Thymic Epithelial Tumors and Benign Thymic Lesions

 

Relative frequencya

Age (y) range (average)

Gender (male:female)

Myasthenia Gravis (+)b range (mean)

Masaoka stage

I (%)

II (%)

III (%)

IVa (%)

IVb (%)

Type A

11.5% (3.1–26.2)

8–88 (64)

1:1.4

0–33 (17)

60

31

8

0.5

0.5

Type AB

27.5% (15–43.0)

11–89 (57)

1:1.4

6–42 (18)

67

26

6

1


Type B1

17.5% (5.9–52.8)

6–83 (50)

1:1.6

7–70 (44)

50

37

9

3

1

Type B2

26.0% (8.0–41.1)

4–83 (49)

1:1

24–71 (54)

32

29

28

8

3c

Type B3

16.0% (3.4–35.1)

8–87 (55)

1:0.8

25–65 (50)

19

36

27

15

3d

MNT2

1.0%

41–80 (65)

1.5:1

Rare

62

36


2


Metaplastic thymoma

<1.0%

28–71 (50)

1:1

Very rare

75

17

8




aAverage (range); bMNT, micronodular thymoma; c,dRanges 0–5% (B2) and 0–15% (B3)



TCs are also derivatives of thymic epithelial cells but histologically resemble carcinomas of other organs. This explains their “conventional” nomenclature (squamous cell carcinomas, adenocarcinomas, etc.). Neoplastic epithelial cells of TCs are usually devoid of thymic functions like the capacity for thymopoiesis. Accordingly, paraneoplastic autoimmune diseases are rare in TCs.



Tumor Heterogeneity, Proper Sampling, Reporting of Histology and Tumor Stage


Tumor heterogeneity due to different histological patterns in a single tumor, is common (up to 50%) in thymomas [4, 5]. Thymomas with WHO type B2 and B3 patterns are the commonest [5]. Heterogeneity is rare among TCs and TETs showing a thymoma and TC component(s) are even rarer [1, 2]. To detect heterogeneity, at least 1 block per cm maximal tumor diameter, preferably a minimum of 5 blocks, should be submitted for histological evaluation in resection specimens [6].

In resection specimens, the components in heterogeneous tumors should be reported in 10% increments. This rule does not apply to type AB thymomas that are, by definition, almost always composed of type A and type B-like components (and the latter should not be subclassified, e.g., as B1 or B2, either) [1, 2]. In heterogeneous thymomas as well as TCs, reporting should start with the most prevalent component, followed by the second most prevalent component, etc. However, if a given TET shows areas of thymoma and TC, reporting should always start with the carcinoma component, even if it is the minor component. Heterogeneous tumors with a small-cell carcinoma or large-cell neuroendocrine carcinoma component of whatever proportion should be labeled as “combined small-cell carcinoma” or “combined large-cell neuroendocrine carcinoma” followed by the other component(s). The common heterogeneity in TETs implies caution when reporting on small biopsies (see below).

Tumor stage of thymomas is reported following the Masaoka-Koga system [7] that reflects the fact that thymomas commonly exhibit pleural spread (stage IVa) while lymph node and extrathoracic metastasis (stage IVb) are rare (~2%). By contrast, lymph node and distant metastases are common in TCs (up to 40%). The Masaoka-Koga system is commonly used also for TCs but a UICC/AJCC approved TNM system will soon replace or complement the Masaoka-Koga system [7].


Epidemiology


Thymomas are rare tumors (incidence 1–4 per 106 per year) without clear gender bias. Although they rarely occur in children, their incidence peaks between 40 and 60 years of age [1, 2]. Thymic carcinomas are even rarer (incidence 0.1–0.4 per 106 per year), more common in men and occur with increasing frequency from adolescence onward [1, 2].


Etiology and Molecular Pathogenesis


The etiology of thymomas and thymic carcinomas is unknown. Rare familial [8] and syndromal thymoma cases (e.g., HNPCC) have been reported [9]. Thymoma patients show a higher frequency of nonthymic solid and hematopoietic cancers than the general population [10]. Since almost all thymomas show medullary and cortical differentiation [11] they are thought to arise from a bipotent thymic epithelial stem cell. In TCs, the (stem) cell of origin is enigmatic.

TETs show recurrent genetic gains and losses that increase from type A to B3 thymomas and further to TCs [12]. Genetic alterations in thymomas and TCs overlap partially [13]. Overlap with other cancers is limited [12]. The commonest chromosomal deletion of thymomas and TCs concerns the 6p25 region that contains the FOXC1 tumor suppressor gene [14]. The most common gains are at 1q23 (PBX1) and 8q24.3 (GLI4) [14, 15]. The commonest alteration overall is a point mutation in the GTF2I oncogene that is most prevalent in type A and AB thymomas (~80%), less common in type B thymomas (20–40%), rare in TCs (5–10%) and apparently absent in other cancers [16]. The only predictive biomarkers identified so far are activating KIT exon 11 gene mutations in 5% of TCs that correlated with transient responses to imatinib [17]. Markers predicting the response of many TCs to multikinase inhibitors are elusive [18].


Clinical Considerations


Thymoma and TC can cause pain, respiratory distress, vena cava superior syndrome or be fortuitous findings. Myasthenia gravis, pure red cell aplasia, acquired hypogammaglobulinemia (Good syndrome), and mucocutaneous candidiasis are “indicator autoimmune diseases” of thymomas but not of other cancers [3]. Therefore, their diagnosis should prompt a search for thymoma and their detection prior to surgery of a mediastinal mass makes the diagnosis of TC, lymphoma, germ cell, or mesenchymal tumor unlikely. Other thymoma-associated autoimmune diseases are not thymoma-specific, e.g., thyroiditis, type I diabetes, hepatitis, encephalitis, and SLE [19]. Autoimmune diseases in TCs (e.g., polymyositis) are very rare and not specific. Enlargement of mediastinal lymph nodes in association with a mediastinal mass argues against thymoma (see staging) and is in favor of a diagnosis of TC, lymphoma, or germ cell tumor (GCT).


Problems with Small Biopsies and Biopsy Indications—General Considerations


Biopsies of mediastinal masses are indicated if there are nonsurgical treatment options, e.g., in neoadjuvant settings or if lymphomas and germ cell tumors are differential diagnostic possibilities. Biopsies are usually dispensable in myasthenic patients, since thymoma is the only likely diagnosis and surgery is the curative therapy. Although small biopsies help to guide treatment [20], reporting of diagnoses based on small biopsies should be cautious for the following reasons:



  • The notorious tumor heterogeneity of thymomas may be missed in small biopsies and may cause discrepancies between a (homogeneous) biopsy and a (heterogeneous) resection specimen.


  • The highly proliferative cortical regions of normal or reactive thymus and lymphocyte-rich thymomas may be difficult to distinguish from each other and from T-lymphoblastic lymphoma.


  • Thymic epithelial hyperplasia, thymic cysts, or thymic Hodgkin lymphoma may mimic thymoma or TC.


  • The rare focal spillover of immature T cells from thymomas into mediastinal fat and the rare loss of keratin expression in thymomas may entail an erroneous diagnosis of T-lymphoblastic lymphoma.


  • Nuclear expression of p63 by tumor cells of primary mediastinal B-cell lymphoma (PMBL) is prone to be mistaken for thymoma or TC in small biopsies.


  • Many mimics of thymomas, e.g., synovial sarcoma resembling type A thymoma may be even more difficult to recognize in small biopsies than in resection specimens as detailed below.


Thymomas



Type A Thymoma, Including the “Atypical Type a Thymoma” Variant


Definition Type A thymoma is composed of bland spindle/oval neoplastic epithelial cells, with few or no admixed immature T cells. The atypical variant can display hypercellularity, increased mitotic activity and focal necrosis, of which the latter appears to be correlated with increased invasiveness.

Epidemiology and stage distribution of type A thymoma are summarized in Table 5.1. This usually indolent tumor has been delineated from “atypical type A thymoma”, i.e., a rare variant that infiltrates extensively, metastasizes, and shows histologic features of aggressiveness [1, 2].

Clinical peculiarities Due to the new definition as a tumor that is lymphocyte-poor throughout, type A thymoma is rarely associated with Myasthenia gravis (see above). Whether the new definition has an impact on the prevalence of other autoimmune diseases is not known.

Macroscopy Type A thymomas are usually encapsulated or well circumscribed (stage I or II). The cut surface is gray to white and usually shows a fascicular texture, few fibrous septae, and small cysts. Atypical type A thymoma may be poorly circumscribed, extend into adjacent organs, and exhibit necrotic areas and distant metastasis [2123].

Histology and immunohistochemistry Type A thymoma can show fascicular, storiform, glandular/adenoid, glomeruloid, solid, rosette-forming, hemangiopericytoma-like, and “pseudoendocrine (paraganglioma-like)” patterns, often within a single tumor. Bland spindle or oval cells with small, spindly, or oval nuclei with fine chromatin und without prominent nucleoli accompanied by thin-walled hemangiopericytomatous vessels are encountered at least focally in almost all properly sampled cases (Fig. 5.1a, b). Polygonal tumor cells are an optional and sometimes predominant feature (Fig. 5.1c–d). By definition, lymphoid cells are scarce (Fig. 5.1e, f). Mitoses, apoptotic cells, and perivascular spaces are uncommon. Coagulation necrosis (e.g., of comedo type) is usually restricted to “atypical type A thymomas” (Fig. 5.2). Hassall corpuscles are absent.

A352649_1_En_5_Fig1_HTML.gif


Fig. 5.1
WHO type A thymoma. a Conventional type A thymom composed of bland-looking spindle cells (right) adjacent to small polygonal cells (left). b Typical microcysts and single glands. c Paraganglioma-like pattern with polygonal tumor cells. d Solid pattern with polygonal tumors cells. e Focal increase of lymphocytes may require TdT staining to distinguish type A from AB thymoma. f Moderate number of immature, TdT(+) T cells; if more than 10% of the investigated tumor area reveals this density of TdT(+) T cells, a diagnosis of type AB thymoma should be rendered (HE: a, b, e ×100; c, d ×200; immunperoxidase: f ×100)


A352649_1_En_5_Fig2_HTML.gif


Fig. 5.2
Atypical WHO type A thymoma. a Comedo-type necrosis in atypical type A thymoma is the most consistent sign of clinical aggressiveness. b Comedo-type necrosis in a more polygonal cell area of atypical type A thymoma (pan-keratin, AE1/3). c P63 expression is typical of all thymoma subtypes, including atypical type A thymoma. d A low proliferation rate (Ki67 index <2% in epithelial cells) is typical of conventional type A thymoma but does not exclude atypical type A thymoma (HE: a ×50; immunperoxidase: bd ×100)

Immunohistochemistry Type A thymomas [1, 2] harbor no or rare immature T cells as identified by staining for TdT (recommended), CD1a, or CD99. Any dense (“crowded“) accumulation of immature T cells or moderate numbers of immature T cells (Fig. 5.1e, f) in >10% of the evaluable tumor area implies a diagnosis of type AB thymoma. Epithelial cells of type A thymoma consistently stain for CK19 und p63/p40, commonly for CD20 (focally) but not for CK20, CD5, and CD117.

Molecular pathology Recurrent structural genetic alterations are rare [13], while hot spot point mutations in the GTF2I gene show the highest prevalence (80%) among all thymomas [16].





  • Problems with small biopsies in type A thymoma
If only lymphocyte-poor (TdT−) areas of type AB thymomas and micronodular thymoma with lymphoid stroma (MNT) are sampled in a small biopsy, an erroneous diagnosis of type A thymoma may be inevitable. Fortunately, this is rarely of clinical relevance. Focal spindle cell areas in rare type B3 thymoma can be challenging: prominent perivascular spaces and Hassall corpuscles argue for B3 thymoma, while epithelial expression of CD20 and a ki67 index <2% [24] in epithelial cells are in favor of type A thymoma, leaving quite some tumors that may defy classification in small biopsies. While spindle cell (sarcomatoid) carcinoma is usually easily recognized by striking atypia, brisk proliferative activity and common CD5 and CD117 expression, spindle cell carcinoids, mesenchymal neoplasms (e.g., synovial sarcoma, solitary fibrous tumor, inflammatory myofibroblastic tumor, nerve sheath tumor), desmoplastic melanoma, various dendritic cell neoplasms [1] and even reactive changes (e.g., sclerosing mediastinitis, thymic fibrosis [25], inflammatory spindle cell tumors [26]) may need immunohistochemical analysis for recognition.

Therapy and prognosis Complete surgical removal is the only definite treatment and usually achievable due to the circumscription and low tumor stage of most classical “type A” thymomas. Removal may be challenging in “atypical” and metastatic cases [22, 27]. The 10-year survival rates reach 80–100% [5, 28, 29]. Survival rates for atypical type A thymomas have not been reported.


Type AB Thymoma


Definition Type AB thymomas are epithelial thymic tumors with a lymphocyte-poor (type A) and lymphocyte-rich (“type B-like”) component in highly variable proportions. The B-like component should not be subtyped (e.g., as B1-like or B2-like).

Epidemiology and stage distribution of this commonest thymoma subtype (up to 40%) [2830] are summarized in Table 5.1.

Clinical peculiarities Myasthenia gravis (occurring in 20–40%) and other paraneoplastic autoimmune diseases [1, 19] are frequent in this usually indolent tumor [27].

Macroscopy Most cases are well circumscribed [28]. On the cut surface, a nodular architecture and biphasic pattern with firm, whitish (lymphocyte-poor) areas separated by coarse septae from lymph node-like gray (lymphocyte-rich) regions is typical. “Atypical cases” with features that are analogous to those of “atypical type A thymoma” (i.e., advanced stage and necrosis) are rare (<5%) [23].

Histology and immunohistochemistry Fibrous septae that separate large tumor lobules are more common than diffuse and fibrosclerotic variants. The type A areas can exhibit the broad spectrum of type A thymoma morphologies, while tumor cells in the B-like areas are typically spindly, oval, or rarely polygonal with small nuclei and inconspicuous nucleoli and, thus, look different from tumor cells in type B1, B2, and B3 thymomas (Fig. 5.3a). Rarely, light staining medullary islands can occur in type AB thymomas. Type A and B-like areas may be sharply separated or show gradual transition. The proportions of type A and B-like areas are variable. Rarely, type AB thymomas are immature T-cell-rich throughout, i.e., type A areas and a biphasic pattern with lymphocyte-poor regions are not absolutely required for the diagnosis. Spindle-shaped tumor cells and a typical immunohistochemical profile (e.g., CD20 expression; see next) are helpful to recognize these challenging cases.

A352649_1_En_5_Fig3_HTML.gif


Fig. 5.3
WHO type AB thymoma. a Classical biphasic pattern with sharply delineated lymphocyte-poor spindle cell areas (type A, upper right) and lymphocyte-rich (type B-like) areas with barely recognizable tumor cells (lower left). b Keratin expression (AE1/3 antibody) of variable density in both areas (serial section). c CD20 expression in epithelial cells with spindly and dendritic morphology but not in small, round B cells (serial section). This pattern of CD20 positivity allows for the diagnosis of type AB thymoma even in apparently monophasic lymphocyte-rich cases. d Lymphoid cells in the lymphocyte-rich areas are mainly immature, TdT(+) T cells. Even small foci with this degree of “crowding” of TdT(+) T cells imply a diagnosis of type AB thymoma (compare with Fig. 5.1f) (HE: a ×100; immunoperoxidase: bd ×100)

Immunhistochemistry reveals that type AB thymomas are epithelial-rich tumors that show a dense but delicate network of tumor cells expressing various keratins (Fig. 5.3b) and p63/p40. Focal epithelial expression of CD20 occurs in 30–50% of cases (Fig. 5.3c), and spindle cells in septal structures often stain for EMA [31]. Complex expression of cortical and medullary markers is typical [11]. Lymphocytes mainly consist of immature, highly proliferative CD3+ TdT+ T cells (ki-67 index >90%) (Fig. 5.3d), but in medullary islands mature CD3+ TdT− Ki67low T cells prevail.

Molecular pathology Structural genetic alterations are more common in type AB than type A thymomas [15, 16, 32], while GTF2I mutations occur at the same high frequency (74–80% of cases) [16].

Problems with small biopsies in type AB thymoma. The commonest challenge is the distinction between lymphocyte-rich type AB, B1, and B2 thymomas. In type AB thymomas epithelial cells and their often oval or elongated nuclei are smaller and less conspicuous than in type B1 and B2 thymomas. Even minor spindle cell areas favor a diagnosis of type AB thymoma. While medullary islands occur in all lymphocyte-rich thymomas, Hassall corpuscles almost never occur in type AB thymomas but are frequent in B1 and not uncommon in B2 thymomas. Keratin stains highlight the consistently dense epithelial network in AB (and B2) thymomas, as compared to the delicate network in B1 thymomas. Epithelial CD20 expression is restricted to AB (and type A) thymomas. For the distinction between lymphocyte-poor type AB from type A thymoma and its mimics, see Fig. 5.1e, f. To separate biphasic AB thymomas from micronodular thymomas with lymphoid stroma (MNT), one has to recognize that the complex lymphoid component of MNTs (see below) is localized outside the epithelial component. Of note, heterogeneous tumors featuring type A or AB thymoma and an MNT component are quite common.

Therapy and prognosis. Complete resection is usually achieved and curative due to the common low tumor stage. The 10-year overall survival rates are over 80% [29, 33]. The impact of exceedingly rare metastases on survival has not been studied [22].


Type B1 Thymoma


Definition Type B1 thymomas resemble the normal childhood thymus in terms of abundance of immature T cells, paucity of epithelial cells, and absence of epithelial cell clustering. The “organoid pattern” with concurrence of prevailing cortical areas and medullary regions is another diagnostic prerequisite, while occurrence of Hassall corpuscles is optional.

Epidemiology and stage distribution of this infrequent thymoma subtype are given in Table 5.1.

Clinical peculiarities Myasthenia gravis is frequent (−45%), while other paraneoplastic autoimmune diseases, e.g., hypogammaglobulinema (Good syndrome) are rare (5%). “Pure” B1 thymomas usually follow an indolent clinical course and show stages I and II in over 80% of cases [5, 28, 33].

Macroscopy Most cases are well circumscribed and may reach large diameters due to late infiltration [28]. While the capsule may be firm, the cut surface is usually soft, and large tumor nodules may be separated by delicate or coarse fibrous septae.

Histology and immunohistochemistry In H&E stained sections, type B1 thymomas usually show little or no lobulation on low power. Dark cortical regions almost always dominate massively over lighter “medullary islands (MIs)” (Fig. 5.4a). In contrast to normal childhood medulla, MIs are not consistently “burried” in cortical areas but are commonly misplaced to the periphery of tumor lobules or along fibrous septae. Hassall corpuscles are common but not obligatory constituents of MIs. By definition, the frequency of epithelial cells must be similar to that of the normal thymus and must not show epithelial cell clustering (i.e., 3 or more contiguous tumor cells) as can be highlighted by p63 stain (Fig. 5.4b). Tumor cell nuclei resemble those of normal thymus, exhibiting round contours, vesicular chromatin, and variably prominent nucleoli. Keratin stains highlight a delicate epithelial network that is attenuated in MIs (Fig. 5.4c). In cortical regions, immature TdT+ T cells may obscure tumor cells. MIs are dominated by CD3+/TdT(−) mature T cells (Fig. 5.4d) and CD20+ B cells, and may harbor desmin-positive myoid cells and AIRE-positive epithelial cells.

A352649_1_En_5_Fig4_HTML.gif


Fig. 5.4
WHO type B1 thymoma. a Obligatory organoid architecture with predominance of dark staining cortical areas and obligatory light staining “medullary islands (MI)” (here without a Hassall’s corpuscle). b P63 expression in scattered, mostly single epithelial cell nuclei both in the cortex and MI. c Delicate network of keratin(+) (AE1/3 antibody) epithelial cells in cortical and medullary regions. d Confluent cells with TdT(+) nuclei in the cortex (HE: a ×50; Immunperoxidase: bd ×100)

Molecular pathology Genetic gains and losses are less frequent than in the more aggressive B2 and B3 thymomas [12, 34, 35]. The GTF2I mutation occurs in 32% of cases [16].





  • Problems with small biopsies in type B1 thymoma
In small biopsies, distinction of B1 thymomas from normal thymus, post-chemotherapeutic rebound hyperplasia, true thymic hyperplasia, B-like areas of AB thymoma, and T-lymphoblastic lymphoma (T-LBL) may be challenging or even impossible—requiring cautious reporting. In contrast to non-neoplastic thymus, excess of cortical over medullary areas, irregular and misplaced medullary islands (MIs), and lack of Hassall corpuscles may be clues to the diagnosis of B1 thymoma, while immunohistochemistry (ki67 index close to 100%; delicate epithelial cell network) is usually not helpful. Lymphocyte-rich areas of AB thymomas may be another challenge, particularly when MIs are present: spindle cells, a denser than normal keratin(+) epithelial cell network and epithelial CD20 expression hint to the diagnosis of AB thymoma (Fig. 5.3). As to T-LBL, the clinical setting, i.e., dramatic local and systemic symptoms and absence of Myasthenia gravis, is usually different. Atypical blastic infiltrates that destroy/attenuate keratin-positive epithelial networks and infiltrate (epithelial-free) mediastinal fat are characteristics of T-LBL. Immunohistochemistry in T-LBL may reveal antigen loss (e.g., of CD1a) or abnormal expression of Notch1 and/or LMO2 [36] in TdT+ lymphoblasts. However, in many T-LBLs, the immunohistochemical profile is normal. Clonality analyses may help to clarify such cases. Rarely, T-LBL develops in thymoma [37].

Therapy and prognosis Over 90% of type B1 thymomas are resectable. Recurrences and metastasis are rare [1]. The 10-year and 20-year survival rates is 85–100% [29, 33].


Type B2 Thymoma


Definition Type B2 thymomas are immature T-cell-rich tumors with a content of polygonal and dendritic neoplastic epithelial cells that is higher than in B1 thymomas and normal thymus. Spindle tumor cells are absent. Cortical differentiation is slightly reduced and medullary features are often strongly attenuated in B2 compared to B1 thymomas [11].

Epidemiology and stage distribution are given in Table 5.1. Occurrence in children is rare [11].

Clinical peculiarities Myasthenia gravis (MG) occurs in up to 50% of cases [5, 28], pure red cell aplasia and hypogammaglobulinemia (with or without accompanying MG) in 5%. Pleural effusions and the SVC syndrome are more common than in A, AB, and B1 thymomas.

Macroscopy Type B2 thymomas are often only partially encapsulated and infiltrate adherent soft tissue and pleura, lung, heart, or large vessels. The cut surface is gray or white and may show poor septation, necrosis, hemorrhage, and cysts.

Histology and immunohistochemistry Fibrous septae typically delineate small tumor lobules, and dominant lymphocytes impart a dark or blue impression (Fig. 5.5a, b). Tumor cell nuclei have distinct membranes, vesicular chromatin, and prominent nucleoli (Fig. 5.5b). Perivascular spaces with palisading of tumor cells (Fig. 5.5c) and pale medullary islands (MIs) with or without Hassall corpuscles are optional findings. Intratumorous lymphoid follicles are common in MG-associated cases (Fig. 5.5d). Neoplastic epithelial cells are either dispersed but more numerous than in normal thymus or B1 thymoma, or occur in clusters of at least three contiguous epithelial cells that can be highlighted by p63 immunohistoochemistry (Fig. 5.5e). Fourty percent of B2 thymomas show a minor B3 or B1 thymoma component (Fig. 5.5e). Tumor cells express many keratins (e.g., CK5/6 and CK19) and p40/p63 but are negative for CK20. Most lymphocytes are immature, highly proliferative CD3+ CD5+ CD4+ CD8+ TdT+ T cells (Fig. 5.5f).

A352649_1_En_5_Fig5_HTML.gif


Fig. 5.5
WHO type B2 thymoma. a Typical lobular architecture and plump invasion into mediastinal fat. b Obligatory blue appearing, lymphocyte-rich tumor with highly increased number of neoplastic epithelial cells (when comparedwith cortical regions of normal thymus); epithelial cells with vesicular nuclei and distinct nucleoli. c Lymphocyte-rich, blue appearing tumor with perivascular space (arrow). d Type B2 thymoma with (optional) medullary island that harbors a lymphoid follicle with germinal center (GC) close to a Hassall’s corpuscle (HC); lymphoid follicles often hint to thymoma-associated Myasthenia gravis. e, f Tumor heterogeneity in thymoma: WHO typ B2 thymoma components on the left, type B1 thymoma component on the right; increased number of focally clustered, p63(+) epithelial cells (arrows) in the B2 component (e); very high density of immature, TdT(+) T cells in the B1 component (f) (HE: a ×25, b ×400; c, d ×100; immunoperoxidase: e, f ×200)

Molecular pathology The number of genetic alterations in B2 thymomas is intermediate between AB thymomas on the one hand, and B3 thymomas on the other hand. B2 and B3 thymomas share losses of 6q25.2–q25.3 and 3p and gains of 1q [12, 34, 35, 38]. 22% show GTF2I mutations [16].

Problems with small biopsies in B2 thymomas Type B2 thymomas must be distinguished from the less aggressive B1 thymomas and from T-LBL. Small tumor lobules, more abundant than normal and clustered polygonal epithelial cells, and a dense keratin(+), p63(+) epithelial network are hints that the diagnosis is type B2 but not B1 thymoma. Cytologically atypical, TdT+ lymphoblasts that destruct and efface the normal thymic epithelial cell network is the hallmark of T-LBL. A pitfall in this respect is the rare (usually partial) loss of keratin expression in B2 thymomas [39]. Since epithelial expression of p40/p63 is almost always maintained in keratin(−) thymomas, it is recommended to stain for p40/p63 whenever the differential diagnosis between thymoma and T-LBL is problematic.

Therapy and Prognosis Complete surgical removal is achieved in 70–90% of cases [29, 34, 40]. The 10-year recurrence rates reach 32 and 41% in completely resected Masaoka stage II and III B2 thymomas, respectively, arguing for adjuvant radiotherapy in stage III thymomas [41, 42]. Postoperative radiotherapy is also used for incompletely resected thymomas, although it is usually not curative [43]. Nonresectable thymomas may be successfully treated by multimodal concepts [44]. The 10-year survival rates of B2 thymoma patients reach 70–90% [5, 28, 29].


Type B3 Thymoma


Definition Type B3 thymomas are epithelial-predominant tumors that harbor a minor number or (rarely) no immature T cells. Neoplastic epithelial cells have largely lost cortical differentiation and medullary features are generally missing [11].

Epidemiology and stage distribution are given in Table 5.1. Occurrence in children is rare [45].

Clinical peculiarities Myasthenia gravis occurs in 40–50% of cases, other paraneoplasias are rare [28]. Since advanced tumor stages occur in 50% of cases, thoracic pain and superior vena cava syndrome are common at presentation.

Macroscopy Type B3 thymomas are often poorly circumscribed due to infiltration into adjacent soft tissue and organs. On the firm, gray/white cut surface, hemorrhage, necrosis, and cysts are common.

Histology and immunohistochemistry Fibrous septae separate tumor lobules that are composed of sheets of polygonal neoplastic epithelial cells. The sheets of tumor cells impart a pink impression on low power (in contrast to the blue hue that is one of the hallmarks of B2 thymomas) (Fig. 5.6a). Plump, sharply delineated lobules of tumor cells form the invasion front of B3 thymomas (Fig. 5.6a), while single-cell infiltration and prominent desmoplasia are more common in thymic carcinoma. Tumor cell nuclei can be varied: bland looking with inconspicuous nucleoli or moderately atypical with anisonucleosis and prominent nucleoli. Sheets of tumor cells are usually sprinkled with few lymphocytes (Fig. 5.6b). Rarely lymphocytes are absent. Perivascular spaces surrounded by palisades of tumor cells are usually conspicuous (Fig. 5.6b). Hassall corpuscles occur rarely. Focal occurrence of spindle cells and clear cell features may occur (Fig. 5.6c, d). Diffuse epithelial expression of keratins (e.g., CK19, CK5/6, CK8/18) (Fig. 5.6e) and p40/p63, and focal expression of GLUT1 and EMA are typical [1], while CD20 is absent (in contrast to many type A and AB thymomas). Focal CD5 and CD117 expression has been described as rare exception. The lymphocytes are mainly TdT+ immature T cells (Fig. 5.6f). Perivascular spaces may harbor mature and immature T cells and plasma cells.

A352649_1_En_5_Fig6_HTML.gif


Fig. 5.6
WHO type B3 thymoma. a Typical lobular architecture and plump invasion front facing desmoplastic stroma. b Pink appearing, epithelial-rich, lymphocyte-poor tumor with minor atypia and prominent perivascular space (PVS). c Small focus of spindle cells; minor spindle cell foci against a background of typical B3 thymoma are compatible with the diagnosis of B3 thymoma. d Focal clear cells features are common in otherwise typical (pink) B3 thymomas. e Keratin(+) sheets of tumor cells with palisading around epithelial-free perivascular spaces (antibody AE1/3). F Paucity of TdT(+) non-neoplastic immature T cells among neoplastic epithelial tumor cells (HE: a, d ×25; b, c ×200; immunoperoxidase: e, f ×200)

Molecular Pathology Type B3 thymomas show the highest prevalence of genetic alterations among thymomas [12, 15, 34, 35]. Changes that are largely restricted to B3 thymomas comprise losses of 13q, 16q, and 17p and gains of 4p and 17q [15, 16, 34, 35, 46]. GTF2I mutations occur in 21% of cases [16].

Problems with small biopsies Distinction of a relatively lymphocyte-rich (but still pink appearing) B3 thymoma from a relatively lymphocyte-poor (but still blue appearing) B2 thymoma is inevitably subjective (but clinically of minor relevance). Tumors showing both B3 and B2 patterns are more common than pure B3 thymomas, and small biopsies often fail to sample this tumor heterogeneity. Therefore, we comment on this possibility if we encounter pure B3 and B2 thymomas in small biopsies. Rare B3 thymomas can show focal spindle epithelial cells, raising the differential diagnosis of type A thymoma. This distinction may be impossible if the focal and inconsistent CD20 expression of type A thymomas, or prominent perivascular spaces of B3 thymomas are missing in small biopsies. The distinction of B3 thymomas from thymic squamous cell carcinomas (TSQCC) is clinically relevant and difficult since well-differentiated TSQCC may have perivascular spaces, and small biopsies may fail to sample the single cell or finger-like infiltration pattern at the invasion front. Prominent intercellular bridges, strong and usually extensive expression of CD5 and CD117 (in 80% of TSQCCs), and absence of TdT+ immature T cells favor a diagnosis of TSQCC [1]. Whenever a suspected “B3 thymoma” shows unusual features, alternative diagnoses should be seriously considered (e.g., melanoma; seminoma; carcinoid; synovial sarcoma; parathyroid adenoma).

Therapy and Prognosis Therapeutic principles are the same as in B2 thymomas (see above). 30% of B3 thymomas recur within 10 years [28]. The 10-year overall survival rates are 50–70% [5, 29]. MG-associated cases may have a better prognosis due to earlier detection [40, 47].


Micronodular Thymoma with Lymphoid Stroma (MNT)


Definition MNTs are biphasic tumors composed of nodules of neoplastic, bland-looking spindle epithelial cells that are surrounded by a non-neoplastic, epithelial-free lymphoid stroma.

Epidemiology and stage distribution of these rare thymomas are given in Table 5.1.

Clinical peculiarities Paraneoplastic phenomena, including Myasthenia gravis (MG), are virtually nonexistent in MNTs. 95% are in stage I or II and fortuitous findings.

Macroscopy Most MNTs are encapsulated or well circumscribed and commonly contain cysts on the soft, gray cut surface.

Histology and immunohistochemistry Multiple, small, and sharply delineated tumor nodules are composed of type A thymoma-like, bland spindle and (rarely) polygonal epithelial cells that may form cysts and rosettes (Fig. 5.7a, b). Inside the nodules there are few or no lymphocytes. Outside the epithelial compartment, the lymphoid stroma is dense and usually shows lymphoid follicles. Some TdT+ immature T cells typically surround the nodules and are accompanied by mature, CD3+ T cells, CD20+ B cells, and lymphoid follicles containing CD23+ follicular dendritic cells. Rarely, monoclonal B cells and low-grade intratumorous B-cell lymphomas occur inside MNTs [48]. In contrast to many type A and AB thymomas, epithelial cells of MNTs are CD20(−).

A352649_1_En_5_Fig7_HTML.gif


Fig. 5.7
Miconodular thymoma with lymphoid stroma (MNT). a Single or confluent nodules of pink appearing spindel cells are surrounded by a lymphocyte-rich, desmoplasia-free stroma including a lymphoid follicle (F). b Nodules are composed of bland-looking spindle and oval cells with inconspicuous nucleoli. c Keratin expression (antibody AE1/3) in the nodules; no expression in the lymphoid stroma. d CD20 expression in a lymphoid follicle within the lymphoid stroma; note typical absence of CD20 expression in epithelial cells of MNT e. BCL2(−) reactive germinal center (GC). f TdT(+) immature T cells occur in the lymphoid stroma close to but barely inside epithelial nodules (HE: a ×25; b ×200; immunoperoxidase: c ×25; df ×100)

Molecular pathology Recurrent genetic alterations have not been reported.

Problems with small biopsies MNT can be mixed up with type AB thymoma, if the lack of epithelial cells in the T-cell- and B-cell-rich compartments of MNT is not appreciated. AB thymomas are epithelial-rich throughout and largely devoid of B cells. Lymphoid follicles in epithelial cell-free areas of MNT may prompt the diagnosis of thymic follicular hyperplasia (TFH) in small biopsies. However, TFH is almost always associated with MG, while MNT almost never is. Low-grade B-cell lymphomas inside MNTs may be impossible to distinguish from primary thymic (e.g., MALT) lymphomas if the characteristic nodules of MNT are missing in small biopsies. Imaging studies may give a hint: MNTs are usually well circumscribed, while conventional thymic lymphomas show fuzzy borders. Micronodular thymic carcinoma with lymphoid hyperplasia [49] shows the same growth pattern as MNT but tumor cells are clearly atypical and TdT+ immature T cells are absent.

Therapy and Prognosis Surgical removal is almost always possible. Recurrences and tumor-related deaths have not been reported.


Metaplastic Thymoma (MPT)


Definition MPTs are biphasic tumors showing solid areas of epithelial cells in addition to a bland, spindle cell (metaplastic) component.

Epidemiology and stage distribution are given in Table 5.1.

Clinical peculiarities MPTs have not been reported to be associated with Myasthenia Gravis (MG) or other autoimmune diseases. Almost all MPTs show low tumor stages and are fortuitous findings.

Macroscopy MPTs are usually well circumscribed with a homogenous gray to white cut surface.

Histology and immunohistochemistry MPTs show no lobular growth pattern. The polygonal or plump spindly cells of the epithelial component form islands or anastomosing trabecular structures, while the spindle cell component resembles bland fibroblasts growing in storiform or fascicular patterns (Fig. 5.8a). The transition between both components can be sharp or gradual. The epithelial cells show mildly atypical nuclei and small nucleoli. Lymphocytes are usually absent. The epithelial component is strongly positive for keratins, p63, and usually EMA, while the fibroblast-like component may express keratins and EMA faintly and focally and is p63(−) (Fig. 5.8b) [1, 2]. CD5, CD20, CD34, and CD117 are not expressed and TdT+ T cells are absent. Proliferative activity is mostly low (ki67 index < 5%).

A352649_1_En_5_Fig8_HTML.gif


Fig. 5.8
Metaplastic thymoma and other rare thymomas. a Metaplastic thymoma exhibiting epitheloid tumor cell islands surrounded by bland-looking spindle-shaped tumor cells. b Nuclear p63 expression exclusively in the epithelioid tumor component. Note moderate degree of nuclear atypia in the epithelioid component. In case of substantial mitotic activity and spindle cell atypia, the differential diagnosis of sarcomatoid carcinoma needs consideration. c So-called “microscopic thymoma” fortuitously detected in a thymectomy specimen resected for Myasthenia gravis. The cells of this supposedly hyperplastic epithelial proliferation resemble those of type A thymoma and are devoid of interepithelial immature T cells. d Sclerosing thymoma (large mediastinal mass): Collagen-rich fibrous stroma surrounds‚ islands’ of epithelial cells. As is the case here, the underlying cause, supposedly a regressively changed thymoma, cannot be classified with certainty (HE: a, c, d ×50; b ×200; immunoperoxidase)

Problems with small biopsies Distinction of MPTs from sarcomatoid thymic carcinomas, biphasic synovial sarcomas, and solitary fibrous tumors (SFTs) may be difficult. Striking atypia and high proliferative rate are typical of sarcomatoid carcinomas, while the immunoprofiles of synovial sarcoma (p63(−), TLE1+) and SFT (p63(−), CD34+, CD99+, BCL2+, nuclear STAT6+) are distinctive.

Therapy and prognosis Complete surgical removal is the treatment of choice. Recurrences are exceedingly rare and a single tumor-related death has been reported [1, 2].


Rare Other Thymomas


So-called “microscopic thymomas” are tiny (<1 mm) epithelial nodules that are found fortuitously on histological examination of non-neoplastic thymuses. The nodules are composed of bland-looking plump spindle and polygonal cells and are typically devoid of immature T cells (Fig. 5.8c). They are of no clinical relevance and need to be separated from “microthymomas” that represent small conventional thymomas measuring <1 cm in diameter [50].

So-called “sclerosing thymoma” is not an entity but a mediastinal mass that mostly seems to represent the fibrous end stage of a regressing thymoma. In most cases, the histotype of the underlying thymoma is not recognizable (Fig. 5.8d). Rarely, sclerosing thymomas seem to result from enigmatic collagen production in perivascular spaces and in the outskirts of conventional thymomas (usually type B2 or B3) [1]. Sclerosing thymoma must be distinguished from sclerosing mediastinitis, diffuse thymic fibrosis [25], sclerosing lymphomas, germ cells tumors, and mesenchymal tumors [1].



Thymic Carcinomas (TCs)


TCs are derivatives of thymic epithelial cells like thymomas but morphologically resemble analogously labeled carcinomas in other organs. There are no subtype-specific clinical peculiarities and tumor stage and resection status (R0 vs. R1/R2) but not histological subtype appear to have an impact on prognosis [5153]. Therefore, TCs will not be described in detail here but typical diagnostic pitfalls and problems with small biopsies in TCs will be highlighted. Since neoplastic epithelial cells of TCs are devoid of thymus-specific functions (e.g., the capacity to support thymopoiesis), TCs are almost never accompanied by Myasthenia gravis and other autoimmune and immunodeficiency diseases that are typical of thymomas. Instead, pain and other local symptoms are common at presentation. The distinction of thymomas from TCs is clinical relevant:



  • The prognosis of TCs is worse than the prognosis of thymomas [29, 51].


  • Lymph node metastases are common (30%) in TCs but rare in thymomas (2%) [54].


  • Distant (hematogenous) metastases are frequent in TCs (50%) while pleural dissemination is the most common type of metastasis in thymomas, and extrathoracic spread is rare [55].


  • Therapeutic management is different in TCs and thymomas. Even completely resected stage II TCs should receive postoperative radiotherapy and rare TCs with activating KIT mutations may be candidates for second-line imatinib treatment. In contrast to thymomas, a subset of otherwise refractory TCs profits from multikinase inhibitor therapy [28, 5658].

Genetic complexity is higher in TCs than thymomas [12, 16, 38, 59]. Despite considerable mutational overlap, some alterations are largely restricted to TCs: gains on 17q and 18p; losses on 3p, 16q, and 17p; and mutations with impact on epigenetics and apoptosis [14, 60, 61]. The most common mutations in TCs concern TP53 (40%), and members of the PI3 K and RAS pathway [60, 62, 63], while mutations of GTF2I are rarer (8%) than in thymomas (20–80%) [16]. Interestingly, thymic mucoepidermoind carcinomas (MECs) show the same MAML2 rearrangement as MECs in other organs [64], while mutational profiles of squamous cell carcinomas (SQCCs) of thymus and lung are very different [12]. This reflects the etiological role of smoking in lung but not thymic SQCCs.


Thymic Squamous Cell Carcinoma (TSQCC)


TSQCC make up 80% of TCs, are more common in males, and typically present in patients over 50 years of age [53, 63]. If Myasthenia gravis is present, this usually hints to a rare thymoma component [65]. 90% of cases are in advanced tumor stages at presentation [66]. Histology can be quite varied (Fig. 5.9). Morphological peculiarities of TSQCCs are: (i) structures resembling perivascular spaces; however, optically empty spaces around central vessels that are typical of thymomas are often effaced by fibrosis; (ii) plump instead of infiltrative invasion in many well-differentiated cases; (iii) epithelial cells expressing CD5 in 60% and CD117 (Fig. 5.10) and FOXN1 in 70–80%; (iv) frequent expression of neuroendocrine markers in a minority (<50%) of tumor cells; (v) rare fully encapsulated (stage I) “cystic well-differentiated TSQCCs” with excellent prognosis [67]. A molecular peculiarity is “targetable” KIT mutations in 5–10% of cases [13]. The 10-year survival rate is 60–65% [53, 68].

A352649_1_En_5_Fig9_HTML.gif


Fig. 5.9
Spectrum of thymic squamous cell carcinoma (TSQCC). a Solid growth pattern of keratinizing TSQCC. b Nonkeratinizing TSQCC. c TSQCC mimicking B3 thymoma due to prominent palisading of tumor cells around vessels; in contrast to optically empty or lymphocyte-filled “true” perivascular spaces, the perivascular zones in TSQCC are typically obliterated by variably collagen-rich fibrosis and commonly harbor plasma cells. d Focal spindling of tumor cells in TSQCC; in case of extensive spindling, the differential diagnoses of spindle cell carcinoma (i.e., a variant of sarcomatoid carcinoma) or of “combined thymic carcinoma” need consideration. e Trabecular growth pattern of TSQCC contrasting with the lobular growth pattern of type A or B3 thymoma. f Infiltrative growth at the invasion front is typical of TSQCC but not thymoma; however, the “pushing border” type of invasion that is characteristic of thymoma occurs in some TSQCC as well (HE: ×200)


A352649_1_En_5_Fig10_HTML.gif


Fig. 5.10
Immunohistochemical profile of thymic squamous cell carcinoma (TSQCC). a Cytokeratin 5/6 (CK5/6) expression is a must and usually extensive. b Epithelial CD5 expression. c Epithelial CD117 expression. d Nuclear p40/p63 positivity in virtually all tumor cells. This “full house pattern” occurs in 60–80% of TSQCC and is almost pathognomonic for a thymic primary. In case of mediastinal CD5-/CD117-squamous cell carcinomas, clinicopathological correlation is needed to distinguish TSQCC from mediastinal involvement by extramediastinal squamous cell carcinomas (e.g., from the lung). If squamous differentiation is lacking, expression of CD5 and/or CD117 is not per se indicative of a thymic origin of a given carcinoma, since nonsquamous cancers from many organs can be CD5+ and CD117+ (Immunoperoxidase, ×100)

Problems with small biopsies and differential diagnosis The strategy to distinguish TSQCC from B3 thymomas has been detailed in the “B3 thymoma chapter”.

TSQCC can be accompanied by dense lymphoid and plasma cell-rich infiltrates, raising the possibility of lymphoepithelioma-like carcinoma (LELC). Since both cancer types can be CK5/6+, CD5+, and CD117+, negativity on EBER in situ hybridization (ISH) and solid instead of syncytial tumor complexes would support diagnosis of TSQCC.

Poorly differentiated carcinomas resembling TSQCC should routinely be checked for EBV association using EBER-ISH. By definition, EBER+ carcinomas are counted among LELCs, even if they resemble SQCCs and completely lack lymphoid stroma (see below) [1].

TSQCCs that show transition to undifferentiated or small-cell carcinoma components should be stained for neuroendocrine markers to exclude “combined large-cell neuroendocrine carcinoma” or “combined small-cell carcinomas” of the thymus; and for NUT protein to exclude the aggressive “NUT carcinoma” [69]. In biopsies the poorly differentiated component of NUT carcinoma can be missed. Therefore, in young patients, centrally located tumors and/or TSQCCs in small biopsies should be tested for NUT expression. To distinguish poorly differentiated TSQCC from undifferentiated carcinoma, staining for CK5/6, p63, and CD5 is required. By definition, all these staining are negative in undifferentiated carcinomas of the thymus [1].

Distinguishing TSQCC from metastases of pulmonary or head-and-neck SQCCs is straightforward in small biopsies, if the TSQCC is CD5+ CD117+ FOXN1+. In SQCCs that are negative for these markers (20% of TSQCCs) imaging studies are needed to resolve this differential diagnostic problem.


Basaloid Carcinomas


Basaloid thymic carcinomas (BTCs) make up 5% of TCs, are more common in males, and typically occur in elderly patients. 60% show stage III and IV disease at presentation [1]. Striking palisading of tumor cells at the periphery of tumor infiltrates is the hallmark of BTCs and necrosis is common (Fig. 5.11a–c). Large, cystic tumors with papillary, nested, or “cylindromatous” growth patterns are frequent. Tumor cells usually are quite monotonous giving a blue or clear cell impression. Keratinization and transition to sarcomatoid components may occur [70]. Like TSQCC most BTCs express CK5/6, p63, CD5, and CD117 (Fig. 5.11d) and are TTF1(−) [70]. The prognosis is similar as in TSQCC.

A352649_1_En_5_Fig11_HTML.gif


Fig. 5.11
Basaloid carcinoma of the thymus. a Typical solid growth pattern of basophilic tumor cells with high nuclear/cytoplasmic ratio showing palisading at the periphery of tumor lobules and around vessels. b High power view of palisading of tumor cells. c Frequent mitoses, apoptotic bodies, and necrotic areas are characteristic. d CD5 expression is at least as common as in thymic squamous cell carcinomas and—in the absence of extensive neuroendocrine features—argues against a diagnosis of large-cell neuroendocrine carcinoma (HE: a, c ×50; b ×200; Immunoperoxidase: d ×200)

Problems with small biopsies and differential diagnoses Distinction of BTC from poorly differentiated TSQCC may be difficult in small biopsies. Atypia in TSQCCs is usually more striking. NUT carcinoma needs consideration and staining for NUT protein in cases with sharp transition between keratinizing and small-cell areas, particularly in young patients [69] might be helpful. Adenoid cystic carcinoma-like tumor usually shows some cribriform areas, less palisading than BTC and is usually CD117(−) [71].


Lymphoepithelioma-Like Carcinoma (LELC)


Thymic LELCs make up 5% of TCs, are more common in males and often occur in children and young adults. 70% are in stage III or IV at presentation. Syncytial growth of poorly delineated cells with large nuclei, vesicular chromatin, and prominent nucleoli in a lymphoid and plasma cell-rich stroma with no or little desmoplasia is typical (Fig. 5.12). Keratinization is rare [53, 72]. 50% of classical, lymphocyte-rich cases are EBV-associated, particularly those in patients under 30. By definition, EBV(+) carcinomas without lymphoid stroma are also called LELCs, even if they look like TSQCC or undifferentiated carcinoma (Fig. 5.13a) [1, 2]. EBV detection needs EBER in situ hybridization (EBER-ISH) due to insensitivity of LMP1 immunohistochemistry. Tumor cells typically express keratins, p63, CD5, and CD117 (Fig. 5.13b–d). Lymphocytes are mature, TdT(−) T and B cells. Plasma cells are polyclonal. The prognosis is similar as in TSQCC.
Oct 14, 2017 | Posted by in PATHOLOGY & LABORATORY MEDICINE | Comments Off on Thymic Epithelial Tumors and Benign Thymic Lesions

Full access? Get Clinical Tree

Get Clinical Tree app for offline access