Context.—

Mesothelioma is an uncommon tumor that can be difficult to diagnose.

Objective.—

To provide updated, practical guidelines for the pathologic diagnosis of mesothelioma.

Data Sources.—

Pathologists involved in the International Mesothelioma Interest Group and others with expertise in mesothelioma contributed to this update. Reference material includes peer-reviewed publications and textbooks.

Conclusions.—

There was consensus opinion regarding guidelines for (1) histomorphologic diagnosis of mesothelial tumors, including distinction of epithelioid, biphasic, and sarcomatoid mesothelioma; recognition of morphologic variants and patterns; and recognition of common morphologic pitfalls; (2) molecular pathogenesis of mesothelioma; (3) application of immunohistochemical markers to establish mesothelial lineage and distinguish mesothelioma from common morphologic differentials; (4) application of ancillary studies to distinguish benign from malignant mesothelial proliferations, including BAP1 and MTAP immunostains; novel immunomarkers such as Merlin and p53; fluorescence in situ hybridization (FISH) for homozygous deletion of CDKN2A; and novel molecular assays; (5) practical recommendations for routine reporting of mesothelioma, including grading epithelioid mesothelioma and other prognostic parameters; (6) diagnosis of mesothelioma in situ; (7) cytologic diagnosis of mesothelioma, including use of immunostains and molecular assays; and (8) features of nonmalignant peritoneal mesothelial lesions.

Pathologic diagnosis of mesothelioma continues to evolve. These updated guidelines1  reflect the authors’ expert opinion, informed by a thorough review of relevant literature. The contents were discussed at the annual meeting of the International Mesothelioma Panel, with subsequent drafts circulated to obtain consensus among the authors. These guidelines are intended to offer a practical reference for the diagnostic pathologist, rather than a mandate.

Challenges in the morphologic classification of pleural and peritoneal mesothelioma are summarized. Morphologic hallmarks and pitfalls are reviewed, acknowledging that many pathologists lack access to some or all of the immunohistochemical and molecular assays discussed here. The role of immunostains to establish mesothelial lineage, separate benign from malignant mesothelial proliferations, and distinguish mesothelioma from other malignant mimics is presented. Given the large number of studies reported from different laboratories, sensitivity and specificity figures quoted in this article for different immunostains should be regarded as average figures obtained by literature review. The evolving role of diagnostic molecular assays is also addressed. Additional topics include recommendations regarding reporting the diagnosis of mesothelioma, the evolving concept of mesothelioma in situ, the role of cytopathology in diagnosis of mesothelioma, and features of nonmalignant peritoneal mesothelial lesions.

Approximately 85% to 90% of mesotheliomas arise in the pleura, with most of the remaining 10% to 15% affecting the peritoneum. Primary pericardial and paratesticular mesothelioma each account for roughly 1%, and considerations specific to these rare locations are not addressed. Location of the tumor (pleural versus peritoneal, or rarely pericardial, paratesticular, or at a metastatic site) and sex of the patient affect the differential diagnosis and thus the diagnostic approach. Regardless of site, a diagnosis of mesothelioma should always be based on compatible morphologic and immunohistochemical results obtained from an adequate tissue sample (typically a biopsy; less often an effusion, exfoliative, or fine-needle aspiration cytology specimen), in the context of appropriate clinical, radiographic, and (when available) surgical findings. A history of asbestos exposure should not be taken into consideration by the pathologist when confirming or excluding mesothelioma. Molecular studies might be necessary in a minority of cases.

When “mesothelioma” is diagnosed without further qualification, it is generally understood to mean diffuse mesothelioma, which represents 99% of pleural and peritoneal mesotheliomas and is characterized by disseminated involvement of the serosal-lined cavity. In contrast, localized mesothelioma (accounting for just 1% of cases) presents as a solitary, circumscribed pleural- or peritoneal-based mass, ranging from 0.5 to 20 cm, with negative effusion cytology. Localized and diffuse mesothelioma are indistinguishable under the microscope, with approximately the same distribution of epithelioid, biphasic, and sarcomatoid tumors and comparable molecular profiles.2,3  Nonetheless, this distinction is clinically important, as localized mesothelioma has a more favorable prognosis than diffuse mesothelioma. Correlation with clinical and radiographic findings is necessary in all cases of mesothelioma to differentiate a localized from a diffuse process.

The 2021 World Health Organization (WHO) classification of mesothelioma4  retains the 3 major histologic subtypes—epithelioid, biphasic, and sarcomatoid—but incorporates several architectural patterns and cytologic and stromal features that are prognostically significant. The diagnostic term mesothelioma is recommended, rather than malignant mesothelioma. To avoid confusion, the lesion previously termed well-differentiated papillary mesothelioma has been renamed well-differentiated papillary mesothelial tumor. As before, the term multicystic mesothelioma is discouraged, in favor of (multiloculated) peritoneal inclusion cyst.5  In short, the term mesothelioma now applies only to malignant tumors.

Epithelioid Mesothelioma

Epithelioid mesothelioma accounts for 60% to 70% of pleural and 80% to 90% of peritoneal mesotheliomas.6–9  These tumors comprise polygonal, oval, or cuboidal cells that often mimic nonneoplastic, reactive mesothelial cells. Epithelioid mesothelioma exhibits several generally familiar architectural patterns, including tubulopapillary (Figure 1), trabecular (Supplemental Figure 1, see the supplemental digital content file, containing 18 figures and 2 tables at https://meridian.allenpress.com/aplm in the November 2024 table of contents.), micropapillary (Figure 2), adenomatoid (Figure 3), and solid (Figure 4). The identified architectural patterns should be reported for each tumor in both biopsy and resection specimens, and in definitive resection specimens (ie, extended pleurectomy/decortication or extrapleural pneumonectomy); percentage representation (to the nearest 10%) of each pattern should also be reported.10 

Figure 1.

Epithelioid mesothelioma, tubulopapillary architecture (hematoxylin-eosin, original magnification ×100).

Figure 2.Epithelioid mesothelioma, micropapillary architecture (hematoxylin-eosin, original magnification ×100).

Figure 3.Epithelioid mesothelioma, adenomatoid architecture (hematoxylin-eosin, original magnification ×200).

Figure 4.Epithelioid mesothelioma, solid architecture, with necrosis (hematoxylin-eosin, original magnification ×100).

Figure 5.Desmoplastic mesothelioma (hematoxylin-eosin, original magnification ×100).

Figure 6.Transitional mesothelioma (hematoxylin-eosin, original magnification ×100).

Figure 1.

Epithelioid mesothelioma, tubulopapillary architecture (hematoxylin-eosin, original magnification ×100).

Figure 2.Epithelioid mesothelioma, micropapillary architecture (hematoxylin-eosin, original magnification ×100).

Figure 3.Epithelioid mesothelioma, adenomatoid architecture (hematoxylin-eosin, original magnification ×200).

Figure 4.Epithelioid mesothelioma, solid architecture, with necrosis (hematoxylin-eosin, original magnification ×100).

Figure 5.Desmoplastic mesothelioma (hematoxylin-eosin, original magnification ×100).

Figure 6.Transitional mesothelioma (hematoxylin-eosin, original magnification ×100).

Close modal

Tubulopapillary, trabecular, and adenomatoid patterns are associated with a more favorable prognosis. Conversely, any micropapillary component or at least 50% solid pattern is associated with worse prognosis.11  Micropapillary pattern correlates with a higher incidence of lymphatic invasion. Necrosis is seen in 30% of epithelioid mesotheliomas and is associated with a worse prognosis.12 

Variant cytologic and stromal features are recognized for epithelioid mesothelioma, and familiarity facilitates proper diagnosis. Those features of known prognostic significance should be reported when present.

Myxoid Stromal Features

Rare epithelioid mesotheliomas comprise clusters of mildly atypical tumor cells in a matrix of loose myxoid stroma (Supplemental Figure 2). Epithelioid mesotheliomas with at least 50% myxoid morphology and less than 50% solid growth pattern have a favorable prognosis.13,14 

Rhabdoid Cytologic Features

Rhabdoid cytologic features are prognostically unfavorable, defined by a variable proportion (15%–75%) of tumor cells morphologically similar to those of rhabdomyoblastic tumors, containing cytoplasmic globules that express cytokeratins but are negative for myogenin (Supplemental Figure 3).15 

Pleomorphic Cytologic Features

Epithelioid mesotheliomas with pleomorphic cytomorphology—defined by nuclear enlargement, hyperchromasia, prominent nucleoli, and (often) multinucleation, forming at least 10% of the tumor—behave like sarcomatoid and biphasic mesotheliomas.16,17  The 2021 WHO classification recommends that tumors with pleomorphic cytomorphology (Supplemental Figure 4) be classified as epithelioid, biphasic, or sarcomatoid, based on the remaining tumor cell morphology, although the presence of a pleomorphic component should be documented and its poor prognostic significance noted.4,18 

Lymphohistiocytoid Cytologic Features

Mesothelioma with lymphohistiocytoid features is defined by polygonal tumor cells morphologically similar to histiocytes, admixed with a marked lymphoid infiltrate (Supplemental Figure 5, A). Immunostains might be necessary to distinguish this entity from a nonneoplastic inflammatory process, lymphoepithelial carcinoma, or lymphoma. “Lymphohistiocytoid features” does not simply refer to markedly inflamed mesothelioma; the tumor cells must show histiocytoid morphology. Mesothelioma with lymphohistiocytoid features can be classified as epithelioid, biphasic, or sarcomatoid from the morphology of the non-lymphohistiocytoid component. Lymphohistiocytoid features in sarcomatoid mesothelioma impart a favorable prognosis, although prognostic significance is less clear for epithelioid tumors.19 

Clear Cell, Deciduoid, Signet Ring, Small Cell, and Adenoid Cystic Features

These features are not prognostically significant (Supplemental Figure 5, B; Supplemental Figure 6). However, familiarity with these morphologies facilitates distinction, respectively, from clear cell carcinomas, sarcoma, and melanoma20 ; florid deciduosis or deciduoid carcinomas21 ; signet ring cell adenocarcinomas of the lung and gastrointestinal tract22 ; small cell carcinomas, sarcomas, and lymphomas23 ; and adenoid cystic carcinoma. Note that mesotheliomas with small cell–like morphology do not show true neuroendocrine differentiation, nor do they stain with neuroendocrine markers, and use of the term small cell mesothelioma is discouraged, to avoid confusion with small cell carcinoma.

Sarcomatoid Mesothelioma

Sarcomatoid mesotheliomas account for 5% to 15% of pleural and less than 5% of peritoneal mesotheliomas.8,9,24  They are composed of infiltrating sheets of spindle cells with variable cytologic atypia (Supplemental Figure 7). Tumors can show necrosis, atypical mitoses, and/or heterologous (including rhabdomyosarcomatous, osteosarcomatous, and chondrosarcomatous) elements, which when extensive must be distinguished from true sarcomas.25  Sarcomatoid mesothelioma has a significantly poorer prognosis than epithelioid mesothelioma.12,24,26 

Desmoplastic Mesothelioma

Desmoplastic mesothelioma is a pattern of sarcomatoid mesothelioma characterized by a hypocellular population of bland spindle cells, arranged in a haphazard (patternless) fashion between bands of dense collagenous stroma (Figure 5). While the stroma is similar to that seen in pleural plaque, the tumor cells are arranged in a haphazard fashion as opposed to a linear arrangement usually parallel to the surface in the latter.27  Desmoplastic mesothelioma might not be suspected unless frankly sarcomatoid areas or areas of invasion into adipose tissue or lung are found. A diagnosis of “desmoplastic mesothelioma” requires desmoplastic features in at least 50% of a tumor in a definitive resection specimen. If desmoplastic morphology is seen in a smaller biopsy specimen, the diagnostic qualification “with desmoplastic features” is recommended.

Transitional Features

Transitional features in mesothelioma are an uncommon and recently described pattern, defined by sheetlike growth and cytomorphology intermediate between sarcomatoid and epithelioid tumor cells, with elongated but plump and cohesive cells (Figure 6). These were likely variably classified historically, but they are now regarded as a pattern of sarcomatoid morphology, as their prognosis and transcriptomic profile is similar to sarcomatoid mesotheliomas.28,29  Reticulin staining can be helpful in identifying the transitional pattern, as staining is present around individual cells as in sarcomatoid mesothelioma, in contrast to surrounding clusters of cells in the epithelioid subtype.29 

Biphasic Mesothelioma

Biphasic mesothelioma accounts for 15% to 30% of pleural and 10% to 20% of peritoneal mesotheliomas.6–9  These tumors contain malignant epithelioid and sarcomatoid components (Supplemental Figure 8), with transitional morphology regarded as sarcomatoid in this context (ie, if transitional features are seen in an otherwise epithelioid mesothelioma, a diagnosis of biphasic mesothelioma should be rendered). In a definitive resection specimen, at least 10% of each component must be present to render the diagnosis of biphasic mesothelioma. In biopsy specimens, biphasic mesothelioma should be diagnosed if malignant epithelioid and sarcomatoid components are present, regardless of the percentage of each component (ie, <10% epithelioid or sarcomatoid component does not preclude a diagnosis of biphasic mesothelioma in a biopsy—this change in the 2021 WHO classification4  reflects the typically higher percentage of sarcomatoid morphology seen in resection versus biopsy specimens).7,11,30  In peritoneal tumors, some evidence suggests that even focal (<10%) sarcomatoid morphology imparts a poorer prognosis than is typical of epithelioid peritoneal mesothelioma,31  so it is prudent to note even focal (<10%) sarcomatoid growth in resected peritoneal mesotheliomas.

The prognosis of biphasic mesothelioma is intermediate between that of pure epithelioid and pure sarcomatoid mesothelioma.8,32  While data are limited, there is some suggestion that biphasic mesotheliomas with predominant (>50%) sarcomatoid morphology have a poorer prognosis,33,34  so the percentage contribution of the epithelioid component and the sarcomatoid component should be reported in biopsies and resections of biphasic mesothelioma.

Historically, expert pathologists showed moderate interobserver agreement in diagnosis of biphasic mesothelioma, though improved agreement can be attained through application of strict diagnostic criteria.28,34,35  Epithelioid mesothelioma associated with a reactive spindled mesothelial population presents a challenging differential for biphasic mesothelioma, particularly when the spindled component lacks overtly atypical morphologic features. In this setting, immunohistochemical and molecular studies may help define the spindled mesothelial component as malignant (see Ancillary Studies in Diagnosis of Biphasic Mesothelioma below).

Although an exhaustive review of the molecular biology of mesothelioma is beyond the scope of these guidelines, a brief overview is provided as context for subsequent discussion of diagnostic molecular assays and immunostains. The genes most commonly affected by somatic mutation and copy number alteration in mesothelioma are BAP1, NF2, CDKN2A, TP53, LATS1/2, and SETD2, with mutation rates in particular genes varying by both tumor site and histotype.36,37  Gene fusions are rare in mesothelioma, though a subset of peritoneal mesotheliomas in young patients harbor ALK rearrangements or EWSR1::ATF1 or EWSR1/FUS::CREB fusions, while rare cases of EWSR1::YY1 fusion are reported in peritoneal mesothelioma in middle-aged patients.38–42 ALK rearrangements are very rarely detected in pleural mesothelioma.43,44 

BAP1

BRCA1-associated protein-1 (BAP1) is a tumor suppressor gene encoding a deubiquitylase with roles in cell cycle regulation, cellular differentiation, cell death, and DNA damage response.45 BAP1 is inactivated in approximately 60% of pleural and 70% of peritoneal mesotheliomas by missense, truncating, and splice site mutations; truncating fusion events; and copy number loss via chr 3p21.1 deletion. BAP1 mutations appear to be an early event in mesothelioma pathogenesis, representing the most common molecular alteration in reported cases of mesothelioma in situ (see Mesothelioma In Situ below). BAP1 alterations are found more often in epithelioid than in sarcomatoid tumors.46  Germline BAP1 mutations produce the BAP1 tumor predisposition syndrome (see Germline Predisposition to Mesothelioma below).

CDKN2A

CDKN2A (p16) resides on the chr 9p21 locus alongside CDKN2B (p15INK5), p14RF, and MTAP. Homozygous deletion of CDKN2A is found in approximately 70% of pleural mesotheliomas (including 90%–100% of sarcomatoid mesotheliomas and 40%–70% of epithelioid and biphasic types) but just 10% to 15% of peritoneal mesotheliomas.47–49 CDKN2A point mutations are exceptionally rare in mesothelioma.48  Like BAP1 alterations, CDKN2A deletions are an early event in mesothelioma pathogenesis and are occasionally detected in mesothelioma in situ (see Mesothelioma In Situ below). Some evidence suggests that CDKN2A deletion plays a role in evolution from epithelioid to biphasic mesothelioma in some cases.48  Approximately 75% to 90% of mesotheliomas with CDKN2A deletion show codeletion of the neighboring MTAP gene,47,49  which can therefore be used as an immunohistochemical surrogate for CDKN2A deletion.

NF2

NF2, located in the chromosomal region 22q12, encodes Merlin, a tumor suppressor protein in the Hippo signaling pathway.50 NF2 inactivation in mesothelioma is predominantly via truncating mutations or gene deletion, and is somewhat more common in biphasic and sarcomatoid (∼70%) than in epithelioid (∼40%) tumors.48 NF2 inactivation appears to be a fairly late event in mesothelioma pathogenesis, and intratumoral heterogeneity for NF2 alterations is common.51 

Reactive and malignant mesothelial proliferations can overlap morphologically with nonmesothelial lesions and malignancies. Establishing mesothelial lineage is a crucial early step in proper diagnosis of a serosal lesion. Although mesothelial cells show certain characteristic morphologic features, panels of “mesothelial” and “epithelial” immunostains are now almost universally applied in routine diagnosis to confirm the morphologic impression of mesothelial differentiation. Markers of specific epithelial lineages also play a role, depending on the clinical and morphologic differential diagnosis. Rarely, the differential for mesothelioma includes nonepithelial (eg, mesenchymal, hematolymphoid) tumors, to which the immunopanel must be tailored.

Before undertaking immunostains for evaluation of mesothelioma, the responsible laboratory should have performed a rigorous validation to determine ideal conditions for routine use in their hands. Immunostains should be interpreted with caution in minute biopsies, in those with crush artifact (which may induce false-positive or false-negative staining), and around the edges of biopsy specimens (which may show artifactual positive immunostaining). Careful attention should be paid to avoid misinterpretation of immunostaining in entrapped benign mesothelial or epithelial structures.

Broad-Spectrum Cytokeratin

Immunohistochemical stains for broad-spectrum cytokeratin (eg, pancytokeratin, AE1/AE3, CAM 5.2, CK OSCAR) are highly sensitive for mesothelioma, including sarcomatoid mesothelioma. In one large study, 93% of sarcomatoid mesotheliomas exhibited immunoreactivity for at least 1 cytokeratin; that percentage may be even higher if a cytokeratin cocktail is used, there is adequate sampling of the tumor, and the tissue is well fixed.24  For sarcomatoid neoplasms, cytokeratin positivity is additionally useful in excluding spindle cell sarcoma or melanoma,52  although rare sarcomas and melanomas can be positive for cytokeratins, and areas of heterologous differentiation in mesothelioma are often cytokeratin negative. Note that reactive mesothelial stroma is also keratin positive; keratin stain does not differentiate benign from malignant mesothelial proliferations.

Broad-spectrum cytokeratins are virtually 100% sensitive for epithelioid mesothelioma. If an epithelioid malignant neoplasm causing diffuse serosal thickening is negative for multiple broad-spectrum cytokeratins, other diagnoses should be considered, such as melanoma, epithelioid hemangioendothelioma or angiosarcoma, and lymphoma.

Occasional tumors do not stain with any marker. This often reflects artifact, such as overfixation in formalin, or alcohol fixation followed by antigen retrieval (commonly used for cytology specimens), so some knowledge about the fixative is important, as is proper laboratory validation for alcohol-fixed tissue.53–55  Assessment of internal controls is helpful. If needed, vimentin can be used to assess baseline immunoreactivity of the tissue.

Mesothelial and Epithelial Immunomarkers

The most common morphologic differential diagnosis for mesothelioma is carcinoma, and panels of mesothelial and epithelial immunomarkers are routinely used to establish mesothelial lineage and distinguish benign or malignant mesothelial proliferations from epithelial mimics. The specific markers used will depend on the differential diagnosis, and as noted above, nonepithelial (eg, mesenchymal, melanocytic, or hematolymphoid) tumors may occasionally enter the differential, requiring appropriate immunopanel modifications.

The best-characterized and most common mesothelial markers include calretinin (Supplemental Figure 9), CK5 or CK5/6 (Supplemental Figure 10), WT1 (Wilms tumor-1; Supplemental Figure 11), and podoplanin (D2-40) (Supplemental Figure 12). Each of these markers shows greater than 80% sensitivity for epithelioid mesothelial proliferations, with lower sensitivity for sarcomatoid proliferations.56,57  HEG1 is a promising mesothelial marker for epithelioid tumors, with similar sensitivity to other mesothelial markers, and possibly greater specificity in the differential with carcinoma of the lung.58  Importantly, none of these markers is entirely specific for mesothelial origin, and all can be positive (usually focal, though sometimes diffuse) in a subset of carcinomas.56  Further, different mesothelial immunostains show different patterns of positive staining. For calretinin, combined cytoplasmic and nuclear staining is typically present in mesothelial cells. For WT1, only nuclear staining is considered positive, and cytoplasmic-only staining should be disregarded. CK5/6 is cytoplasmic, and D2-40 and HEG1 show membranous staining. While there is no validated standard for the percentage tumor cell staining required to be called “positive,” using 10% seems like a reasonable minimum.

The most common and generally most reliable epithelial markers are claudin-4 (Supplemental Figure 13), MOC-31 (Supplemental Figure 14), and Ber-EP4, all of which show membranous staining. Of note, claudin-4 may occasionally show dotlike cytoplasmic reactivity, which should be interpreted as negative. A variety of older markers (including CEA [carcinoembryonic antigen], CD15 [LeuM1], BG-8, and B72.3) also remain reliable options. When properly validated, each of these markers is greater than 80% sensitive for epithelial lineage (albeit less sensitive for sarcomatoid carcinoma) and greater than 80% specific in distinction from mesothelial tissues.56,57  Note that diffuse MOC-31 and Ber-EP4 expression are more specific for epithelial lineage than patchy staining, as ∼10% to 15% of mesotheliomas show patchy MOC-31 or Ber-EP4 staining.56,59 

Since none of these markers are perfectly sensitive or specific, it is recommended that, in addition to broad-spectrum cytokeratin, 2 mesothelial and 2 epithelial markers be included in a first-line immunopanel to establish mesothelial lineage. If results are concordant, the diagnosis can be considered established. If discordant, the immunopanel can be expanded for a second round of staining, with additional antibodies selected according to the differential diagnosis. A different tissue block can also be stained, if available. Given the range of reliable options and the likelihood of interlaboratory variation, no specific first-line antibody panel is recommended. Instead, each laboratory should test staining conditions for the antibodies of choice, ideally verifying sensitivity and specificity of at least 80% with appropriate controls.

Emerging evidence suggests that the novel mesothelial marker HEG1 and the epithelial marker claudin-4 may be sufficiently sensitive and specific to be used as a 2-marker panel to distinguish epithelioid mesothelioma from non–small cell lung carcinoma.58  In the authors’ experience, claudin-4 is sufficiently reliable to serve as the sole epithelial marker in most differentials59  (though claudin-4 is not expressed in proximal renal tubules or hepatocytes and is therefore not highly sensitive for renal cell or hepatocellular carcinoma). Despite its high sensitivity, HEG1 is not yet in widespread clinical use, and its expression in 50% of serous ovarian cancers and 100% of thyroid cancers limits its application. At present, it seems prudent to continue using immunopanels to establish mesothelial lineage.59–62 

Markers Useful in Specific Differentials

In addition to the broad-spectrum epithelial markers discussed above, immunomarkers specific to particular types of carcinoma are useful in certain differential diagnoses.

Immunohistochemistry in Diagnosis of Epithelioid Mesothelioma

Tables 1 and 2 list markers that are useful in distinguishing epithelioid pleural mesothelioma from adenocarcinoma and squamous cell carcinoma of the lung, respectively. TTF-1 (thyroid transcription factor-1; 8G73/1 DAKO clone) and Napsin A are highly specific for lung adenocarcinoma in the differential with epithelioid mesothelioma.63,64  WT1 is expressed in just 2% of squamous cell carcinomas and is therefore highly specific for epithelioid mesothelioma in this differential,65  while claudin-4 and p40 (and, to a lesser degree, p63) are specific for squamous cell carcinoma in this setting.65–69  p40 also assists in distinguishing squamous cell carcinoma from adenocarcinoma.70 

Table 1.

Immunohistochemical Markers Used in the Differential Diagnosis of Pleural Epithelioid Mesothelioma Versus Lung Adenocarcinomaa

Immunohistochemical Markers Used in the Differential Diagnosis of Pleural Epithelioid Mesothelioma Versus Lung Adenocarcinomaa
Immunohistochemical Markers Used in the Differential Diagnosis of Pleural Epithelioid Mesothelioma Versus Lung Adenocarcinomaa
Table 2.

Immunohistochemical Markers Used in the Differential Diagnosis of Pleural Epithelioid Mesothelioma Versus Squamous Cell Carcinoma of the Lunga

Immunohistochemical Markers Used in the Differential Diagnosis of Pleural Epithelioid Mesothelioma Versus Squamous Cell Carcinoma of the Lunga
Immunohistochemical Markers Used in the Differential Diagnosis of Pleural Epithelioid Mesothelioma Versus Squamous Cell Carcinoma of the Lunga

Because most breast carcinomas express estrogen receptor, gross cystic disease fluid protein-15, and/or mammaglobin, these markers are often useful in distinguishing metastatic breast carcinoma and mesothelioma.71  Calretinin and CK5/6 can be positive in high-grade basal-type breast carcinomas, which may also be negative for estrogen and progesterone receptor.72,73  SOX10 expression favors breast cancer in this scenario. One-third to one-half of epithelioid mesotheliomas are positive for GATA3, limiting its usefulness in this scenario.71,74 

Supplemental Table 1 lists markers that are considered useful in distinguishing mesothelioma and metastatic renal cell carcinoma. Because of their sensitivity and specificity, calretinin, D2-40, and CK5/6 are the best mesothelial markers in this context.75  A panel of epithelial markers may be necessary, as claudin-4, MOC-31, and Ber-EP4 are expressed in 90%, 40%, and 40% of renal cell carcinomas, respectively. Carbonic anhydrase IX is expressed in virtually all epithelioid pleural mesotheliomas and therefore not useful to distinguish it from renal cell carcinomas.76  PAX8 or PAX2 can be very useful, as these are expressed in most renal cell carcinomas but not in pleural mesotheliomas,76–78  though PAX8 (using both polyclonal and monoclonal antibodies) is positive in ∼15% of peritoneal mesotheliomas.79  The sensitivity and specificity of renal cell carcinoma (RCC) marker and CD15 for renal cell carcinoma are not high.76  Most renal cell carcinomas express CD10, but half of epithelioid mesotheliomas are also positive.80 

Serous ovarian carcinomas are virtually always positive for WT1 and may be positive for HEG1 (50%), CK5/6 (∼30%), and D2-40 (∼20%).56,81  Conversely, as noted above, ∼15% of peritoneal mesothelioma are PAX8 positive, more often in women (25%) than men (5%).79,82  Estrogen and progesterone receptor expression is rare in peritoneal mesothelioma (7% and 2%, respectively) (Table 3).82  Strong diffuse p53 (ie, >80% staining throughout tumor) does not exclude peritoneal mesothelioma, as 15% harbor TP53 mutation.83 

Table 3.

Peritoneal Mesothelioma Versus Serous Ovarian Carcinomaa

Peritoneal Mesothelioma Versus Serous Ovarian Carcinomaa
Peritoneal Mesothelioma Versus Serous Ovarian Carcinomaa

Adenocarcinomas of the gastrointestinal tract (Supplemental Table 2) and prostate can be distinguished from epithelioid mesotheliomas by the demonstration of CDX284  and prostate-specific antigen (and more recently NKX3.1),85  respectively. SATB2 (a marker of colorectal adenocarcinoma and osteosarcoma) is typically negative in mesothelioma.86 

Immunohistochemistry in Diagnosis of Sarcomatoid Mesothelioma

The role of broad-spectrum cytokeratin immunostains in the diagnosis of sarcomatoid mesothelioma is discussed above. In a cytokeratin-positive sarcomatoid malignancy, distinguishing sarcomatoid mesothelioma from sarcomatoid carcinoma requires a panel of immunomarkers, as sarcomatoid mesotheliomas are often negative for 1 or more mesothelial markers, and none of these markers is entirely specific. D2-40 and calretinin are each expressed in 50% to 60% of sarcomatoid mesotheliomas,24,87–89  though staining may be focal, and specificity is limited, as D2-40 and calretinin each stain ∼20% to 25% of sarcomatoid lung carcinomas. D2-40 reactivity in entrapped lymphatics or reactive mesothelial elements is a potential pitfall. Claudin-4, MOC31, and BerEP4 also show low sensitivity for sarcomatoid areas of carcinomas—approximately 33%, 38%, and 23%, respectively, across studies.59  Positive staining for TTF-1, Napsin A, or p40/p63 supports a diagnosis of sarcomatoid lung carcinoma. Diffuse GATA3 expression is seen in 70% of sarcomatoid mesotheliomas, but focal GATA3 expression can also be seen in sarcomatoid lung carcinoma,90–92  while 29% of sarcomatoid urothelial carcinomas and 50% of sarcomatoid (metaplastic) breast carcinomas express GATA3. PAX8 is positive in 44% to 69% of sarcomatoid renal cell carcinomas but generally negative in sarcomatoid mesothelioma.93  Conversely, sarcomatoid renal cell carcinoma is reportedly negative for CK5/6 and calretinin, though CK5/6 is particularly limited by its low sensitivity for sarcomatoid mesothelioma.75 

Synovial sarcoma is characteristically cytokeratin positive, and angiosarcoma and melanoma may also express cytokeratins. The diagnosis of synovial sarcoma can be confirmed with mutation-specific SS18-SSX and SSX-C-terminus immunostains, or with molecular confirmation of the distinctive X;18 translocation. Immunohistochemistry for TLE-1 can be used but is less specific.94 

After extensive workup and with appropriate clinical and radiologic features, 5% to 10% of sarcomatoid mesotheliomas are cytokeratin negative.25,89,95,96  In the absence of convincing cytokeratin positivity, positive calretinin and/or D2-40 staining should not be interpreted as evidence of mesothelial differentiation, as these markers are variably positive in some sarcomas (including synovial sarcoma, malignant peripheral nerve sheath tumor, and angiosarcoma),87,97  for which additional immunohistochemical markers would be warranted. The expanded differential might include epithelioid hemangioendothelioma, angiosarcoma, liposarcoma, myogenic or neurogenic sarcoma, undifferentiated pleomorphic sarcoma, malignant solitary fibrous tumor, melanoma, and histiocytic sarcomas. Morphology and clinical context should guide the differential and selection of appropriate immunohistochemical stains. Note that muscle-specific actin (HHF-35) and α-smooth muscle actin are often positive (occasionally diffusely) in sarcomatoid mesothelioma,98  though desmin expression is quite rare in sarcomatoid mesothelioma.98,99  Rare tumors cause diffuse pleural thickening, show extensive heterologous differentiation (eg, osteosarcomatous), and are negative for cytokeratin and mesothelial markers. After exclusion of a separate primary, these can be regarded as consistent with mesothelioma.

Histochemical Stains for Cytoplasmic Mucin

In current practice, histochemical staining is rarely used by experienced practitioners to distinguish mesothelioma from carcinoma (eg, in tumors expressing contradictory immunohistochemical markers). The cytoplasmic epithelial mucin in adenocarcinomas is positive by periodic acid–Schiff after diastase digestion (PAS-D) or by Alcian blue after hyaluronidase treatment. In contrast, cytoplasmic vacuoles in mesothelioma are generally negative by PAS-D, and the cytoplasmic hyaluronic acid in mesotheliomas stains positively with Alcian blue but is digestible by hyaluronidase.100  PAS-D can be positive in hyaluronidase crystals. Mucicarmine can be positive in mesothelioma or adenocarcinoma and is not recommended for this distinction.

Electron Microscopy

The ultrastructural features of mesothelioma are well described.101  However, the advent of novel immunomarkers has reduced the role of electron microscopy in routine diagnosis. Electron microscopy occasionally helps establish a diagnosis of mesothelioma when immunohistochemistry is equivocal, though tumors without diagnostic morphologic and immunophenotypic features of mesothelioma frequently lack specific ultrastructural findings, as well.102,103  Formalin-fixed material retrieved from a paraffin block may be satisfactory, as microvilli and tonofilament bundles tend to be preserved. As routine diagnostic experience with electron microscopy wanes, such cases are likely best referred to subspecialists with expertise in this domain.

Separating benign from malignant mesothelial proliferations requires certainty that the process is mesothelial (see Establishing Mesothelial Lineage above). The diagnostic approach used when distinguishing reactive mesothelial hyperplasia from epithelioid mesothelioma differs from that used when distinguishing fibrous pleuritis from desmoplastic mesothelioma.104  While morphology is paramount, a supportive immunophenotype is necessary for a definitive diagnosis of mesothelioma. The role of molecular studies in routine diagnosis is also evolving.

Reactive Mesothelial Hyperplasia Versus Epithelioid Mesothelioma

Reactive mesothelial proliferations may mimic mesothelioma, as they can show high cellularity, numerous mitoses, cytologic atypia, necrosis, papillary formations, and mesothelial entrapment within fibrosis, mimicking invasion (Figure 7).104  Morphologic features that help distinguish reactive mesothelial hyperplasia from mesothelioma are summarized in Table 4.

Figure 7.

Reactive mesothelial hyperplasia within fibrous tissue mimicking invasion (hematoxylin-eosin, original magnification ×100).

Figure 8.Desmoplastic mesothelioma. A, Bland-appearing spindle cells with haphazard growth. B, In a different focus, keratin highlights infiltration into fat (hematoxylin-eosin, original magnification ×100 [A]; original magnification ×100 [B]).

Figure 9.“Fake fat.” A, Fake fat in a pleural biopsy specimen from a patient with effusion and fibrosis. B, Keratin AE1/AE3 highlights horizontal, keratin-positive, reactive spindle cells around fake fat. C, S100 is negative in fake fat (hematoxylin-eosin, original magnification ×40 [A]; original magnifications ×100 [B] and ×200 [C]).

Figure 7.

Reactive mesothelial hyperplasia within fibrous tissue mimicking invasion (hematoxylin-eosin, original magnification ×100).

Figure 8.Desmoplastic mesothelioma. A, Bland-appearing spindle cells with haphazard growth. B, In a different focus, keratin highlights infiltration into fat (hematoxylin-eosin, original magnification ×100 [A]; original magnification ×100 [B]).

Figure 9.“Fake fat.” A, Fake fat in a pleural biopsy specimen from a patient with effusion and fibrosis. B, Keratin AE1/AE3 highlights horizontal, keratin-positive, reactive spindle cells around fake fat. C, S100 is negative in fake fat (hematoxylin-eosin, original magnification ×40 [A]; original magnifications ×100 [B] and ×200 [C]).

Close modal
Table 4.

Reactive Versus Malignant Mesothelial Proliferations

Reactive Versus Malignant Mesothelial Proliferations
Reactive Versus Malignant Mesothelial Proliferations

Demonstration of tissue invasion is a key feature in diagnosis of mesothelioma (Supplemental Figure 15). Invasion by mesothelioma is often subtle, involving only a few layers of collagenous tissue subjacent to the mesothelial space and eliciting no obvious desmoplastic reaction. Invasion may be highlighted with immunostains, such as pancytokeratin or calretinin. However, inflammatory pleural processes can entrap mesothelial cells in granulation tissue deep to the pleura, typically arranged parallel to the pleural surface. Tubular collections of reactive mesothelial cells may also be seen, again parallel to the pleural surface. These patterns do not connote malignancy. Diagnosis is best performed in a well-oriented specimen (ie, cut perpendicularly to the pleural surface, including the full thickness of the pleura with adjacent adipose tissue, skeletal muscle, and/or lung parenchyma), as a tangential section (ie, taken parallel to the pleural surface) can give a false impression of a full-thickness mesothelial proliferation.

When a substantial amount of solid, malignant tumor (ie, tumor nodule[s]) with histologic features of mesothelioma is identified, the presence of invasion is not required for diagnosis. Tumor necrosis is also a feature of malignancy.

Fibrous Pleuritis Versus Sarcomatoid or Desmoplastic Mesothelioma

Identification of malignant features in desmoplastic mesothelioma requires adequate tissue, and large surgical biopsy samples are generally (but not always) needed. Features separating fibrous pleuritis from desmoplastic mesothelioma are shown in Table 5.

Table 5.

Fibrous Pleuritis Versus Sarcomatoid or Desmoplastic Mesothelioma

Fibrous Pleuritis Versus Sarcomatoid or Desmoplastic Mesothelioma
Fibrous Pleuritis Versus Sarcomatoid or Desmoplastic Mesothelioma

Fibrous pleuritis tends to show a uniformity of growth, with regular sheets and sweeping parallel fascicles of bland spindle cells that respect mesothelial boundaries. In contrast, sarcomatoid or desmoplastic mesothelioma shows disorganized growth, haphazardly intersecting fascicles, and expansile nodules of varying sizes with abrupt demarcation and changes in cellularity between nodules and their surrounding tissue (Figure 8, A). These different patterns can be highlighted by a broad-spectrum cytokeratin immunostain. Note that reactive mesothelial stroma is also keratin positive; keratin stain does not differentiate benign from malignant mesothelial proliferations; however, it can show the pattern of growth.

Stromal invasion is often more difficult to recognize in spindle cell than in epithelioid proliferations, as the invasive malignant cells in the former are often deceptively bland, resembling fibroblasts. Broad-spectrum cytokeratin staining is invaluable in highlighting bland, cytokeratin-positive malignant cells in areas where they would not normally be present (eg, adipose tissue, skeletal muscle, or lung or other visceral tissue in the pleura or peritoneum) (Figure 8, B). Although identification of invasion is often straightforward with the aid of broad-spectrum cytokeratin staining, fatlike spaces (termed fake fat) can be encountered in organizing pleuritis, probably reflecting artifactual changes in dense, fibrous connective tissue (Figure 9, A).105  In such cases, horizontally oriented, cytokeratin-positive cells may be seen around the fatlike spaces (Figure 9, B). In addition, S100 protein, laminin, and collagen IV are usually positive in true adipose tissue and can help distinguish it from “fake fat,” which is negative for all 3 markers (Figure 9, C).

Immunohistochemical and Molecular Studies

In small biopsy specimens, morphology alone may be inadequate for a definitive diagnosis of malignancy. Many immunostains previously purported to distinguish benign from malignant mesothelial proliferations—including GLUT1, IMP3, desmin, and epithelial membrane antigen (EMA)—are of little diagnostic value in individual cases. However, loss of nuclear BAP1 by immunohistochemistry, loss of cytoplasmic methylthioadenosine phosphorylase (MTAP) by immunohistochemistry, and homozygous deletion of CDKN2A by FISH, though not present in all cases of mesothelioma, are by definition never found in benign mesothelium (Supplemental Figures 16 and 17).48,106–113  Both BAP1 and MTAP immunostains must be interpreted in the presence of a positive internal control, typically intratumoral inflammatory or stromal cells. These 3 techniques are very useful and can be applied in an algorithmic fashion in both tissue sections and cell block preparations (Figure 10).

Figure 10.

Algorithm for tissue diagnosis of mesothelial proliferations. An immunopanel of 2 epithelial and 2 mesothelial markers is generally advisable for confirming mesothelial lineage. Abbreviations: CK, cytokeratin; D2-40, podoplanin; FISH, fluorescence in situ hybridization; IHC, immunohistochemistry; WT1, Wilms tumor-1.

Figure 10.

Algorithm for tissue diagnosis of mesothelial proliferations. An immunopanel of 2 epithelial and 2 mesothelial markers is generally advisable for confirming mesothelial lineage. Abbreviations: CK, cytokeratin; D2-40, podoplanin; FISH, fluorescence in situ hybridization; IHC, immunohistochemistry; WT1, Wilms tumor-1.

Close modal

Sensitivities of BAP1 immunohistochemistry, MTAP immunohistochemistry, and CDKN2A FISH depend on both tumor histologic subtype and primary site. Loss of nuclear BAP1 staining is seen in 60% to 70% of epithelioid pleural mesotheliomas but just 20% of sarcomatoid tumors.48,107,112,114  Conversely, CDKN2A deletion and corresponding loss of cytoplasmic MTAP staining are seen in 60% and 40% of epithelioid pleural mesotheliomas, respectively, compared to 93% and 75%, respectively, of sarcomatoid pleural mesotheliomas.107,115,116  Recent data indicate that MTAP immunohistochemistry may be significantly less sensitive than CDKN2A FISH in desmoplastic mesothelioma, given its scant cytoplasm, highlighting the continued role of CDKN2A FISH in clinical diagnosis.116 

By primary site, BAP1 loss is seen in 50% to 60% of pleural and 60% to 70% of peritoneal mesotheliomas. Conversely, MTAP loss is seen in 50% of pleural compared to just 5% to 10% of peritoneal mesotheliomas, reflecting the different proportions of histologic subtypes and relative rates of underlying CDKN2A deletion at these sites.48,83 

BAP1 loss can also support a diagnosis of mesothelioma in the differential with certain carcinomas. In particular, BAP1 loss is seen in less than 1% of lung cancers and serous ovarian carcinomas.117,118  However, BAP1 loss is not specific for mesothelioma in isolation or in all contexts, as it may be seen in ∼15% of clear cell renal cell carcinomas and in subsets of intrahepatic cholangiocarcinoma, thymic carcinoma, and melanoma. MTAP loss does not help distinguish mesothelioma from other malignancies in any reported context.

p53 immunostaining has a controversial history in diagnosis of mesothelioma, largely because of prior misunderstanding about wild-type versus aberrant staining, but recent data indicate that strong diffuse p53 immunostaining (ie, in ≥80% of tumor cells) can be seen in mesothelioma but not in reactive mesothelial proliferations. Sensitivity is limited, with strong diffuse p53 staining seen in only ∼10% to 15% of pleural mesotheliomas, including rare cases in which this might be the only immunohistochemical evidence for malignancy.48,119  There is conflicting data on the specificity of null-pattern (ie, complete absence of) p53 staining for underlying TP53 mutation.

Homozygous or heterozygous NF2 deletion (detected by FISH or molecular sequencing) is specific for mesothelioma in the differential with reactive mesothelial proliferation.120  In recent reports, immunohistochemical loss of the NF2 protein product, Merlin, correlates strongly with underlying NF2 mutation, and Merlin loss has been detected in 40% to 50% of pleural mesotheliomas.48,121  Preliminary results are promising, but full endorsement of Merlin immunohistochemistry for this application awaits additional data.

Ancillary Studies in Diagnosis of Biphasic Mesothelioma

Morphology alone cannot always reliably distinguish biphasic mesothelioma from epithelioid mesothelioma associated with a reactive spindled mesothelial population. Immunohistochemical and molecular studies can attempt to establish a clonal relationship between the epithelioid and spindled populations. Although studies using this approach have shown variable results,34,122–124  recent data show strong concordance for BAP1 staining pattern (ie, lost versus retained) in the epithelioid and sarcomatoid components of biphasic mesothelioma. In contrast, MTAP staining is more often discordant, with retained expression in the epithelioid component and loss in the sarcomatoid component.48  At present, it is recommended that BAP1 or MTAP loss (or CDKN2A deletion by FISH) in the spindle cell component of a biphasic mesothelial proliferation be regarded as evidence of malignancy (ie, supporting diagnosis of biphasic mesothelioma). Conversely, if BAP1 or MTAP loss is confined to the epithelioid component (ie, retained expression in the spindled component), a diagnosis of biphasic mesothelioma should be made only if the spindled population shows unequivocal morphologic features of malignancy.

Molecular Sequencing in Routine Diagnosis and Management

Tumor molecular profiling with large next-generation sequencing panels has the potential to provide diagnostic, prognostic, and therapeutic information in a single assay. In one study of resection specimens, a 447-gene next-generation sequencing panel showed 95% sensitivity for diagnosis of mesothelioma.48  At present, routine genomic sequencing of mesotheliomas is performed only in select referral or academic centers, and it is not currently recommended for routine clinical use. Immunohistochemical studies remain the ancillary assay of choice, with targeted molecular studies (eg, CDKN2A FISH, ALK FISH, sequencing) in select cases.

The International Collaboration on Cancer Reporting has recently published a 3rd edition to their guidelines for reporting mesothelioma, which provide a valuable resource for routine practice.125 

Staging Pleural Mesothelioma

The Union for International Cancer Control and American Joint Committee on Cancer (AJCC) Cancer Staging Manual, 8th edition,126  represents the most widely applied TNM system and should be reported for all pleural mesotheliomas resected via extended pleurectomy/decortication or extrapleural pneumonectomy (now rarely performed). The TNM staging system for pleural mesothelioma evaluates resectability but is generally not a good predictor of prognosis. Importantly, the AJCC 8th edition does not include mesothelioma in situ. There is no consensus TNM staging for peritoneal, pericardial, or paratesticular mesothelioma.

Pathologic Predictors of Prognosis and Therapy Responsiveness

The 2021 WHO classification4  also recognizes a variety of important pathologic factors beyond tumor histologic subtype (ie, favorable prognosis for epithelioid, intermediate prognosis for biphasic, and poor prognosis for sarcomatoid tumors).10,127  As noted above, certain architectural, cytologic, and stromal features are linked to prognosis, and pathologists should routinely report this information.11  In brief, prognostically favorable morphologic findings include tubulopapillary, trabecular, and adenomatoid architecture; low nuclear grade; high tumor-associated immune microenvironment (eg, lymphohistiocytoid cytologic features); and myxoid-rich stromal matrix.12,13,128  Conversely, adverse prognosis is associated with any micropapillary or greater than 50% solid architecture; high nuclear grade; rhabdoid, pleomorphic, transitional, or desmoplastic morphology; and necrosis.15–17,29 

Grading Epithelioid Mesothelioma

Several studies have validated a 3-tiered grading system for epithelioid mesothelioma of the pleura and peritoneum, based on mitotic activity and nuclear atypia.8,12,128  After clinician input at a multidisciplinary meeting of mesothelioma experts, necrosis was added to mitotic activity and nuclear atypia to create a 2-tiered grading system (ie, low versus high grade), which better facilitates clinical decision-making (Table 6). The prognostic significance of the 2-tiered system has been validated in a large series of pleural mesothelioma,11,26  though its applicability to peritoneal mesothelioma remains unclear.8  When assigning nuclear grade, tumor foci with the highest-grade features should be used. The current 2021 WHO classification recommends routine reporting according to this 2-tiered grading system for biopsies and resections of epithelioid diffuse pleural mesothelioma.4 

Table 6.

Grading of Epithelioid Pleural Mesothelioma

Grading of Epithelioid Pleural Mesothelioma
Grading of Epithelioid Pleural Mesothelioma

Molecular Prognostic Factors

Homozygous deletion of CDKN2A and MTAP loss by immunohistochemistry both portend poor prognosis (shorter overall and disease-free interval) among mesothelioma cases.36,129,130  In contrast, loss of nuclear BAP1 by immunohistochemistry is a favorable prognostic marker, at least partly reflecting the significantly improved prognosis and treatment responsiveness in patients with germline BAP1 mutation.131,132  Rates of CDKN2A deletion, MTAP loss, and BAP1 correlate with tumor histologic subtype and primary site (see Distinguishing Benign Versus Malignant Mesothelial Proliferations above).

Germline Predisposition to Mesothelioma

Germline testing should now be considered for all patients with mesothelioma, as it affords improved response to platinum-based chemotherapy and relatively favorable prognosis (despite advanced stage at presentation), potential access to novel therapies, and genetic counseling for the patient and family, which is relevant to surveillance for other tumors.132–134 

Genomic profiling indicates that at least 12% of mesotheliomas arise in carriers of pathogenic germline mutations.45,132,133,135 BAP1 germline mutations (which cannot be distinguished from somatic mutations by BAP1 immunohistochemistry) account for approximately half of such cases. The prevalence of germline predisposition is higher in younger patients (ie, germline mutations identified in >50% of patients with mesothelioma and younger than 50 years134 ), in those with peritoneal disease, in tumors with low-grade epithelioid morphology and a high tumor immune response, in patients with longer overall survival (median survival >5 years135 ), and in those with a personal or family history of multiple cancers (especially melanoma, clear cell renal cell carcinoma, and breast cancer).132 

Targeted Therapies

Pathogenic germline mutations in patients with mesothelioma most commonly affect DNA damage repair pathways, which serve as a potential therapeutic target for poly (ADP-ribose) polymerase (PARP) inhibitors.136  Use of PARP inhibitors alone or combined with platinum-based regimens in mesotheliomas with germline homologous recombination defects is under evaluation.

US Food and Drug Administration approval for combined nivolumab and ipilimumab in untreated unresectable diffuse pleural mesothelioma followed publication of the CheckMate 743 trial.137  The benefits are primarily observed among patients with nonepithelioid disease, which is typically most refractory to conventional chemotherapy. By immunohistochemistry, PD-L1 is positive (>1% tumor cell staining) in 10% to 49% of epithelioid, 9% to 67% of biphasic, and 22% to 100% of sarcomatoid mesotheliomas,138–140  with some variability between PD-L1 antibody clones.139,140  Routine PD-L1 immunostaining is not currently indicated for mesothelioma.

ALK rearrangements can be identified by immunohistochemistry or molecular testing in a small number of mesotheliomas, with a predilection for children and young adults, women, and peritoneal tumors.40,141,142 ALK rearrangement appears mutually exclusive with other genetic events commonly observed in mesothelioma. ALK-fusion–positive peritoneal mesotheliomas afford a small group of patients access to novel targeted treatment with tyrosine kinase inhibitors, with reportedly dramatic treatment response.143  There is no formal guideline on screening for ALK rearrangement in mesothelioma, but given its clinical implications, it appears reasonable to perform ALK immunohistochemistry in young patients and patients with peritoneal tumors, particularly if other molecular alterations (eg, BAP1 loss, MTAP loss, Merlin loss) are not detected.

Mesothelioma in situ (MIS) as the noninvasive precursor to diffuse mesothelioma was initially proposed in 1992.144  The morphology of MIS is now recognized as variable and includes flat or cuboidal cells with or without cytologic atypia, small or complex papillary proliferations, or small surface nodules with moderate to severe cytologic atypia. Invasion is absent by definition, and there must be no clinically or radiographically identifiable mass lesion or diffuse process. MIS cannot be diagnosed by morphology alone, and immunohistochemical loss of nuclear BAP1 and/or demonstration of CDKN2A homozygous deletion (by FISH or by MTAP immunohistochemistry) must also be demonstrated on a rigorously validated assay with appropriate controls (Figure 11, A through D).47,111,112,115,145–147 

Figure 11.

Mesothelioma in situ. A, Papillary proliferation lined by a flat, cytomorphologically banal mesothelium, with (B) BAP1 loss. C, A separate case shows flat mesothelial lining with (D) BAP1 loss (hematoxylin-eosin, original magnifications ×100 [A and C] and ×200 [B]; original magnification ×100 [D]).

Figure 12.Mesothelioma, cytology preparation. Features of mesothelioma include (A) cell-in-cell arrangement and (B) a hump at the cell periphery (Papanicolaou, original magnification ×1000 [A and B]).

Figure 13.Well-differentiated papillary mesothelial tumor (hematoxylin-eosin, original magnification ×40).

Figure 11.

Mesothelioma in situ. A, Papillary proliferation lined by a flat, cytomorphologically banal mesothelium, with (B) BAP1 loss. C, A separate case shows flat mesothelial lining with (D) BAP1 loss (hematoxylin-eosin, original magnifications ×100 [A and C] and ×200 [B]; original magnification ×100 [D]).

Figure 12.Mesothelioma, cytology preparation. Features of mesothelioma include (A) cell-in-cell arrangement and (B) a hump at the cell periphery (Papanicolaou, original magnification ×1000 [A and B]).

Figure 13.Well-differentiated papillary mesothelial tumor (hematoxylin-eosin, original magnification ×40).

Close modal

There are no published criteria on minimum acceptable sample size, but caution is warranted when biopsy samples are very small or crushed. The WHO recommends thoracoscopic evaluation with large biopsy specimens (ideally 100–200 mm2) from different areas of the pleura in patients with nonresolving effusions.4  Similarly, no criteria are published for which samples should be tested for BAP1, MTAP, and CDKN2A alterations, but a low threshold is suggested for patients with unexplained recurrent effusions, history of occupational exposures, genetic predisposition, history of chest radiation, or atypical histologic features.

The differential diagnosis of MIS includes reactive mesothelial atypia and well-differentiated papillary mesothelial tumor (WDPMT), depending on lesional architecture.148,149  BAP1 and MTAP immunostains should be performed in WDPMT-like lesions identified on investigation of an effusion or related symptoms, with aberrant results supporting MIS with WDPMT-like morphology, in the correct clinical context. Absence of BAP1, MTAP, and CDKN2A alterations does not exclude MIS, and an expert opinion is advisable in difficult cases, given the potential for malignant behavior.

The WHO classification4  currently only describes MIS in the pleural space, but peritoneal, pericardial, and paratesticular presentations are described, and the same diagnostic criteria can be applied.150–152  At present, MIS has only been established for epithelioid mesothelioma.

The 2021 WHO classification4  emphasizes that MIS is a multidisciplinary diagnosis. Communication with the clinical team is especially important because time to progression may range from 1 year146,149  to 15 years.153  No treatment guidelines exist currently.

Up to 90% of patients with diffuse pleural mesothelioma present with a pleural effusion. Consequently, cytology fluid specimens are often the first (and in patients who cannot tolerate additional procedures, the only) sample available for diagnosis.

Cytomorphology

The typical cytologic features of epithelioid mesothelioma were described more than 50 years ago and have been refined in numerous subsequent publications.154–156  Mesothelioma can manifest cytologically as a highly cellular effusion with obvious nuclear atypia, numerous large tissue fragments and cell clusters, or as cellular fluid with single and clustered mesothelial cells exhibiting only subtle atypia, overlapping with reactive mesothelium. Cell-in-cell arrangements, a hump at the cell periphery, multinucleated cells, papillary groups with basement membrane cores, and orangeophilic cells are features concerning for mesothelioma (Figure 12, A and B). Sarcomatoid mesotheliomas generally do not shed malignant cells into effusions.

Ancillary Studies

As in tissue specimens, the diagnosis of mesothelioma in cytology specimens is a 2-step process in which both mesothelial lineage and malignancy must be established.157–159  Immunocytochemistry plays an invaluable role in both steps and can be performed on smears or cell blocks, although use of cell blocks is preferable, as they undergo processing similar to formalin-fixed, paraffin-embedded tissue specimens, thereby enabling preparation of serial sections for immunostaining and/or molecular studies. Given that cell blocks can be prepared in various ways, it is important to recognize that differences in fixation can affect immunostaining results and to use appropriate controls.53,54  When cell blocks are not prepared, the cell-transfer technique (in which a Papanicolaou-stained sample is divided into several pieces and transferred to multiple slides) can facilitate multiple immunocytochemical stains on limited materials.160 

The same antibodies used to distinguish mesothelial from epithelial lineage in tissue samples (see Establishing Mesothelial Lineage above) can be applied to cytology specimens, provided they have been validated for this application in the responsible laboratory. As in tissue specimens, it is currently recommended to use 2 epithelial and 2 mesothelial markers, though claudin-4 immunocytochemistry can be used as the sole epithelial marker if well validated. When a cytology specimen comprises single cells or loose aggregates, the differential diagnosis may also include melanoma, lymphoma, sarcoma, germ cell, and other tumors, and the immunopanel should be tailored to the differential diagnosis.

BAP1 and MTAP immunocytochemistry, and CDKN2A FISH have been validated for use in cell block preparations (Supplemental Figure 18).108,110,154,158,159,161,162  In distinction of mesothelioma from reactive mesothelial proliferation in pleural effusion cytology specimens, a recent meta-analysis of 65 studies found 100% specificity for CDKN2A homozygous deletion by FISH, and 99% specificity for both BAP1 loss and MTAP loss by immunocytochemistry. The same study found a diagnostic sensitivity of 83% for the combination of BAP1 immunocytochemistry and CDKN2A FISH.163  FISH for NF2 deletion is also reportedly specific for mesothelioma in effusion cytology, though not yet in widespread use.120,164  Although previously viewed as potentially useful markers, it is now clear that positive staining for EMA, IMP-3, CD146, or GLUT1 alone is insufficient to diagnose a cytology specimen as malignant.

Limitations

The inability to assess stromal invasion and sarcomatous elements, coupled with the grim prognosis and expensive and toxic therapy associated with the diagnosis, has contributed to a general reluctance to render a primary definitive diagnosis of mesothelioma based solely on effusion cytology. The reported sensitivity of cytologic diagnosis for mesothelioma is 30% to 75%, though specificity is 99% to 100%.157,165  Sensitivity is almost certainly lower for sarcomatoid mesothelioma, which does not typically shed cells in effusions. Although the epithelioid component of biphasic mesothelioma can shed mesothelioma cells into effusions, biphasic mesothelioma cannot be differentiated from epithelioid mesothelioma on cytology effusion. Additionally, cytology alone cannot distinguish invasive mesothelioma from MIS,166  and correlation with clinical and radiographic findings is recommended in all cases. Architectural subtyping and grading of epithelioid mesothelioma also cannot be performed on cytology specimens.

Non–Effusion Cytology Specimens

The above discussion pertains principally to effusion cytology, which is by far the most common setting for a cytologic diagnosis of mesothelioma. Similar considerations apply to other types of cytology samples. Mesothelioma may rarely be diagnosed in sputum or bronchial washing, lavage, or brushing, and a few studies from the 1980s report use of transthoracic or endoscopic bronchial ultrasound-directed fine-needle aspiration biopsy (FNAB) for diagnosis of mesothelioma. Though rarely reported, definitive primary diagnosis of mesothelioma using these techniques is possible in the appropriate clinical and radiologic context. The cytomorphology in such cases resembles that of mesothelioma in effusions, with high cellularity and papillary clusters, though other features characteristic of mesothelioma in effusions (eg, cell-in-cell arrangements, multinucleated cells) are less commonly observed. Because FNAB directly samples a clinical mass lesion, sarcomatoid mesothelioma is more likely to be diagnosed in an FNAB specimen than in effusion cytology.

Peritoneal Inclusion Cysts

The WHO Classification of Tumors of the female genital tract (which includes peritoneal mesothelial tumors) encourages the diagnostic term peritoneal inclusion cyst and discourages use of multicystic mesothelioma (and similar terms) to avoid confusion with (malignant) mesothelioma.5  Peritoneal inclusion cyst(s) may comprise 1 or multiple cysts lined by bland mesothelial cells without significant stratification, papillary formations, or infiltration of soft tissues. Lesions can be unifocal or multifocal within the pelvis and abdomen. Local recurrence rates as high as 50% have been reported in studies of florid multifocal lesions,167  though a recent study including a more representative population of peritoneal inclusion cysts, as currently defined in the WHO, found a local recurrence rate of just 3%.168  It remains unclear whether peritoneal inclusion cysts represent reactive or neoplastic lesions, though BAP1 and MTAP are universally retained.169 

Well-Differentiated Papillary Mesothelial Tumor

Well-differentiated papillary mesothelial tumor (WDPMT) occurs principally in the peritoneum and is often an incidental surgical finding. Tumors are generally unifocal and small (<2 cm), though larger and/or multifocal examples are reported with otherwise classic morphology.170  WDPMT is composed of slender papillae with hyalinized to myxoid cores lined by a single layer of bland mesothelial cells (Figure 13).171  Mitoses are rare to absent. Infiltration of underlying soft tissue is absent by definition, though occasional WDPMTs show so-called invasive foci, characterized by confluent papillary growth and/or percolating mesothelial nests/cords within papillary stroma (ie, confined to the WDPMT).172  Recurrent mutations in TRAF7, CDC42, EHD1, ATM, FBXO10, and SH2D2A have been reported.173,174 BAP1 and CDKN2A alterations are absent, with BAP1 and MTAP retained by immunohistochemistry.169  Most (60%–95%) are positive for PAX8.171,175 

“Invasive foci” and multifocality are associated with increased recurrence risk and should be reported when present. Malignant transformation is rare and can occur years or decades after original diagnosis. It is uncertain whether such cases represent true WDPMTs, versus MIS with WDPMT-like morphology and/or early epithelioid mesotheliomas with WDPMT-like foci.176  BAP1 immunohistochemistry (and, if necessary, MTAP immunohistochemistry or CDKN2A FISH) should be performed on WDPMT-like lesions discovered during clinical workup for effusion or associated with multifocal or diffuse serosal involvement. BAP1 or MTAP loss supports a diagnosis of WDPMT-like mesothelioma or MIS, but retained staining does not exclude mesothelioma and requires multidisciplinary correlation.148 

Adenomatoid Tumor

Adenomatoid tumors are small, circumscribed, nodular lesions, most often involving the uterus and fallopian tube, and rarely other peritoneal sites.177,178  An association with immunosuppression has been noted.177  Tumors comprise acini, cords, and nests of plump to flattened, bland mesothelial cells. Single cells may be noted, often with a signet ring appearance. Stringlike bridges characteristically span tumor lumina, which may contain hyaluronic acid–rich myxoid material. BAP1 and MTAP are retained.179 TRAF7 mutations have been reported in adenomatoid tumors at various sites.177 

Unusual and Provisional Entities

Rare peritoneal mesothelial lesions show mixed features of peritoneal inclusion cyst, WDPMT, and adenomatoid tumor. The pathogenesis and prognosis of these hybrid lesions remain unclear.171  Occasional noninvasive, morphologically bland, and unifocal or oligofocal nodular mesothelial proliferations defy easy classification. A provisional entity of “solid papillary mesothelial tumor” attempts to encompass some of these lesions, which appear to show indolent behavior.180  In such cases, a descriptive diagnosis with recommendation for clinical and radiographic correlation with close follow-up is generally appropriate.

This article provides broad guidelines for diagnosis of mesothelioma, which, though uncommon, carries a grave prognosis and frequently has medicolegal implications. We emphasize that morphology remains the cornerstone for classification of mesothelial proliferations in both biopsy and resection specimens. Numerous prognostically and clinically significant morphologic features are discussed, and these should be routinely reported in the diagnostic report, whenever possible. Immunohistochemistry and molecular studies play a growing and evolving role in diagnosis and management of mesothelioma. Immunohistochemical panels are routinely applied to establish mesothelial lineage, distinguish mesothelioma from malignant mimics, and distinguish mesothelioma from reactive mesothelial proliferations. Specific immunopanels should be tailored to the clinical and morphologic differential, and immunostains should be carefully validated for optimal performance. Molecular studies, including FISH and targeted sequencing panels, are useful in challenging cases with nondiagnostic morphologic and immunophenotypic findings. These same principles apply to the cytologic diagnosis of mesothelioma, with recognition that cytology does not permit evaluation of stromal invasion, that tissue fixation protocols may impact immunostain performance in cytology preparations, and that sarcomatoid mesothelioma does not typically shed in effusions. Importantly, the pathologist must always correlate morphology and ancillary study results with clinical, radiographic, and operative findings. Despite (or perhaps in part because of) rapid evolution in the field of mesothelioma diagnosis, this remains a challenging and evolving area of surgical and cytopathology, and expert opinion should be sought in difficult cases, when needed.

This article has been endorsed by the Board of the International Mesothelioma Interest Group.

1.
Husain
AN,
Colby
TV,
Ordóñez
NG,
et al.
Guidelines for Pathologic Diagnosis of Malignant Mesothelioma 2017 Update of the Consensus Statement From the International Mesothelioma Interest Group
.
Arch Pathol Lab Med
.
2018
;
142
(
1
):
89
108
.
2.
Marchevsky
AM,
Khoor
A,
Walts
AE,
et al.
Localized malignant mesothelioma, an unusual and poorly characterized neoplasm of serosal origin: best current evidence from the literature and the International Mesothelioma Panel
.
Mod Pathol
.
2020
;
33
(
2
):
281
296
.
3.
Hung
YP,
Dong
F,
Dubuc
AM,
Dal Cin
P,
Bueno
R,
Chirieac
LR.
Molecular characterization of localized pleural mesothelioma
.
Mod Pathol
.
2020
;
33
(
2
):
271
280
.
4.
Sauter
JL,
Bueno
R,
Dacic
S,
et al.
Diffuse pleural mesothelioma. In:
Thoracic Tumours
. 5th ed.
Lyon, France
:
International Agency for Research on Cancer
;
2021
:
204
-
219
. WHO Classification of Tumours; vol 5.
5.
Malpica
A,
Baker
P,
Cheung
AN,
Djordjevic
B.
Peritoneal inclusion cysts. In:
Female Genital Tumours
. 5th ed.
Lyon, France
:
International Agency for Research on Cancer
;
2020
:
202
. WHO Classification of Tumours; vol 4.
6.
Verma
V,
Ahern
CA,
Berlind
CG,
et al.
Survival by histologic subtype of malignant pleural mesothelioma and the impact of surgical resection on overall survival
.
Clin Lung Cancer
.
2018
;
19
(
6
):
e901
e912
.
7.
Chirieac
LR,
Hung
YP,
Foo
WC,
et al.
Diagnostic value of biopsy sampling in predicting histology in patients with diffuse malignant pleural mesothelioma
.
Cancer
.
2019
;
125
(
23
):
4164
4171
.
8.
Chapel
DB,
Schulte
JJ,
Absenger
G,
et al.
Malignant peritoneal mesothelioma: prognostic significance of clinical and pathologic parameters and validation of a nuclear-grading system in a multi-institutional series of 225 cases
.
Mod Pathol
.
2021
;
34
(
2
):
380
395
.
9.
Malpica
A,
Euscher
ED,
Marques-Piubelli
ML,
et al.
Malignant mesothelioma of the peritoneum in women: a clinicopathologic study of 164 cases
.
Am J Surg Pathol
.
2021
;
45
(
1
):
45
58
.
10.
Sauter
JL,
Dacic
S,
Galateau-Salle
F,
et al.
The 2021 WHO Classification of Tumors of the Pleura: advances since the 2015 classification
.
J Thorac Oncol
.
2022
;
17
(
5
):
608
622
.
11.
Nicholson
AG,
Sauter
JL,
Nowak
AK,
et al.
EURACAN/IASLC proposals for updating the histologic classification of pleural mesothelioma: towards a more multidisciplinary approach
.
J Thorac Oncol
.
2020
;
15
(
1
):
29
49
.
12.
Rosen
LE,
Karrison
T,
Ananthanarayanan
V,
et al.
Nuclear grade and necrosis predict prognosis in malignant epithelioid pleural mesothelioma: a multi-institutional study
.
Mod Pathol
.
2018
;
31
(
4
):
598
606
.
13.
Alchami
FS,
Attanoos
RL,
Bamber
AR.
Myxoid variant epithelioid pleural mesothelioma defines a favourable prognosis group: an analysis of 191 patients with pleural malignant mesothelioma
.
J Clin Pathol
.
2017
;
70
(
2
):
179
182
.
14.
Shia
J,
Qin
J,
Erlandson
RA,
et al.
Malignant mesothelioma with a pronounced myxoid stroma: a clinical and pathological evaluation of 19 cases
.
Virchows Arch
.
2005
;
447
(
5
):
828
834
.
15.
Ordóñez
NG.
Mesothelioma with rhabdoid features: an ultrastructural and immunohistochemical study of 10 cases
.
Mod Pathol
.
2006
;
19
(
3
):
373
383
.
16.
Kadota
K,
Suzuki
K,
Sima
CS,
Rusch
VW,
Adusumilli
PS,
Travis
WD.
Pleomorphic epithelioid diffuse malignant pleural mesothelioma: a clinicopathological review and conceptual proposal to reclassify as biphasic or sarcomatoid mesothelioma
.
J Thorac Oncol
.
2011
;
6
(
5
):
896
904
.
17.
Ordóñez
NG.
Pleomorphic mesothelioma: report of 10 cases
.
Mod Pathol
.
2012
;
25
(
7
):
1011
1022
.
18.
Roy
S,
Galateau-Sallé
F,
Le Stang
N,
et al.
Molecular characterization of pleomorphic mesothelioma: a multi-institutional study
.
Mod Pathol
.
2022
;
35
(
1
):
82
86
.
19.
Galateau-Sallé
F,
Attanoos
R,
Gibbs
AR,
et al.
Lymphohistiocytoid variant of malignant mesothelioma of the pleura: a series of 22 cases
.
Am J Surg Pathol
.
2007
;
31
(
5
):
711
716
.
20.
Ordóñez
NG.
Mesothelioma with clear cell features: an ultrastructural and immunohistochemical study of 20 cases
.
Hum Pathol
.
2005
;
36
(
5
):
465
473
.
21.
Ordóñez
NG.
Deciduoid mesothelioma: report of 21 cases with review of the literature
.
Mod Pathol
.
2012
;
25
(
11
):
1481
1495
.
22.
Ordóñez
NG.
Mesothelioma with signet-ring cell features: report of 23 cases
.
Mod Pathol
.
2013
;
26
(
3
):
370
384
.
23.
Ordóñez
NG.
Mesotheliomas with small cell features: report of eight cases
.
Mod Pathol
.
2012
;
25
(
5
):
689
698
.
24.
Klebe
S,
Brownlee
NA,
Mahar
A,
et al.
Sarcomatoid mesothelioma: a clinical-pathologic correlation of 326 cases
.
Mod Pathol
.
2010
;
23
(
3
):
470
479
.
25.
Klebe
S,
Mahar
A,
Henderson
DW,
Roggli
VL.
Malignant mesothelioma with heterologous elements: clinicopathological correlation of 27 cases and literature review
.
Mod Pathol
.
2008
;
21
(
9
):
1084
1094
.
26.
Zhang
YZ,
Brambilla
C,
Molyneaux
PL,
et al.
Utility of nuclear grading system in epithelioid malignant pleural mesothelioma in biopsy-heavy setting: an external validation study of 563 cases
.
Am J Surg Pathol
.
2020
;
44
(
3
):
347
356
.
27.
Hashimoto
K,
Okuma
Y,
Hosomi
Y,
Hishima
T.
Malignant mesothelioma of the pleura with desmoplastic histology: a case series and literature review
.
BMC Cancer
.
2016
;
16
(
1
):
718
.
28.
Dacic
S,
Le Stang
N,
Husain
A,
et al.
Interobserver variation in the assessment of the sarcomatoid and transitional components in biphasic mesotheliomas
.
Mod Pathol
.
2020
;
33
(
2
):
255
262
.
29.
Galateau Salle
F,
Le Stang
N,
Tirode
F,
et al.
Comprehensive molecular and pathologic evaluation of transitional mesothelioma assisted by deep learning approach: a multi-institutional study of the International Mesothelioma Panel from the MESOPATH Reference Center
.
J Thorac Oncol
.
2020
;
15
(
6
):
1037
1053
.
30.
Schulte
JJ,
Chapel
DB,
Attanoos
R,
et al.
Comparison of nuclear grade, necrosis, and histologic subtype between biopsy and resection in pleural malignant mesothelioma: an international multi-institutional analysis
.
Am J Clin Pathol
.
2021
;
156
(
6
):
989
999
.
31.
Borczuk
AC,
Taub
RN,
Hesdorffer
M,
et al.
P16 loss and mitotic activity predict poor survival in patients with peritoneal malignant mesothelioma
.
Clin Cancer Res
.
2005
;
11
(
9
):
3303
3308
.
32.
Sugarbaker
PH,
Welch
LS,
Mohamed
F,
Glehen
O.
A review of peritoneal mesothelioma at the Washington Cancer Institute
.
Surg Oncol Clin N Am
.
2003
;
12
(
3
):
605
621
,
xi
.
33.
Vigneswaran
WT,
Kircheva
DY,
Ananthanarayanan
V,
et al.
Amount of epithelioid differentiation is a predictor of survival in malignant pleural mesothelioma
.
Ann Thorac Surg
.
2017
;
103
(
3
):
962
966
.
34.
Galateau Salle
F,
Le Stang
N,
Nicholson
AG,
et al.
New insights on diagnostic reproducibility of biphasic mesotheliomas: a multi-institutional evaluation by the International Mesothelioma Panel from the MESOPATH Reference Center
.
J Thorac Oncol
.
2018
;
13
(
8
):
1189
1203
.
35.
Brcic
L,
Vlacic
G,
Quehenberger
F,
Kern
I.
Reproducibility of malignant pleural mesothelioma histopathologic subtyping
.
Arch Pathol Lab Med
.
2018
;
142
(
6
):
747
752
.
36.
Hmeljak
J,
Sanchez-Vega
F,
Hoadley
KA,
et al.
Integrative molecular characterization of malignant pleural mesothelioma
.
Cancer Discov
.
2018
;
8
(
12
):
1548
1565
.
37.
Bueno
R,
Stawiski
EW,
Goldstein
LD,
et al.
Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations
.
Nat Genet
.
2016
;
48
(
4
):
407
416
.
38.
Ren
H,
Rassekh
SR,
Lacson
A,
et al.
Malignant mesothelioma with EWSR1-ATF1 fusion in two adolescent male patients
.
Pediatr Dev Pathol
.
2021
;
24
(
6
):
570
574
.
39.
Argani
P,
Harvey
I,
Nielsen
GP,
et al.
EWSR1/FUS-CREB fusions define a distinctive malignant epithelioid neoplasm with predilection for mesothelial-lined cavities
.
Mod Pathol
.
2020
;
33
(
11
):
2233
2243
.
40.
Hung
YP,
Dong
F,
Watkins
JC,
et al.
Identification of ALK rearrangements in malignant peritoneal mesothelioma
.
JAMA Oncol
.
2018
;
4
(
2
):
235
238
.
41.
Dermawan
JK,
Torrence
D,
Lee
CH,
et al.
EWSR1::YY1 fusion positive peritoneal epithelioid mesothelioma harbors mesothelioma epigenetic signature: report of 3 cases in support of an emerging entity
.
Genes Chromosomes Cancer
.
2022
;
61
(
10
):
592
602
.
42.
Hung
YP,
Dong
F,
Torre
M,
Crum
CP,
Bueno
R,
Chirieac
LR.
Molecular characterization of diffuse malignant peritoneal mesothelioma
.
Mod Pathol
.
2020
;
33
(
11
):
2269
2279
.
43.
Salvi
S,
Varesano
S,
Boccardo
S,
et al.
FISH analysis of crizotinib target genes ROS1/ALK/MET in malignant mesothelioma
.
J Thorac Oncol
.
2017
;
12
(
8
):
e116
e118
.
44.
Leal
JL,
Peters
G,
Szaumkessel
M,
et al.
NTRK and ALK rearrangements in malignant pleural mesothelioma, pulmonary neuroendocrine tumours and non-small cell lung cancer
.
Lung Cancer
.
2020
;
146
:
154
159
.
45.
Carbone
M,
Harbour
JW,
Brugarolas
J,
et al.
biological mechanisms and clinical significance of BAP1 mutations in human cancer
.
Cancer Discov
.
2020
;
10
(
8
):
1103
1120
.
46.
Leblay
N,
Leprêtre
F,
Le Stang
N,
et al.
BAP1 is altered by copy number loss, mutation, and/or loss of protein expression in more than 70% of malignant peritoneal mesotheliomas
.
J Thorac Oncol
.
2017
;
12
(
4
):
724
733
.
47.
Illei
PB,
Rusch
VW,
Zakowski
MF,
Ladanyi
M.
Homozygous deletion of CDKN2A and codeletion of the methylthioadenosine phosphorylase gene in the majority of pleural mesotheliomas
.
Clin Cancer Res
.
2003
;
9
(
6
):
2108
2113
.
48.
Chapel
DB,
Hornick
JL,
Barlow
J,
Bueno
R,
Sholl
LM.
Clinical and molecular validation of BAP1, MTAP, P53, and Merlin immunohistochemistry in diagnosis of pleural mesothelioma
.
Mod Pathol
.
2022
;
35
(
10
):
1383
1397
.
49.
Chapel
DB,
Dubuc
AM,
Hornick
JL,
Sholl
LM.
Correlation of methylthioadenosine phosphorylase (MTAP) protein expression with MTAP and CDKN2A copy number in malignant pleural mesothelioma
.
Histopathology
.
2021
;
78
(
7
):
1032
1042
.
50.
Petrilli
AM,
Fernández-Valle
C.
Role of Merlin/NF2 inactivation in tumor biology
.
Oncogene
.
2016
;
35
(
5
):
537
548
.
51.
Meiller
C,
Montagne
F,
Hirsch
TZ,
et al.
Multi-site tumor sampling highlights molecular intra-tumor heterogeneity in malignant pleural mesothelioma
.
Genome Med
.
2021
;
13
(
1
):
113
.
52.
Chirieac
LR,
Pinkus
GS,
Pinkus
JL,
Godleski
J,
Sugarbaker
DJ,
Corson
JM.
The immunohistochemical characterization of sarcomatoid malignant mesothelioma of the pleura
.
Am J Cancer Res
.
2011
;
1
(
1
):
14
24
.
53.
Buonocore
DJ,
Konno
F,
Jungbluth
AA,
et al.
CytoLyt fixation significantly inhibits MIB1 immunoreactivity whereas alternative Ki-67 clone 30-9 is not susceptible to the inhibition: critical diagnostic implications
.
Cancer Cytopathol
.
2019
;
127
(
10
):
643
649
.
54.
Sauter
JL,
Grogg
KL,
Vrana
JA,
Law
ME,
Halvorson
JL,
Henry
MR.
Young investigator challenge: validation and optimization of immunohistochemistry protocols for use on cellient cell block specimens
.
Cancer Cytopathol
.
2016
;
124
(
2
):
89
100
.
55.
Fitzgibbons
PL,
Bradley
LA,
Fatheree
LA,
et al.
Principles of analytic validation of immunohistochemical assays: guideline from the College of American Pathologists Pathology and Laboratory Quality Center
.
Arch Pathol Lab Med
.
2014
;
138
(
11
):
1432
1443
.
56.
Chapel
DB,
Schulte
JJ,
Husain
AN,
Krausz
T.
Application of immunohistochemistry in diagnosis and management of malignant mesothelioma
.
Transl Lung Cancer Res
.
2020
;
9
(suppl
1
):
S3
S27
.
57.
Chapel
DB,
Vivero
M,
Sholl
LM.
Mesothelioma. In:
Practical Pulmonary Pathology
. Elsevier;
2024
;
755
792
.
58.
Churg
A,
Naso
JR.
Hypothesis: HEG1 and claudin-4 staining will allow a diagnosis of epithelioid and biphasic mesothelioma versus non-small-cell lung carcinoma with only two stains in most cases
.
Histopathology
.
2023
;
82
(
3
):
385
392
.
59.
Naso
JR,
Churg
A.
Claudin-4 shows superior specificity for mesothelioma vs non-small-cell lung carcinoma compared with MOC-31 and Ber-EP4
.
Hum Pathol
.
2020
;
100
:
10
14
.
60.
Hiroshima
K,
Wu
D,
Koh
E,
et al.
Membranous HEG1 expression is a useful marker in the differential diagnosis of epithelioid and biphasic malignant mesothelioma versus carcinomas
.
Pathol Int
.
2021
;
71
(
9
):
604
613
.
61.
Naso
JR,
Tsuji
S,
Churg
A.
HEG1 is a highly specific and sensitive marker of epithelioid malignant mesothelioma
.
Am J Surg Pathol
.
2020
;
44
(
8
):
1143
1148
.
62.
Patel
A,
Borczuk
AC,
Siddiqui
MT.
Utility of Claudin-4 versus BerEP4 and B72.3 in pleural fluids with metastatic lung adenocarcinoma
.
J Am Soc Cytopathol
.
2020
;
9
(
3
):
146
151
.
63.
Bishop
JA,
Sharma
R,
Illei
PB.
Napsin A and thyroid transcription factor-1 expression in carcinomas of the lung, breast, pancreas, colon, kidney, thyroid, and malignant mesothelioma
.
Hum Pathol
.
2010
;
41
(
1
):
20
25
.
64.
Klebe
S,
Swalling
A,
Jonavicius
L,
Henderson
DW.
An immunohistochemical comparison of two TTF-1 monoclonal antibodies in atypical squamous lesions and sarcomatoid carcinoma of the lung, and pleural malignant mesothelioma
.
J Clin Pathol
.
2016
;
69
(
2
):
136
141
.
65.
Ordóñez
NG.
The diagnostic utility of immunohistochemistry in distinguishing between epithelioid mesotheliomas and squamous carcinomas of the lung: a comparative study
.
Mod Pathol
.
2006
;
19
(
3
):
417
428
.
66.
Ordóñez
NG.
Value of claudin-4 immunostaining in the diagnosis of mesothelioma
.
Am J Clin Pathol
.
2013
;
139
(
5
):
611
619
.
67.
Tatsumori
T,
Tsuta
K,
Masai
K,
et al.
p40 is the best marker for diagnosing pulmonary squamous cell carcinoma: comparison with p63, cytokeratin 5/6, desmocollin-3, and sox2
.
Appl Immunohistochem Mol Morphol
.
2014
;
22
(
5
):
377
382
.
68.
Mawas
AS,
Amatya
VJ,
Kushitani
K,
et al.
MUC4 immunohistochemistry is useful in distinguishing epithelioid mesothelioma from adenocarcinoma and squamous cell carcinoma of the lung
.
Sci Rep
.
2018
;
8
(
1
):
134
.
69.
Kushitani
K,
Amatya
VJ,
Okada
Y,
et al.
Utility and pitfalls of immunohistochemistry in the differential diagnosis between epithelioid mesothelioma and poorly differentiated lung squamous cell carcinoma
.
Histopathology
.
2017
;
70
(
3
):
375
384
.
70.
Pelosi
G,
Fabbri
A,
Bianchi
F,
et al.
ΔNp63 (p40) and thyroid transcription factor-1 immunoreactivity on small biopsies or cellblocks for typing non-small cell lung cancer: a novel two-hit, sparing-material approach
.
J Thorac Oncol
.
2012
;
7
(
2
):
281
290
.
71.
Ordóñez
NG,
Sahin
AA.
Diagnostic utility of immunohistochemistry in distinguishing between epithelioid pleural mesotheliomas and breast carcinomas: a comparative study
.
Hum Pathol
.
2014
;
45
(
7
):
1529
1540
.
72.
Taliano
RJ,
Lu
S,
Singh
K,
et al.
Calretinin expression in high-grade invasive ductal carcinoma of the breast is associated with basal-like subtype and unfavorable prognosis
.
Hum Pathol
.
2013
;
44
(
12
):
2743
2750
.
73.
Duhig
EE,
Kalpakos
L,
Yang
IA,
Clarke
BE.
Mesothelial markers in high-grade breast carcinoma
.
Histopathology
.
2011
;
59
(
5
):
957
964
.
74.
Miettinen
M,
McCue
PA,
Sarlomo-Rikala
M,
et al.
GATA3: a multispecific but potentially useful marker in surgical
pathology: a systematic analysis of 2500 epithelial and nonepithelial tumors. Am J Surg Pathol
.
2014
;
38
(
1
):
13
22
.
75.
Ordóñez
NG.
The diagnostic utility of immunohistochemistry in distinguishing between mesothelioma and renal cell carcinoma: a comparative study
.
Hum Pathol
.
2004
;
35
(
6
):
697
710
.
76.
Ordóñez
NG.
Value of PAX8, PAX2, napsin A, carbonic anhydrase IX, and claudin-4 immunostaining in distinguishing pleural epithelioid mesothelioma from metastatic renal cell carcinoma
.
Mod Pathol
.
2013
;
26
(
8
):
1132
1143
.
77.
Laury
AR,
Perets
R,
Piao
H,
et al.
A comprehensive analysis of PAX8 expression in human epithelial tumors
.
Am J Surg Pathol
.
2011
;
35
(
6
):
816
826
.
78.
Ordóñez
NG.
Value of PAX8, PAX2, claudin-4, and h-caldesmon immunostaining in distinguishing peritoneal epithelioid mesotheliomas from serous carcinomas
.
Mod Pathol
.
2013
;
26
(
4
):
553
562
.
79.
Chapel
DB,
Husain
AN,
Krausz
T,
McGregor
SM.
PAX8 expression in a subset of malignant peritoneal mesotheliomas and benign mesothelium has diagnostic implications in the differential diagnosis of ovarian serous carcinoma
.
Am J Surg Pathol
.
2017
;
41
(
12
):
1675
1682
.
80.
Butnor
KJ,
Nicholson
AG,
Allred
DC,
et al.
Expression of renal cell carcinoma-associated markers erythropoietin, CD10, and renal cell carcinoma marker in diffuse malignant mesothelioma and metastatic renal cell carcinoma
.
Arch Pathol Lab Med
.
2006
;
130
(
6
):
823
827
.
81.
Ordóñez
NG.
Value of immunohistochemistry in distinguishing peritoneal mesothelioma from serous carcinoma of the ovary and peritoneum: a review and update
.
Adv Anat Pathol
.
2006
;
13
(
1
):
16
25
.
82.
Tandon
RT,
Jimenez-Cortez
Y,
Taub
R,
Borczuk
AC.
Immunohistochemistry in peritoneal mesothelioma: a single-center experience of 244 cases
.
Arch Pathol Lab Med
.
2018
;
142
(
2
):
236
242
.
83.
Offin
M,
Yang
SR,
Egger
J,
et al.
Molecular characterization of peritoneal mesotheliomas
.
J Thorac Oncol
.
2022
;
17
(
3
):
455
460
.
84.
Ordóñez
NG.
Application of immunohistochemistry in the diagnosis of epithelioid mesothelioma: a review and update
.
Hum Pathol
.
2013
;
44
(
1
):
1
19
.
85.
Bonk
S,
Kluth
M,
Hube-Magg
C,
et al.
Prognostic and diagnostic role of PSA immunohistochemistry: a tissue microarray study on 21,000 normal and cancerous tissues
.
Oncotarget
.
2019
;
10
(
52
):
5439
5453
.
86.
Lin
F,
Shi
J,
Zhu
S,
et al.
Cadherin-17 and SATB2 are sensitive and specific immunomarkers for medullary carcinoma of the large intestine
.
Arch Pathol Lab Med
.
2014
;
138
(
8
):
1015
1026
.
87.
Ordóñez
NG.
D2-40 and podoplanin are highly specific and sensitive immunohistochemical markers of epithelioid malignant mesothelioma
.
Hum Pathol
.
2005
;
36
(
4
):
372
380
.
88.
Hinterberger
M,
Reineke
T,
Storz
M,
Weder
W,
Vogt
P,
Moch
H.
D2-40 and calretinin—a tissue microarray analysis of 341 malignant mesotheliomas with emphasis on sarcomatoid differentiation
.
Mod Pathol
.
2007
;
20
(
2
):
248
255
.
89.
Attanoos
RL,
Dojcinov
SD,
Webb
R,
Gibbs
AR.
Anti-mesothelial markers in sarcomatoid mesothelioma and other spindle cell neoplasms
.
Histopathology
.
2000
;
37
(
3
):
224
231
.
90.
Terra
SBSP,
Roden
AC,
Aubry
MC,
Yi
ESJ,
Boland
JM.
Utility of immunohistochemistry for MUC4 and GATA3 to aid in the distinction of pleural sarcomatoid mesothelioma from pulmonary sarcomatoid carcinoma
.
Arch Pathol Lab Med
.
2021
;
145
(
2
):
208
213
.
91.
Berg
KB,
Churg
A.
GATA3 Immunohistochemistry for distinguishing sarcomatoid and desmoplastic mesothelioma from sarcomatoid carcinoma of the lung
.
Am J Surg Pathol
.
2017
;
41
(
9
):
1221
1225
.
92.
Prabhakaran
S,
Hocking
A,
Kim
C,
Hussey
M,
Klebe
S.
The potential utility of GATA binding protein 3 for diagnosis of malignant pleural mesotheliomas
.
Hum Pathol
.
2020
;
105
:
1
8
.
93.
Chang
A,
Brimo
F,
Montgomery
EA,
Epstein
JI.
Use of PAX8 and GATA3 in diagnosing sarcomatoid renal cell carcinoma and sarcomatoid urothelial carcinoma
.
Hum Pathol
.
2013
;
44
(
8
):
1563
1568
.
94.
Baranov
E,
McBride
MJ,
Bellizzi
AM,
et al.
A novel SS18-SSX fusion-specific antibody for the diagnosis of synovial sarcoma
.
Am J Surg Pathol
.
2020
;
44
(
7
):
922
933
.
95.
Mayall
FG,
Goddard
H,
Gibbs
AR.
The diagnostic implications of variable cytokeratin expression in mesotheliomas
.
J Pathol
.
1993
;
170
(
2
):
165
168
.
96.
Lucas
DR,
Pass
HI,
Madan
SK,
et al.
Sarcomatoid mesothelioma and its histological mimics: a comparative immunohistochemical study
.
Histopathology
.
2003
;
42
(
3
):
270
279
.
97.
Miettinen
M,
Limon
J,
Niezabitowski
A,
Lasota
J.
Calretinin and other mesothelioma markers in synovial sarcoma: analysis of antigenic similarities and differences with malignant mesothelioma
.
Am J Surg Pathol
.
2001
;
25
(
5
):
610
617
.
98.
Kung
IT,
Thallas
V,
Spencer
EJ,
Wilson
SM.
Expression of muscle actins in diffuse mesotheliomas
.
Hum Pathol
.
1995
;
26
(
5
):
565
570
.
99.
Trupiano
JK,
Geisinger
KR,
Willingham
MC,
et al.
Diffuse malignant mesothelioma of the peritoneum and pleura, analysis of markers
.
Mod Pathol
.
2004
;
17
(
4
):
476
481
.
100.
Hammar
SP,
Bockus
DE,
Remington
FL,
Rohrbach
KA.
Mucin-positive epithelial mesotheliomas: a histochemical, immunohistochemical, and ultrastructural comparison with mucin-producing pulmonary adenocarcinomas
.
Ultrastruct Pathol
.
1996
;
20
(
4
):
293
325
.
101.
Hammar
SP.
Macroscopic, histologic, histochemical, immunohistochemical, and ultrastructural features of mesothelioma
.
Ultrastruct Pathol
.
2006
;
30
(
1
):
3
17
.
102.
Dardick
I,
Al-Jabi
M,
McCaughey
WT,
Srigley
JR,
van Nostrand
AW,
Ritchie
AC.
Ultrastructure of poorly differentiated diffuse epithelial mesotheliomas
.
Ultrastruct Pathol
.
1984
;
7
(
2-3
):
151
160
.
103.
Dardick
I,
Jabi
M,
McCaughey
WT,
Deodhare
S,
van Nostrand
AW,
Srigley
JR.
Diffuse epithelial mesothelioma: a review of the ultrastructural spectrum
.
Ultrastruct Pathol
.
1987
;
11
(
5-6
):
503
533
.
104.
Churg
A,
Colby
TV,
Cagle
P,
et al.
The separation of benign and malignant mesothelial proliferations
.
Am J Surg Pathol
.
2000
;
24
(
9
):
1183
1200
.
105.
Churg
A,
Cagle
P,
Colby
TV,
et al.
The fake fat phenomenon in organizing pleuritis: a source of confusion with desmoplastic malignant mesotheliomas
.
Am J Surg Pathol
.
2011
;
35
(
12
):
1823
1829
.
106.
Churg
A,
Sheffield
BS,
Galateau-Salle
F.
New markers for separating benign from malignant mesothelial proliferations: are we there yet
?
Arch Pathol Lab Med
.
2016
;
140
(
4
):
318
321
.
107.
Hwang
HC,
Pyott
S,
Rodriguez
S,
et al.
BAP1 immunohistochemistry and p16 FISH in the diagnosis of sarcomatous and desmoplastic mesotheliomas
.
Am J Surg Pathol
.
2016
;
40
(
5
):
714
718
.
108.
Hwang
HC,
Sheffield
BS,
Rodriguez
S,
et al.
Utility of BAP1 immunohistochemistry and p16 (CDKN2A) FISH in the diagnosis of malignant mesothelioma in effusion cytology specimens
.
Am J Surg Pathol
.
2016
;
40
(
1
):
120
126
.
109.
Hida
T,
Hamasaki
M,
Matsumoto
S,
et al.
BAP1 immunohistochemistry and p16 FISH results in combination provide higher confidence in malignant pleural mesothelioma diagnosis: ROC analysis of the two tests
.
Pathol Int
.
2016
;
66
(
10
):
563
570
.
110.
Walts
AE,
Hiroshima
K,
McGregor
SM,
Wu
D,
Husain
AN,
Marchevsky
AM.
BAP1 immunostain and CDKN2A (p16) FISH analysis: clinical applicability for the diagnosis of malignant mesothelioma in effusions
.
Diagn Cytopathol
.
2016
;
44
(
7
):
599
606
.
111.
Hida
T,
Hamasaki
M,
Matsumoto
S,
et al.
Immunohistochemical detection of MTAP and BAP1 protein loss for mesothelioma diagnosis: comparison with 9p21 FISH and BAP1 immunohistochemistry
.
Lung Cancer
.
2017
;
104
:
98
105
.
112.
Chapel
DB,
Schulte
JJ,
Berg
K,
et al.
MTAP immunohistochemistry is an accurate and reproducible surrogate for CDKN2A fluorescence in situ hybridization in diagnosis of malignant pleural mesothelioma
.
Mod Pathol
.
2020
;
33
(
2
):
245
254
.
113.
Churg
A,
Naso
JR.
The separation of benign and malignant mesothelial proliferations: new markers and how to use them
.
Am J Surg Pathol
.
2020
;
44
(
11
):
e100
e112
.
114.
McGregor
SM,
McElherne
J,
Minor
A,
et al.
BAP1 immunohistochemistry has limited prognostic utility as a complement of CDKN2A (p16) fluorescence in situ hybridization in malignant pleural mesothelioma
.
Hum Pathol
.
2017
;
60
:
86
94
.
115.
Kinoshita
Y,
Hamasaki
M,
Yoshimura
M,
et al.
A combination of MTAP and BAP1 immunohistochemistry is effective for distinguishing sarcomatoid mesothelioma from fibrous pleuritis
.
Lung Cancer
.
2018
;
125
:
198
204
.
116.
Sa-Ngiamwibool
P,
Hamasaki
M,
Kinoshita
Y,
et al.
Challenges and limitation of MTAP immunohistochemistry in diagnosing desmoplastic mesothelioma/sarcomatoid pleural mesothelioma with desmoplastic features
.
Ann Diagn Pathol
.
2022
;
60
:
152004
.
117.
Andrici
J,
Jung
J,
Sheen
A,
et al.
Loss of BAP1 expression is very rare in peritoneal and gynecologic serous adenocarcinomas and can be useful in the differential diagnosis with abdominal mesothelioma
.
Hum Pathol
.
2016
;
51
:
9
15
.
118.
Andrici
J,
Parkhill
TR,
Jung
J,
et al.
Loss of expression of BAP1 is very rare in non-small cell lung carcinoma
.
Pathology
.
2016
;
48
(
4
):
336
340
.
119.
Naso
JR,
Tessier-Cloutier
B,
Senz
J,
Huntsman
DG,
Churg
A.
Significance of p53 immunostaining in mesothelial proliferations and correlation with TP53 mutation status
.
Mod Pathol
.
2022
;
35
(
1
):
77
81
.
120.
Kinoshita
Y,
Hamasaki
M,
Yoshimura
M,
Matsumoto
S,
Iwasaki
A,
Nabeshima
K.
Hemizygous loss of NF2 detected by fluorescence in situ hybridization is useful for the diagnosis of malignant pleural mesothelioma
.
Mod Pathol
.
2020
;
33
(
2
):
235
244
.
121.
Martin
SD,
Cheung
S,
Churg
A.
Immunohistochemical demonstration of Merlin/NF2 loss in mesothelioma
.
Mod Pathol
.
2023
;
36
(
1
):
100036
.
122.
Wu
D,
Hiroshima
K,
Yusa
T,
et al.
Usefulness of p16/CDKN2A fluorescence in situ hybridization and BAP1 immunohistochemistry for the diagnosis of biphasic mesothelioma
.
Ann Diagn Pathol
.
2017
;
26
:
31
37
.
123.
Righi
L,
Duregon
E,
Vatrano
S,
et al.
BRCA1-associated protein 1 (BAP1) immunohistochemical expression as a diagnostic tool in malignant pleural mesothelioma classification: a large retrospective study
.
J Thorac Oncol
.
2016
;
11
(
11
):
2006
2017
.
124.
McGregor
SM,
Dunning
R,
Hyjek
E,
Vigneswaran
W,
Husain
AN,
Krausz
T.
BAP1 facilitates diagnostic objectivity, classification, and prognostication in malignant pleural mesothelioma
.
Hum Pathol
.
2015
;
46
(
11
):
1670
1678
.
125.
Klebe
S,
Brcic
L,
Galateau Salle
F,
et al.
Mesothelioma in the Pleura, Pericardium and Peritoneum
. 3rd ed. https://www.iccr-cancer.org/datasets/published-datasets/thorax/mesothelioma/ Accessed April 3, 2023.
126.
Rusch
VW,
Chansky
K,
Nowak
AK,
et al.
Malignant pleural mesothelioma. In:
AJCC Cancer Staging Manual
. 8th ed.
Springer
;
2017
:
457
-
468
.
127.
Meyerhoff
RR,
Yang
CFJ,
Speicher
PJ,
et al.
Impact of mesothelioma histologic subtype on outcomes in the Surveillance, Epidemiology, and End Results database
.
J Surg Res
.
2015
;
196
(
1
):
23
32
.
128.
Kadota
K,
Suzuki
K,
Colovos
C,
et al.
A nuclear grading system is a strong predictor of survival in epitheloid diffuse malignant pleural mesothelioma
.
Mod Pathol
.
2012
;
25
(
2
):
260
271
.
129.
Jean
D,
Daubriac
J,
Le Pimpec-Barthes
F,
Galateau-Salle
F,
Jaurand
MC.
Molecular changes in mesothelioma with an impact on prognosis and treatment
.
Arch Pathol Lab Med
.
2012
;
136
(
3
):
277
293
.
130.
Singhi
AD,
Krasinskas
AM,
Choudry
HA,
et al.
The prognostic significance of BAP1, NF2, and CDKN2A in malignant peritoneal mesothelioma
.
Mod Pathol
.
2016
;
29
(
1
):
14
24
.
131.
Baumann
F,
Flores
E,
Napolitano
A,
et al.
Mesothelioma patients with germline BAP1 mutations have 7-fold improved long-term survival
.
Carcinogenesis
.
2015
;
36
(
1
):
76
81
.
132.
Hassan
R,
Morrow
B,
Thomas
A,
et al.
Inherited predisposition to malignant mesothelioma and overall survival following platinum chemotherapy
.
Proc Natl Acad Sci U S A
.
2019
;
116
(
18
):
9008
9013
.
133.
Panou
V,
Gadiraju
M,
Wolin
A,
et al.
Frequency of germline mutations in cancer susceptibility genes in malignant mesothelioma
.
J Clin Oncol
.
2018
;
36
(
28
):
2863
2871
.
134.
Carbone
M,
Pass
HI,
Ak
G,
et al.
Medical and surgical care of patients with mesothelioma and their relatives carrying germline BAP1 mutations
.
J Thorac Oncol
.
2022
;
17
(
7
):
873
889
.
135.
Pastorino
S,
Yoshikawa
Y,
Pass
HI,
et al.
A subset of mesotheliomas with improved survival occurring in carriers of BAP1 and other germline mutations
.
J Clin Oncol
.
2018
;
36
(
35
):
JCO2018790352
.
136.
Hiltbrunner
S,
Mannarino
L,
Kirschner
MB,
et al.
Tumor immune microenvironment and genetic alterations in mesothelioma
.
Front Oncol
.
2021
;
11
:
660039
.
137.
Baas
P,
Scherpereel
A,
Nowak
AK,
et al.
First-line nivolumab plus ipilimumab in unresectable malignant pleural mesothelioma (CheckMate 743): a multicentre, randomised, open-label, phase 3 trial
.
Lancet
.
2021
;
397
(
10272
):
375
386
.
138.
Muller
S,
Victoria Lai
W,
Adusumilli
PS,
et al.
V-domain Ig-containing suppressor of T-cell activation (VISTA), a potentially targetable immune checkpoint molecule, is highly expressed in epithelioid malignant pleural mesothelioma
.
Mod Pathol
.
2020
;
33
(
2
):
303
311
.
139.
Combaz-Lair
C,
Galateau-Sallé
F,
McLeer-Florin
A,
et al.
Immune biomarkers PD-1/PD-L1 and TLR3 in malignant pleural mesotheliomas
.
Hum Pathol
.
2016
;
52
:
9
18
.
140.
Chapel
DB,
Stewart
R,
Furtado
LV,
Husain
AN,
Krausz
T,
Deftereos
G.
Tumor PD-L1 expression in malignant pleural and peritoneal mesothelioma by Dako PD-L1 22C3 pharmDx and Dako PD-L1 28-8 pharmDx assays
.
Hum Pathol
.
2019
;
87
:
11
17
.
141.
Loharamtaweethong
K,
Puripat
N,
Aoonjai
N,
Sutepvarnon
A,
Bandidwattanawong
C.
Anaplastic lymphoma kinase (ALK) translocation in paediatric malignant peritoneal mesothelioma: a case report of novel ALK-related tumour spectrum
.
Histopathology
.
2016
;
68
(
4
):
603
607
.
142.
Argani
P,
Lian
DWQ,
Agaimy
A,
et al.
Pediatric mesothelioma with ALK fusions: a molecular and pathologic study of 5 cases
.
Am J Surg Pathol
.
2021
;
45
(
5
):
653
661
.
143.
Rüschoff
JH,
Gradhand
E,
Kahraman
A,
et al.
STRN -ALK rearranged malignant peritoneal mesothelioma with dramatic response following ceritinib treatment
.
JCO Precis Oncol
.
2019
;
3
:
PO.19.00048
.
144.
Whitaker
D,
Henderson
DW,
Shilkin
KB.
The concept of mesothelioma in situ: implications for diagnosis and histogenesis
.
Semin Diagn Pathol
.
1992
;
9
(
2
):
151
161
.
145.
Churg
A,
Hwang
H,
Tan
L,
et al.
Malignant mesothelioma in situ
.
Histopathology
.
2018
;
72
(
6
):
1033
1038
.
146.
Churg
A,
Galateau-Salle
F,
Roden
AC,
et al.
Malignant mesothelioma in situ: morphologic features and clinical outcome
.
Mod Pathol
.
2020
;
33
(
2
):
297
302
.
147.
Dacic
S,
Roy
S,
Lyons
MA,
von der Thusen
JH,
Galateau-Salle
F,
Churg
A.
Whole exome sequencing reveals BAP1 somatic abnormalities in mesothelioma in situ
.
Lung Cancer
.
2020
;
149
:
1
4
.
148.
Churg
A,
Galateau-Salle
F.
Well differentiated papillary mesothelial tumor: a new name and new problems
.
Mod Pathol
.
2022
;
35
(
10
):
1327
1333
.
149.
Klebe
S,
Nakatani
Y,
Dobra
K,
et al.
The concept of mesothelioma in situ, with consideration of its potential impact on cytology diagnosis
.
Pathology
.
2021
;
53
(
4
):
446
453
.
150.
Kobayashi
Y,
Yasuhara
Y,
Arai
H,
Honda
M,
Hiramatsu
M,
Goya
S.
Mesothelioma in situ of the spermatic cord arising from a patent processus vaginalis: a case report
.
Urol J
.
2020
;
17
(
6
):
671
673
.
151.
MacLean
A,
Churg
A,
Johnson
ST.
Bilateral pleural mesothelioma in situ and peritoneal mesothelioma in situ associated with BAP1 germline mutation: a case report
.
JTO Clin Res Rep
.
2022
;
3
(
8
):
100356
.
152.
Fukasawa
N,
Agemi
Y,
Shiba
A,
et al.
A case of slowly progressive malignant pericardial mesothelioma suggesting the involvement of BAP1 loss
.
Respirol Case Rep
.
2022
;
10
(
9
):
e01004
.
153.
Hidaka
K,
Takeda
T,
Kinoshita
Y,
et al.
Development of mesothelioma in situ and its progression to invasive disease observed in a patient with uncontrolled pleural effusions for 15 years
.
Pathol Int
.
2020
;
70
(
12
):
1009
1014
.
154.
Hjerpe
A,
Ascoli
V,
Bedrossian
C,
et al.
Guidelines for cytopathologic diagnosis of epithelioid and mixed type malignant mesothelioma: complementary statement from the International Mesothelioma Interest Group, also endorsed by the International Academy of Cytology and the Papanicolaou Society of Cytopathology
.
Cytojournal
.
2015
;
12
:
26
.
155.
Whitaker
D.
The cytology of malignant mesothelioma
.
Cytopathology
.
2000
;
11
(
3
):
139
151
.
156.
Whitaker
D,
Shilkin
KB.
Diagnosis of pleural malignant mesothelioma in life–a practical approach
.
J Pathol
.
1984
;
143
(
3
):
147
175
.
157.
Henderson
DW,
Reid
G,
Kao
SC,
van Zandwijk
N,
Klebe
S.
Challenges and controversies in the diagnosis of mesothelioma: part 1—cytology-only diagnosis, biopsies, immunohistochemistry, discrimination between mesothelioma and reactive mesothelial hyperplasia, and biomarkers
.
J Clin Pathol
.
2013
;
66
(
10
):
847
853
.
158.
Chevrier
M,
Monaco
SE,
Jerome
JA,
Galateau-Salle
F,
Churg
A,
Dacic
S.
Testing for BAP1 loss and CDKN2A/p16 homozygous deletion improves the accurate diagnosis of mesothelial proliferations in effusion cytology
.
Cancer Cytopathol
.
2020
;
128
(
12
):
939
947
.
159.
Hiroshima
K,
Wu
D,
Hasegawa
M,
et al.
Cytologic differential diagnosis of malignant mesothelioma and reactive mesothelial cells with FISH analysis of p16
.
Diagn Cytopathol
.
2016
;
44
(
7
):
591
598
.
160.
Wu
HH,
Jones
KJ,
Cramer
HM.
Immunocytochemistry performed on the cell-transferred direct smears of the fine-needle aspirates: a comparison study with the corresponding formalin-fixed paraffin-embedded tissue
.
Am J Clin Pathol
.
2013
;
139
(
6
):
754
758
.
161.
Hiroshima
K,
Wu
D,
Hamakawa
S,
et al.
HEG1, BAP1, and MTAP are useful in cytologic diagnosis of malignant mesothelioma with effusion
.
Diagn Cytopathol
.
2021
;
49
(
5
):
622
632
.
162.
Berg
KB,
Churg
AM,
Cheung
S,
Dacic
S.
Usefulness of methylthioadenosine phosphorylase and BRCA-associated protein 1 immunohistochemistry in the diagnosis of malignant mesothelioma in effusion cytology specimens
.
Cancer Cytopathol
.
2020
;
128
(
2
):
126
132
.
163.
Girolami
I,
Lucenteforte
E,
Eccher
A,
et al.
Evidence-based diagnostic performance of novel biomarkers for the diagnosis of malignant mesothelioma in effusion cytology
.
Cancer Cytopathol
.
2022
;
130
(
2
):
96
109
.
164.
Sa-Ngiamwibool
P,
Hamasaki
M,
Kinoshita
Y,
et al.
Usefulness of NF2 hemizygous loss detected by fluorescence in situ hybridization in diagnosing pleural mesothelioma in tissue and cytology material: a multi-institutional study
.
Lung Cancer
.
2022
;
175
:
27
35
.
165.
Segal
A,
Sterrett
GF,
Frost
FA,
et al.
A diagnosis of malignant pleural mesothelioma can be made by effusion cytology: results of a 20 year audit
.
Pathology
.
2013
;
45
(
1
):
44
48
.
166.
Louw
A,
van Vliet
C,
Peverall
J,
et al.
Analysis of early pleural fluid samples in patients with mesothelioma: a case series exploration of morphology, BAP1, and CDKN2A status with implications for the concept of mesothelioma in situ in cytology
.
Cancer Cytopathol
.
2022
;
130
(
5
):
352
362
.
167.
Nizri
E,
Baratti
D,
Guaglio
M,
et al.
Multicystic mesothelioma: operative and long-term outcomes with cytoreductive surgery and hyperthermic intra peritoneal chemotherapy
.
Eur J Surg Oncol
.
2018
;
44
(
7
):
1100
1104
.
168.
Karpathiou
G,
Casteillo
F,
Dridi
M,
Peoc’h
M.
Mesothelial cysts
.
Am J Clin Pathol
.
2021
;
155
(
6
):
853
862
.
169.
Shinozaki-Ushiku
A,
Ushiku
T,
Morita
S,
Anraku
M,
Nakajima
J,
Fukayama
M.
Diagnostic utility of BAP1 and EZH2 expression in malignant mesothelioma
.
Histopathology
.
2017
;
70
(
5
):
722
733
.
170.
Malpica
A,
Sant’Ambrogio
S,
Deavers
MT,
Silva
EG.
Well-differentiated papillary mesothelioma of the female peritoneum: a clinicopathologic study of 26 cases
.
Am J Surg Pathol
.
2012
;
36
(
1
):
117
127
.
171.
Sun
M,
Zhao
L,
Weng Lao
I,
Yu
L,
Wang
J.
Well-differentiated papillary mesothelioma: a 17-year single institution experience with a series of 75 cases
.
Ann Diagn Pathol
.
2019
;
38
:
43
50
.
172.
Churg
A,
Allen
T,
Borczuk
AC,
et al.
Well-differentiated papillary mesothelioma with invasive foci
.
Am J Surg Pathol
.
2014
;
38
(
7
):
990
998
.
173.
Stevers
M,
Rabban
JT,
Garg
K,
et al.
Well-differentiated papillary mesothelioma of the peritoneum is genetically defined by mutually exclusive mutations in TRAF7 and CDC42
.
Mod Pathol
.
2019
;
32
(
1
):
88
99
.
174.
Shrestha
R,
Nabavi
N,
Volik
S,
et al.
Well-differentiated papillary mesothelioma of the peritoneum is genetically distinct from malignant mesothelioma
.
Cancers
.
2020
;
12
(
6
).
175.
Xing
D,
Banet
N,
Sharma
R,
Vang
R,
Ronnett
BM,
Illei
PB.
Aberrant Pax-8 expression in well-differentiated papillary mesothelioma and malignant mesothelioma of the peritoneum: a clinicopathologic study
.
Hum Pathol
.
2018
;
72
:
160
166
.
176.
Lee
HE,
Molina
JR,
Sukov
WR,
Roden
AC,
Yi
ES.
BAP1 loss is unusual in well-differentiated papillary mesothelioma and may predict development of malignant mesothelioma
.
Hum Pathol
.
2018
;
79
:
168
176
.
177.
Karpathiou
G,
Hiroshima
K,
Peoc’h
M.
Adenomatoid tumor: a review of pathology with focus on unusual presentations and sites, histogenesis, differential diagnosis, and molecular and clinical aspects with a historic overview of its description
.
Adv Anat Pathol
.
2020
;
27
(
6
):
394
407
.
178.
Hissong
E,
Graham
RP,
Wen
KW,
Alpert
L,
Shi
J,
Lamps
LW.
Adenomatoid tumours of the gastrointestinal tract - a case-series and review of the literature
.
Histopathology
.
2022
;
80
(
2
):
348
359
.
179.
Itami
H,
Fujii
T,
Nakai
T,
et al.
TRAF7 mutations and immunohistochemical study of uterine adenomatoid tumor compared with malignant mesothelioma
.
Hum Pathol
.
2021
;
111
:
59
66
.
180.
Churg
A,
Le Stang
N,
Dacic
S,
et al.
Solid papillary mesothelial tumor
.
Mod Pathol
.
2022
;
35
(
1
):
69
76
.

Author notes

Supplemental digital content is available for this article at https://meridian.allenpress.com/aplm in the November 2024 table of contents.

Competing Interests

The authors have no relevant financial interest in the products or companies described in this article.

This work was presented at the International Mesothelioma Interest Group meeting; June 27, 2023; Lille, France.

Attanoos provides expert testimony in asbestos litigation for claimants/plaintiffs, defendants, and on a joint basis. Roggli and Beasley consult with attorneys representing plaintiffs and defendants in asbestos litigation.

Supplementary data