Add Your Heading Text Here
Fine-needle aspiration of basaloid scalp lesion: Potential diagnostic pitfall
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 [
(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).
1. What is the most likely diagnosis?
Pilomatricoma Melanoma Merkle cell carcinoma Squamous cell carcinoma Proliferating trichilemmal tumor
Answer: a
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.[
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.[
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.[
Option e: Proliferating trichilemmal tumor is a tumor originating from the outer root sheath of a hair follicle.[
The lesion showed [
(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
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.
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
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.[
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.[
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.[
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.[
Pilomatricomas are commonly misdiagnosed and are rarely considered as part of the differential diagnosis due to multiple overlapping cytologic features as shown in
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.[
Epidermal inclusion cyst was considered on the differential and usually occurs on the scalp, neck, back, and extremities.[
MCCs do not have distinctive clinical features and can present as asymptomatic red or pink or blue papules or nodules.[
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.[ |
Anucleate and nucleated squamous cells without atypia, basaloid cells without peripheral palisading but with abrupt keratinization and keratinous debris.[ |
EIC | Asymptomatic, slowly enlarging, usually occur on the scalp, skin of head and neck, back and extremities.[ |
Firm, round, and subcutaneous nodule with central punctum.[ |
EIC is lined by a stratified squamous epithelium with a granular layer and contains lamellated keratin flakes.[ |
numerous anucleate squames alongside nucleated benign squamous cells. With secondary infection, aspirates turn turbid, showing inflammatory cells like neutrophils and histiocytes.[ |
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.[ |
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.[ |
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.[ |
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.[ |
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.[ |
TC: Trichilemmal cyst, EIC: Epidermal inclusion cyst, BCC: Basal cell carcinoma, SCC: Squamous cell carcinoma, MCC: Merkel cell carcinoma
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.
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.
FNAC – Fine-needle aspiration cytology.
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
An unusual neck mass diagnosed by fine needle aspiration: Cytological findings and challenges
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 [
(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).
Q1. What is the most likely diagnosis?
Papillary thyroid carcinoma Salivary gland myoepithelioma Polymorphous adenocarcinoma (PAC) Pleomorphic adenoma Basal cell adenoma.
Answer: C
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.[
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 [
(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).
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.[
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.[
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.
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.[
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.
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.[
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 [
(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).
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.[
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.[
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.[
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.[
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%).[
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.[
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.[
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.[
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.[
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.
FNA – Fine needle aspiration
PAC – Polymorphous adenocarcinoma.
References
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
What is the most likely diagnosis?
Secretory carcinoma Oncocytic carcinoma Apocrine carcinoma Lipid-rich carcinoma
c. Apocrine carcinoma
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 [
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.[
Which of the following immunohistochemical marker is generally negative in apocrine carcinoma of breast?
ER AR HER2 GCDFP-15
a. ER
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 [
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).
To be designated as “Carcinoma with apocrine differentiation,” _________% of the tumor cells should have distinct apocrine morphology.
50% 25% 75% 90%
d. 90%
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
Is apocrine carcinoma associated with BRCA 1 or 2?
Yes No Not applicable Cannot be commented.
b. No
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.[
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
a. Triple-negative invasive duct carcinoma of no special type
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.[
Invasive carcinoma of breast with apocrine differentiation is a special subtype of breast carcinoma.[
Apocrine carcinoma was first described by Krompecher
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
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 [
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.[
Cytogenetic analysis of apocrine carcinoma cells reveals gains of 1p, 1q, 2q, 7 and 17, losses of 1p, 12q, 16q, 17q, and 22q.[
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.[
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.[
Next-generation sequencing frequently shows loss of PIK3CA/PTEN/AKT and TP53, followed by mutations of KRAS, NRAS, and BRAF.[
Genetic studies on the AR gene showed the highest CAG repeats in DCIS with apocrine differentiation compared to fibroadenomas and invasive breast carcinomas.[
The expression of PD-L1 is low in apocrine carcinoma. They are found to be microsatellite stable.[
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.[
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.
The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.
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
LCIS – Lobular carcinoma
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
Squash surprise in an elderly female
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 [
(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.
What is the diagnosis based on the clinical history, imaging findings, and photomicrographs [ 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
1. (c) Metastatic carcinoma
2. (a) CK7, CK20, TTF-1, GATA3, PAX-8, and CDX2
Squash smear cytology is a rapid and reliable method for intraoperative diagnosis of inflammatory and neoplastic brain lesions.[
Squash smears were moderately cellular and showed cohesive clusters and glandular arrangement of tumor cells against a thin mucoid background [
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 [
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
3. (b) Metastas es
4. (d) All of the above
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.
Metastases to the brain are a known complication in approximately 20% of malignancies.[
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.[
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.
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
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 [
(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
The correct cytological interpretation is d.
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.
Q2. Hematoxylin-eosin(HE) stained sections are prepared from the tissue cores [
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×.
The correct cytological interpretation is d.
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 [
(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
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.
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[
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
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.[
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.[
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.
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.
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.[
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.[
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.[
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.[
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,[
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.
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.
CT - Computed Tomography
EHE - Epithelioid Hemangioendothelioma
FISH - Fluorescence in situ Hybridization
HE - Hematoxylin-Eosin staining
IHC - Immunohistochemical
NSCLC - Non-small Cell Lung Cancer.
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.
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
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) [
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.
The findings in Pap smear [
Low-grade squamous intraepithelial lesion (LGSIL) Atypical glandular cells of uncertain significance (AGUS) Adenocarcinoma Carcinosarcoma (Malignant-mixed Mullerian tumor)
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).[
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 [
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).
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.[
The carcinomatous component of UCS is more readily identifiable than the mesenchymal component [
Publish metadata.
S. No. | Author/Year | No. of cases | Pap type | Findings |
---|---|---|---|---|
1. | Tenti |
10 | Conventional | 7 positive smears (2 diagnosed as a mixed tumor at first examination, 1 upon revision). |
2. | Costa |
21 | Conventional | Sarcoma component was absent in 4 out of 9 (44%) positive smears available for cytology review. |
3. | Casey |
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 |
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 |
1 | SurePath (LBC) | Cervical smear positive for glandular elements and some spindle-shaped cells. |
6. | Gupta |
8 | SurePath (LBC) | Malignant epithelial component in all eight cases Sarcomatous elements in three cases. |
7. | Hanley |
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.[
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.[
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 [
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.[
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,
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
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.
Most common mutations in uterine carcinosarcoma?
TP53 mutations HER2 TERT KRAS.
Most consistent independent predictor of outcome
Age at presentation Stage Angioinvasion Depth of invasion Number of mitotic figures.
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.[
It is biologically a de-differentiated endometrial carcinoma with its own pathogenesis and molecular profile.[
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.[
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.[
Recommended treatment for UCS involves surgical staging with total hysterectomy with BSO, pelvic lymphadenectomy, para-aortic lymph node sampling, and peritoneal washings.[
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
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
(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.[
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.[
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.[
PEH is an exuberant intravascular, endothelial cell proliferation mimicking a neoplastic tumor of vascular origin with papillary tufting and “hobnail” morphology.[
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
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 [
(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 [
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.[
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.[
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.[
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.[
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.[
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.[
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
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 [
(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.
Degenerated cells Adenocarcinoma Glioma Small blue round cell tumor.
The correct cytopathological interpretation is d. Small blue round cell tumor.
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 [
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 [
(a) Tumor cells arranged in nest, chords and trabeculae (H & E ×10); (b) Tumor cells having round nucleus and powdery chromatin (H & E ×40).
All of the following are among the differentials except
Germ cell tumor Astrocytoma Melanoma Neuroendocrine tumor.
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 [
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 [
Immunohistochemistry: Strong positive synaptophysin (a), Positive Chromogranin (b), Pancytokeratin focal positive (c), and Glial fibrillary acidic protein negative (d), Ki67 >20% (e) (×40).
Which marker helped in excluding the primary nature of the lesion?
Synaptophysin Chromogranin Pan-cytokeratin GFAP.
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.
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.).
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 [
Tumor cells are positive for TTF-1 (×40).
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.
NEN can develop at any of the initial mentioned systems where the neuroendocrine cells are present.[
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 [
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.[
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
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 [
(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).
Q1. What is your diagnosis for the above findings?
Squamous cell carcinoma. Endocervical adenocarcinoma Carcinosarcoma Neuroendocrine carcinoma.
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.
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 [
(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).
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 [
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).
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.[
Our cytological features overlapped with previously described reports,[
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.[
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.
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
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
(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).
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
d. Metastatic Hurthle cell carcinoma.
The scalp swelling was [
(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).
Who gave the description of Hurthle cells?
Max Askanazy Karl Hurthle James Ewing
a. Max Askanazy.
HCC is a rare differentiated thyroid carcinoma of follicular origin, and it accounts for approximately 3–7% of all thyroid carcinomas.[
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.[
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.[
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
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 [
(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).
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?
All of the above
Answer: D. There are many studies which have identified
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.
Granulomatous mastitis is a rare benign inflammatory entity described by Kessler and Wolloch in 1972.[
Various risk factors were studied, but none were specifically related to granulomatous mastitis. Possible etiological factors include an inflammatory response to epithelial damage,
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
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 [
(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).
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.
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.”
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 [
(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.[
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.[
Furthermore, benign features suggestive of AIP do not rule out false negative results induced by sampling error.[
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.
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.[
Q3. (b) AIP most often involves the head of the pancreas.[
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.[
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
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 [
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.
The correct cytological interpretation is b. Cysticercosis.
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 [
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 (
Filariasis (Option D) is another vector borne disease and shows microfilariae. Most common causative agents of human filariasis include
Feco-oral contamination Droplet transmission Inhalation Sexual contact.
Cysticercosis is caused by larval stage of 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: Q2-a, Q3-b.
The life-cycle of
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
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.[
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.[
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.[
Cysticercosis is an infection caused due to larval stage of the parasite
References
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 (
(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 Metastatic adenocarcinoma with microfilaria of Metastatic squamous cell carcinoma with microfilaria of Metastatic adenocarcinoma with microfilaria of
Answer: a. Metastatic squamous cell carcinoma with microfilaria of
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 (
Table 1 elaborates the key differentiating features of commonly occurring sheathed filarial nematodes.
Key differentiating features of commonly occurring sheathed filarial nematodes.
Nematode | |||
---|---|---|---|
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
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
Filariasis is a vector borne public health issue and is endemic all over India. In India,
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.[
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
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
(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 [
Tuberculous bacilli are seen as a pink rod shaped slender bacilli on ZN stain [
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
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.[
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
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 [
(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).
What is the most likely interpretation of FNA using the Milan system? [
Non-neoplastic Atypia of undetermined significance Neoplasm: Benign Neoplasm: Salivary gland neoplasm of uncertain malignant potential (SUMP) Malignant.
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, “
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 “
An excisional biopsy was performed after 12 days of FNAC. On the basis of gross and microscopic findings, what is the diagnosis? [
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).
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.
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,
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.
a. “Pseudocribriform” architecture displaying false punched-out spaces and “Roman bridge” formation without invasion.
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.
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 [
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.
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.
d. Negative surgical margins and absence of stromal invasion.
Low-grade intraductal carcinoma (LG-IC) is a rare salivary gland malignant tumor.[
In 1996, Delgado
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.[ Intercalated duct-like (most common)[ Apocrine[ Mixed/hybrid Possibly oncocytic, but may be a variant of intercalated duct type.[
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.[
A systematic review performed by Giovacchini
Nakazawa
Kuo
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,[
Immunohistochemically, Giovacchini
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
References
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.
Q1. What is the interpretation/diagnosis?
Breast carcinoma Granulomatous mastitis (GM) in accessory axillary breast Fibrocystic change Phyllodes tumor.
b- GM in accessory axillary breast.
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 [
(a-c) Fine needle aspirate biopsy smear from left axillary swelling stained with papanicolaou and may-grunwald-giemsa stain.
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.
Phyllodes Mastitis Fibroadenoma
All of the above.
Tuberculous mastitis Sarcoidosis Both Galactocele.
Answers: Q2-a, Q3-d, Q4-c.
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.[
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
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 [
(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.[
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.[
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 [
(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
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.
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.[
The cytologic features of metastatic low-grade ESS have rarely been described in the literature since it is rarely subjected to fine-needle aspiration.[
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
Ronen
Mindiola-Romero
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.[
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
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) [
(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? Tuberculosis Non-Hodgkin’s lymphoma Ductal carcinoma. 1. a. 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.
Actinomycosis is a chronic infection caused by
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.[
2. c. Gram
3. a.
There is no conflict of interest in this paper.
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.
FNAC was done after proper consent as routine diagnostic test.
PAS- Periodic Acid Schiff
ZN- Ziehl Nelson
To ensure the integrity and highest quality of CytoJournal publications, the review process of this manuscript was conducted under a
References
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 [
(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).
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
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 [
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 [
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 [
Total thyroidectomy specimen with tumour mass showing solid-cystic areas.
Microscopy [
(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.
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
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.
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.
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.
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.
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.
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.
SS is an exceedingly rare malignant mesenchymal neoplasm accounting for about 10% of soft-tissue sarcomas.[
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.[
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.[
Pre-operative diagnosis based on FNAC is usually not supportive, although HPE may be contributory to some extent.[
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
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 [
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).
The correct cytological interpretation is
a. Atypical epithelial cells, suspicious for carcinoma.
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.
Q2. H&E stained sections are prepared from the tissue cores [
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 [
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
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.”
Acinar cell carcinoma (ACC) is a rare pancreatic malignancy that comprises about 1% of pancreatic neoplasia.[
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%.[
References
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 [
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).
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
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 [
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.[
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).
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).
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%.[
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
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.
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.
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.[
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).[
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 [
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.[
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.[
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 [
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 [
Answers for Question 2 through 5:
2. B
3. E
4. B
5. E
References
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 [
(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.[
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.[
Fibromatosis occurs almost at every site of the body.[
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.[
The mass was surgically excised. Grossly, it measured 10 cm in maximum dimension, and externally, it was skin-covered [
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.[
Keloid is a reactive condition resulting from excessive scar formation due to an aberrant healing process.[
Keloids are most frequent among African population and less common in Caucasians.[
Although histopathological features of keloid have been aptly described in the literature, cytomorphology of keloid is rarely reported in the literature.[
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.[
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.
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.
The authors have no conflicts of interest.
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.
The informed and written consent was obtained from the patient. The case was submitted without identifiers.
References
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
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).
Q1: What is your interpretation?
Ulcerated pilar cyst Pilomatrixoma Trichoepithelioma Proliferating trichilemmal tumor (PTT) Squamous cell carcinoma d
d. Proliferating trichilemmal tumor.
Explanation: The fine-needle aspiration cytology (FNAC) smears were moderately cellular and showed anucleate and nucleated squamous cells, basaloid cells [
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 [
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.[ 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.[ 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.[ 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.
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
Fine needle aspiration of hematolymphoid lesions of the thyroid: Onsite adequacy and ancillary testing
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 [
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).
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 [
Immunophenotyping by flow cytometry analysis [
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 [
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.[
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.[
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).[
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).[
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).[
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).[
Thyroid nodules are uncommon in the pediatric population as compared to adults and account for 0.2–2% of cases.[
Advances in ultrasonographic examinations have resulted in increased numbers of incidental thyroid gland lesions than previously reported.[
The differential diagnosis for thyroid lesion in children typically includes nodular goiter, lymphocytic thyroiditis, colloid cysts, follicular adenomas, degenerating nodules, and malignant thyroid nodules.[
Thymus is a primary lymphoid organ that plays an important role in the differentiation of T-cells.[
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 [
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[
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
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 [
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.[
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.[
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.[
A definitive diagnosis of scrotal calcinosis can be made only on the basis of histology.[
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.[
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).
ISC is rare and has a benign course.[
Pathogenesis still remains unknown and continues to be debated.[
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.[
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.[
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
A diagnostic challenge in a rare variant of invasive breast carcinoma – How far one can go
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
(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 [
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[ 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[ 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.[
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.[
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.[
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.
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 [
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.
Pure IMPC is a rare variant of IBC constituting 0.9–2%.[
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.[
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.[
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
Epithelioid cell granulomas in urine cytology smears: A diagnostic approach
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.
(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).
What is your interpretation?
Tuberculosis of urinary tract Urothelial changes with treatment effect Malignancy Cystitis cystica glandularis.
The correct cytopathological interpretation is (a) Urinary tract tuberculosis.
Three consecutive urine samples showed the presence of epithelioid cell granulomas and multinucleated giant cells along with reactive urothelial cells and inflammatory cells [
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 [
(a) Ultrasonography KUB and (b) computed tomography showing evidence of hydroureteronephrosis.
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.
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
Q4. c Fibrosis due to
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.
GUTB is a term coined by Wildbolz in 1937.[
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.[
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.[
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.[
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.[
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%.[
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.
The authors declare that they have no competing interest.
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.
As this is case without identifiers, our institution does not require approval from Institutional Review Board (or its equivalent).
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 –
RT PCR – Reverse transcription polymerase chain reaction
SOL – Space-occupying lesion
USG KUB – Ultrasonography for kidney, ureter, and urinary bladder
ZN – Ziehl–Neelsen.
To ensure the integrity and highest quality of CytoJournal publications, the review process of this manuscript was conducted under a
References
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
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].
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.
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 [
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.[
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.
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
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)[ |
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 [
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).
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.
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.
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.[
About 80% of pancreatic cystic lesions identified on imaging are non-neoplastic, including pseudocysts.[
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.[
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.[
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.[
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.[
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.[
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
Plasmacytoid cells in a thyroid aspirate – Look before you leap
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
(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
The correct answer is b. Non-Hodgkin’s lymphoma.
Thyroid FNA smears were highly cellular, showing predominantly discohesive plasmacytoid cells of variable sizes [
Cells expressing LCA immunoreactivity (ICC, ×400).
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.
Q1.b; Q2.d; Q3.c.
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.[
The presence of LGBs, cartwheel chromatin pattern, and Dutcher bodies can indicate malignant lymphoma and plasmacytoma.[
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
Cytologic evaluation of solitary thyroid nodule in a child
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
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).
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).
Cytology smears were cellular, comprising predominantly of dispersed cells and few loose cellular aggregates [
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: 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.[
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.[
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.[
Q4 (d) – All of the above. The correct answer is A. Immunocytochemically, MTC shows immunoreactivity for calcitonin, chromogranin, and synaptophysin with negativity for thyroglobulin.[
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%.[
The aspirates of MTC are cellular, comprising dispersed cells to loosely cohesive groups of plasmacytoid, spindle, and/or epithelioid cells.[
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
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 [
(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).
What is you interpretation?
Granulomatous lesion Olfactory neuroblastoma Metastatic nasopharyngeal carcinoma (NPC), undifferentiated type Metastasis from CNS tumor Malignant melanoma.
The correct cytopathological interpretation is:
C. Metastatic nasopharyngeal carcinoma (NPC), undifferentiated type.
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.[
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
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 [
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 [
(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).
<1 case/100,000 population 5–10 cases/100,000 population 15–20 cases/100,000 population 20–25 cases/100000 population.
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.
CK 5/6 CK 19 Pan CK CK 7.
Answers: Q1-a, Q2-b, Q3-d.
Q1– NPC is an uncommon tumor with annual incidence of <1 case/100,000 population.[
Q2– It has been shown in many studies that non-keratinizing NPC (NK-NPC), especially undifferentiated type has a strong association with EBV infection.[
Q3– NPC stains strongly for p63, panCK, and CK19. Immunohistochemical staining for CK 7 and 14 is negative.[
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.[
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.
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.
The authors declare that they have no competing interest.
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.
As this is a quiz case without identifiers, our institution does not require approval from Institutional Review Board (IRB).
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.
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
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
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).
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.
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 [
Histopathological examination revealed features of a well-differentiated squamous cell carcinoma (SCC) comprising almost 100% of the tumor area [
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).
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
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.
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.[
Macia
Cytologic diagnosis of SCCB has been cited in the literature as rare case reports only.[
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.[
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.[
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).[
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.[
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
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
(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).
What is your diagnosis?
Reactive mesothelial cells Metastatic adenocarcinoma Positive for malignancy, possibly myxoid chondrosarcoma Positive for malignancy, cannot characterize further.
The correct cytological interpretation is C. Positive for malignancy, possibly myxoid chondrosarcoma.
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.
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.[
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.[
EMC is a rare soft-tissue sarcoma, first described by Stout and Vernar,[
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.[
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
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[
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).
Q1: A; Q2: D; Q3: A.
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
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 [
(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×.
Echinococcus Cysticercus cellulosae Wuchereria bancrofti Enterobius vermicularis.
Answer
The correct cytological interpretation is
b. Cysticercus cellulosae.
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 [
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.[
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.[
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.
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.
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
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 [
(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.[
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.
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 [
(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.
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.[
EMH in Thalassemia usually occurs at liver, spleen, kidney, lymph nodes, and paravertebral region.[
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.[
Cytologically EMH composed of a combination of myeloid, erythroid, and megakaryocyte precursors. The erythroid precursors predominate in children, whereas megakaryocytes predominate in adults.[
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.[
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.[
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.[
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.[
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.
The authors declare that they have no competing interests.
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.
As this is case without identifiers, our institution does not require approval from institutional review board (IRB) (or its equivalent).
CT – Computed tomography
EMH – Extramedullary hematopoiesis
FNAC – Fine needle aspiration cytology
nRBC – Nucleated red blood cells
TM – Thalassemia Major
WBC – White blood cells.
To ensure the integrity and highest quality of CytoJournal publications, the review process of this manuscript was conducted under a
References
A small round blue cell tumor in urine: cytomorphology and differential diagnosis
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 [
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).
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).
The correct interpretation is E: Metastatic MCC.
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 [
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 [
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.
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.
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.[
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[
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.[
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.[
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.
MCC, previously known as trabecular carcinoma/sweat gland carcinoma/Toker’s tumor, was first described in 1972 by Toker.[
MCC is more common in males, with the most common primary site being head and neck. Heath
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.[
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
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 [
FNAC of the right iliac lymph node (Arrowhead-plexiform vasculature, Arrow- lipoblast), PAP, ×100 (original).
Angiomyxoma Myxofibrosarcoma Myxoid dermatofibrosarcoma protuberans Myxoid liposarcoma.
Question 1 answer – d-Myxoid liposarcoma.
Smears made from FNA of the right iliac lymph node showed tissue fragments with myxoid matrix and plexiform vascular network [
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 [
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 [
Hypercellular areas showing predominantly sheets of round to ovoid cells. (H and E, ×100) (original).
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).
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.
Vulvar sarcomas account for only 1–3% of all vulvar malignancies and the most frequent primary vulvar sarcoma is leiomyosarcoma.[
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[ |
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[ |
26 | Left labium majus et minor | 4 years | Bartholin cyst | 10 | Local excision followed by reexcision | NED+ 9 m |
Wu and Tarn[ |
45 | Right labium majus | 72 months | Lipoma | 7 | Local excision; reexcision of 6 cm recurrence 16 months later | NED, 28 m |
Schoolmeester |
34 | Left vulval mass | 11 months | Unknown | 11.7 | Local excision | NED, 28 m |
Baek |
33 | Bilateral perineum | 4 months | Unknown | Wide excision | NED, 2 yr | |
Kwak |
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.[
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 [
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 | ||||||
Angio |
Reproductive age (30 years) | Superficial and deep | Polypoidal, partly circumscribed |
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 | CD34 |
t(12;21) |
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 | Vimentin |
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 mode |
CD34 | t(17;22) |
Myxoid |
Middle to older | Deep soft tissue | Well-demarcated |
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) |
Multi |
Moderate | Plump, spindle or stellate cells having large atypical hyperchromatic nucleus | Pseudolipo |
Curvilinear, elongated blood vessels with perivascular condensation of tumor cells | Myxoid | High | MSA, SMA | Complex karyotype |
MLS | Young adults | Deep soft tissue | Well-circumscribed, multinodular |
Nodular | Mode-rate | Mixture of uniform round-oval cells and bland fusiform cells | +nt | Plexiform, branching | Myxoid | Rare | Vimentin |
t(12;16) |
LB: Lipoblast, RMS: Rhabdomyosarcoma, DFSP: Dermatofibrosarcoma protuberans, MFS: Myxofibrosarcoma, MLS: Myxoid liposarcoma, -nt: Absent, +nt: Present
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
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 [
(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.[
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.[
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.[
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.[
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
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 [
(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).
Plasma cell lesion