayed treatment.[9,10] Like in our case, a clinical diagnosis of vulval cyst was made and patient presented with iliac lymph node metastasis which was reported as low-grade MLS. However, on histopathology of labial mass, we encountered the round cell component as well, thus rendering it as high-grade MLS. Sometimes in hypercellular variant, stroma is less myxoid and capillary network is less prominent leading to erroneous diagnosis of round cell tumor on FNAC. Hence, there is a need for extensive sampling.

Histologically, MLS can be confused with other myxoid tumors more common in the vulva such as aggressive angiomyxomas, botryoid embryonal rhabdomyosarcoma, myxoid dermatofibrosarcoma protuberans, myxofibrosarcoma, and myxoid leiomyosarcoma [Table 2].

Differential diagnosis of myxoid lesions of vulva.

Differential diagnosis Age Location Gross Microscopy IHC Genetics
Growth pattern Cellularity Morphology LB Blood vessels Background Mitotic activity
Angiomyxoma Reproductive age (30 years) Superficial and deep Polypoidal, partly circumscribedcut surface is homogenously gelatinous Infiltrative Low Spindle and stellate cells with small round hyperchromatic nuclei -nt Small thin walled to large hyalinized blood vessels Myxoid with fine collagen fibrils Rare absent CD34VimentinERPR t(12;21)(q15;q21.1)
Botryoid RMS Children (<10 years) Mucosa lined hollow organs Polypoidal with clusters of small sessile or pedunculated nodules Polypoidal Mode-rate Subepithelial condensation of tumor cells (cambium layer) comprising of primitive small round cells, stellate cells and rhabdomyoblast -nt - Myxoid Low to mode rate VimentinMyoD1Myogenin Loss of heterozygosity chromosome 11p15.5
Myxoid DFSP Young-middle-aged adults Superficial Multinodular cutaneous masses, gray-white cut surface with gelatinous areas Diffuse infiltrative Mode-rate Uniform spindle cells with plump elongate nuclei arranged in storiform pattern -nt Prominent thin-walled vessels Myxoid Low to moderate CD34 t(17;22)(q21.3;q13.1)(COL1A1-PDGFB)
Myxoid leiomyosarcoma Middle to older Deep soft tissue Well-demarcatedcut surface is fleshy white-gray mass with whorled app & foci of gelatinous changes Ill defined Mode-rate Elongated spindle cells with blunt-ended nuclei arranged in long dissecting fascicles -nt Not seen Myxoid Low SMA, CALDESMON Complex with genetic instability
MFS Elderly (60–80 yrs) Superficial and deep Multiple gelatinous nodules (superficial)Single mass with infiltrative margin (deep)cut surface is variably gelatinous Multinodular Moderate Plump, spindle or stellate cells having large atypical hyperchromatic nucleus Pseudolipoblast Curvilinear, elongated blood vessels with perivascular condensation of tumor cells Myxoid High MSA, SMA Complex karyotype
MLS Young adults Deep soft tissue Well-circumscribed, multinodularcut surface is gelatinous to fleshy Nodular Mode-rate Mixture of uniform round-oval cells and bland fusiform cells +nt Plexiform, branching Myxoid Rare VimentinS100 t(12;16)(q13;p11)(FUS-DDIT3)

LB: Lipoblast, RMS: Rhabdomyosarcoma, DFSP: Dermatofibrosarcoma protuberans, MFS: Myxofibrosarcoma, MLS: Myxoid liposarcoma, -nt: Absent, +nt: Present

SUMMARY

Vulvar MLS is an extremely rare case reported in the literature. The present case marks the seventh reporting of vulval myxoid/ round cell liposarcoma and the first one presenting with iliac lymph node metastasis. Both pathologists and clinicians should be aware of the occurrence of this entity in vulval region to ensure the correct diagnosis and appropriate management of the patient with this potentially curable neoplasm.


References

Yokouchi J Negishi Y Abe K Shirasawa K Mernyei M Radiotherapy for liposarcoma of the vulva Gynecol Oncol 2000793157 10.1006/gyno.2000.5939 11063664
DiSaia PJ Rutledge F Smith JP Sarcoma of the vulva-report of 12 patients Obstet Gynecol 1971381804
Brooks JJ LiVolsi VA Liposarcoma presenting on the vulva Am J Obstet Gynecol 1987156735 10.1016/0002-9378(87)90206-7
Donnellan R Moodley M Vulval myxoid liposarcoma Int J Gynecol Cancer 2001113212 10.1136/ijgc-00009577-200107000-00012
Wu TC Tarn JJ Vulvar myxoid liposarcoma Taiwan J Obstet Gynecol 2007462934 10.1016/S1028-4559(08)60039-X
Schoolmeester JK Leifer AJ Wang L Hameed MR Vulvar myxoid liposarcoma and well differentiated liposarcoma with molecular cytogenetic confirmation: Case reports with review of malignant lipomatous tumors of the vulvaInt J Gynecol Pathol 2015343905 10.1097/PGP.0000000000000170 25851712
Baek JY Park CS Joo SH A case of liposarcoma of the perineum Korean J Obstet Gynecol 2007509325
Kwak JH Shin SM Kim JW Lee NW Unusual bilateral vulvar liposarcoma Obstet Gynecol Sci 20145754952 10.5468/ogs.2014.57.6.549 25469348
Genton CY Maroni ES Vulval liposarcoma Arch Gynecol 1987240636 10.1007/BF02134066 3827316
Nucci MR Fletcher CD Liposarcoma (atypical lipomatous tumors) of the vulva: A clinicopathologic study of six cases Int J Gynecol Pathol 1998171723 10.1097/00004347-199801000-00004 9475187


Non-cellular morphologic markers in pleomorphic adenoma: A rare observation

A 43-year-old male presented with complaint of swelling in the left parotid region for the past 7–8 years. The swelling was painless, insidious in onset, and progressively increased in size. Local examination revealed a single, well-defined, non-tender, and firm swelling measuring 3.5 × 3 cm in the left preauricular region. Fine-needle aspiration cytology (FNAC) stained smears show the following findings [Figure 1a-d].

(a) Abundant fibrillary chondromyxoid material, numerous basophilic refractile structures (Giemsa, ×400), (b) Myoepithelial cell admixed with clusters of orangeophilic, refractile structures (Papanicolaou stain, ×1000), (c) Radially arranged, glossy, petal-like structures (Thin red Arrow) with blunt ends of refractile structures. (Papanicolaou stain, ×1000), (d) Few epithelial cells have intranuclear cytoplasmic inclusion (Thick Red Arrow) (Papanicolaou stain, ×1000).

Q1. What is your interpretation?

Tyrosine-rich crystalloid

Collagenous crystalloid

Amylase crystalloid

Calcium oxalate crystalloid

Answer: Q1-A. Tyrosine-rich crystalloid

Tyrosine-rich crystalloids (TRCs) are round to oval, refractile, floret-shaped orangeophilic (Papanicolaou stain) structures with symmetrical lobulated contour, central rounded core, and peripheral “rosette like” arrangement.[1] TRCs appear reddish-brown with Millon’s stain and turn deep purplishred with the diazotisation coupling method, confirming the presence of tyrosine.[1] Other histochemical reactions such as tryptophan, arginine, and sulfhydryl groups are also positive in TRC.[2] Collagenous crystalloids (CCs) are non-refractile, eosinophilic, rounded structures composed of radially arranged needle-shaped fibers with central clear space, occupied by a fibroblast or a small blood vessel.[3] Histochemically, CC stains bright blue with trichrome, black with methenamine silver, and gray to black on reticulin staining; while it is negative for Millon’s stain or diazotization coupling reaction.[3] Alpha-amylase crystalloids (ACs) are non-birefringent having various shape such as rod like, rhomboid, cuboidal, square, polyhedral, or needle like, appear bright orange with Papanicolaou stains, deep blue by Giemsa stains, and pink by hematoxylin-eosin stains.[4] Millon’s reagent or diazotization coupling reactions are negative.[4] Calcium oxalate crystals (COCs) are birefringent, irregular or polygonal, transparent to pinkish structure.[5] Millon’s reagent or diazotization coupling reactions are negative.[5]

Q2. What is your most probable diagnosis?

Mucoepidermoid carcinoma

Basal cell adenoma

Warthin tumor

Pleomorphic adenoma (PA)

Answer: Q2-D

PA is characterized by a variable admixture of ductal epithelial cells, myoepithelial cells, and mesenchymal matrix. The mesenchymal matrix may be either chondromyxoid matrix as in most of the cases or less commonly reveal amyloid, collagenous substance, and/or elastic fibers.[6] Rare cytomorphological variations include presence of crystalloids such as TRC, CC, calcium oxalate crystalloid, and amylase crystalloids along with the presence of nuclear changes in epithelial component such as inclusions/grooves are also identified in PA.[6,7]

Q3. Which of the following statement is incorrect?

TRCs are refractile structures seen in both neoplastic and non-neoplastic lesions of parotid glands

Histochemical reactions (Millon’s stain and diazotization coupling reaction) help to confirm the presence of tyrosine.

Intranuclear cytoplasmic inclusions are the most commonly observed features in pleomorphic adenoma.

All of the above

Answer: Q3-C

TRCs are found in both neoplastic and non-neoplastic salivary gland lesions such as PA, myoepithelioma, carcinoma ex pleomorphic adenoma, adenoid cystic carcinoma, polymorphous low-grade adenocarcinoma, terminal duct adenocarcinoma, and parotid cysts.[8-12] Intranuclear cytoplasmic inclusions in the epithelial component of PA are rare findings described in a few case reports.[13]

BRIEF REVIEW OF THE TOPIC

In 1953, Bullock first described the “innumerable crystalline aggregate” present in histologic section of PA as tyrosine crystals because its structure resembles that of tyrosine.[14] Nochomovitz and Kahn (1974) described the physical and histochemical properties as refractile, rosette-like structure exhibiting a positive reaction with Millon’s reagent.[15] On electron microscopy, TRCs are non-birefringent, electron dense, granular without internal structure thus designated as crystalloid.[8] Based on the histochemical reaction and ultrastructural findings, the correct nomenclature is tyrosine-rich crystalloids rather than tyrosine crystal.[8] The incidence of TRC in PA varies from 1.8 to 5%; however, Lemos et al. reported an incidence of up to 33% in African patients.[2,9,15,16] The pathogenesis of TRC remains ambiguous. Thomas et al. proposed that as tyrosine plays an important role in pigment metabolism, therefore, there is high prevalence of TRC in parotid tumors in Black races.[9] This hypothesis was refuted as tyrosine crystals do not get deposited in the tissues of patients suffering from tyrosinosis.[9,17] Many authors hypothesized that TRC deposition is either a manifestation of disturbed protein synthesis or by precipitation onto stromal collagen secreted by myoepithelial cells.[2,9,17] Morphology and histochemical stains may help to differentiate TRC from other crystalloids. Nuclear changes such as nuclear inclusion/grooves are rare cytomorphological variations seen in PA.[13] In the present case, we emphasize on the presence of two rare cytomorphological features seen in PA.

SUMMARY

Since TRCs are the most common crystalloids found in PA, some authors suggest that TRC can serve as a non-cellular morphologic marker of salivary gland neoplasia which might be helpful in paucicellular smears. This case report also aims to emphasize the importance of this novel observation to the novice budding cytopathologists.


References

Warfield AT Smallman LA Simultaneous bilateral pleomorphic adenomas of the parotid glands with unilateral tyrosine rich crystalloids J Clin Pathol 1994473624 10.1136/jcp.47.4.362 8027378
Chaplin AJ Darke P Patel S Tyrosine-rich crystals in pleomorphic adenomas of parotid glands J Oral Pathol 1983123426 10.1111/j.1600-0714.1983.tb00346.x 6195320
Bellizzi AM Mills SE Collagenous crystalloids in myoepithelial carcinoma: Report of a case and review of the literature Am J Clin Pathol 200813035562 10.1309/J3YVV58V22AKXHWR 18701407
Kuwabara H Ishizaki S Akashi S Yuki M Shibayama Y α-amylase crystalloid granuloma in the parotid gland Diagn Cytopathol 2015431146 10.1002/dc.23119 24554643
Pérez-Guillermo M Pérez JS Parra FJ Asteroid bodies and calcium oxalate crystals: Two infrequent findings in fine-needle aspirates of parotid sarcoidosis Diagn Cytopathol 1992824852 10.1002/dc.2840080312 1318826
Faquin WC Powers CN Matrix-containing tumors: Pleomorphic adenoma and adenoid cystic carcinoma Rosenthal DL Ali SZ, Clark DP, Erozan YS, editors. Salivary Gland Cytopathology. New York Springer Science Business Media 200895
Handa U Dhingra N Chopra R Mohan H Pleomorphic adenoma: Cytologic variations and potential diagnostic pitfalls Diagn Cytopathol 200937115 10.1002/dc.20951 18973121
Gould AR van Arsdall LR Hinkle SJ Harris WR Tyrosine-rich crystalloids in adenoid cystic carcinoma: Histochemical and ultrastructural observations J Oral Pathol 19831247890 10.1111/j.1600-0714.1983.tb00360.x 6317832
Thomas K Hutt MS Tyrosine crystals in salivary gland tumours J Clin Pathol 19813410035 10.1136/jcp.34.9.1003 6268668
Hindermann W Mentzel T Katenkamp D Tyrosine-rich crystalloids in a myoepithelioma of the minor salivary glands in the smooth palate Pathologe 19992028891 10.1007/s002920050358 10501926
Harris BR Shipkey F Tyrosine-rich crystalloids in neoplasms and tissues of the head and neck Arch Pathol Lab Med 198611070912
Cleveland DB Cosgrove MM Martin SE Tyrosine-rich crystalloids in a fine needle aspirate of a polymorphous low grade adenocarcinoma of a minor salivary gland. A case report Acta Cytol 19943824751
Das DK Anim JT Pleomorphic adenoma of salivary gland: To what extent does fine needle aspiration cytology reflect histopathological features? Cytopathology 2005166570 10.1111/j.1365-2303.2004.00208.x 15787647
Bullock WK Mixed tumor of parotid gland with tyrosine crystals in the matrix; report of case Am J Clin Pathol 19532312389 10.1093/ajcp/23.12.1238 13104364
Nochomovitz LE Kahn LB Tyrosine crystals in pleomorphic adenomas of the salivary gland Arch Pathol 1974971412
Lemos LB Baliga M Brister T Cason Z Cytomorphology of tyrosine-rich crystalloids in fine needle aspirates of salivary gland adenomas Acta Cytol 199741170913 10.1159/000333173 9390129
Gilcrease MZ Nelson FS Guzman-Paz M Tyrosine-rich crystals associated with oncocytic salivary gland neoplasms Arch Pathol Lab Med 19981226449


Parietal swelling in an old female: A diagnostic conundrum

A 60-year-old female presented with a swelling on the left side of scalp for the past 10 months. The swelling was gradually progressing in size and was associated with on and off pain. There was no history of neurological deficits, however, there was a history of chest pain for the past 1 month. On examination, there was a 4 × 3 cm bony hard swelling over the left parietal region of scalp. Her vital signs were stable and fundus examination was normal. Neurological examination did not reveal any signs of raised intracranial pressure, cranial nerve palsy, or any neurological deficit. Her routine hematological and biochemical investigations were within normal range except for an elevated erythrocyte sedimentation rate of 60 mm/h. X-ray of the skull showed a well-circumscribed lytic lesion in the left parietal convexity.

Fine-needle aspiration (FNA) from the swelling on the scalp was performed [Figure 1].

(a) FNA smears of scalp swelling showing clusters and dispersed round cells (Giemsa, ×100). (b) FNA smears of scalp swelling showing uni- and binucleate cells with eccentric nuclei (Giemsa, ×400), inset: Plasma blast-like cell with dispersed chromatin and moderate cytoplasm. (c) Squash cytology smears of parietal mass showing sheets and scattered oval cells (Giemsa, ×100). (d) Squash cytology smears of parietal mass showing plasmacytoid cells with abundant cytoplasm and eccentrically placed nuclei (Pap, ×400).

Q1. What is your interpretation?

Plasma cell lesion