What's New in Soft Tissue Tumor Pathology?

2021-02-15 病理維基百科

SPECIAL ARTICLE

The 2020 WHO Classification

What’s New in Soft Tissue Tumor Pathology?

Michael E. Kallen, MD* and Jason L. Hornick, MD, PhD†

Abstract: The fifth edition of the World Health Organization Classification of Tumors of Soft Tissue and Bone was published in early 2020. The revisions reflect a consensus among an inter- national expert editorial board composed of soft tissue and bone pathologists, geneticists, a medical oncologist, surgeon, and ra- diologist. The changes in the soft tissue tumor chapter notably include diverse, recently described tumor types (eg, atypical spindle cell/pleomorphic lipomatous tumor, angiofibroma of soft tissue, and CIC-rearranged sarcoma), new clinically significant prognostic information for a variety of existing entities (eg, de- differentiated liposarcoma and solitary fibrous tumor), and a plethora of novel genetic alterations, some of practical diagnostic relevance (eg, NAB2-STAT6 in solitary fibrous tumor, FOSB rearrangements in epithelioid hemangioma and pseudomyogenic hemangioendothelioma, and SUZ12 or EED mutations in ma- lignant peripheral nerve sheath tumor, leading to loss of H3K27 trimethylation). In this review, we highlight the major changes to the soft tissue chapter in the 2020 World Health Organization Classification, as well as the new chapter on undifferentiated small round cell sarcomas, with a focus on updates in diagnostic categories, prognostication, and novel markers. Recent discov- eries in molecular genetics are also discussed, particularly those of immediate utility in differential diagnosis, including protein correlates detectable using immunohistochemistry.

Key Words: sarcoma, soft tissue tumors, solitary fibrous tumor,

NAB2-STAT6 fusion, malignant peripheral nerve sheath tumor,

H3K27me3

(Am J Surg Pathol 2020;00:000–000)

he new fifth edition of the World Health Organization (WHO) Classification of Tumors of Soft Tissue and Bone was published in early 2020,1 7 years following publication of the fourth edition.2 The revisions reflect a consensus among an international expert editorial board composed of soft tissue and bone pathologists, geneticists,


a medical oncologist, surgeon, and radiologist. Updates  to the classification reflect extensive new genetic data,  prompting expansion and reorganization of several categories. The soft tissue tumor chapter in particular in- cludes the addition of recently described tumor types; in some cases, increasing outcome data and comparative genetic studies argued for splitting into separate tumor  types or changes in nomenclature and managerial categories. Despite the expanding contribution of genetics to our understanding of the  molecular  pathogenetic  basis for soft tissue tumors, the new classification empha- sizes the continued central diagnostic importance of morphology.

In addition to a discussion of 「new」 soft tissue tu- mor entities, this review will highlight updated prognostic information for familiar tumor types, such as dediffer- entiated liposarcoma (DDLPS) and solitary fibrous tumor (SFT). Finally, we will include a focused discussion of the dizzying array of recently described genetic alterations in soft tissue tumors. The past decade has seen a plethora of novel gene fusions in rare soft tissue tumor types; a comprehensive and in depth examination of these alter- ations is beyond the scope of this review. Instead, we will focus on selected genetic alterations with practical diag- nostic relevance. An example is the undifferentiated small round cell sarcoma chapter; detection of novel gene fu- sions has led to subclassification of the former wastebasket of 「Ewing-like」 sarcomas into new categories with distinct presentations, clinical behavior, and morphologic and immunohistochemical (IHC) features. Additional prac- tical examples include the identification of recurrent ge- netic alterations in SFT and malignant peripheral nerve sheath tumor (MPNST), leading to the introduction of highly sensitive and specific IHC markers, which are now in routine use in surgical pathology.

NEW SOFT TISSUE TUMOR TYPES

The fifth edition of the WHO Classification features

                         「new」 soft tissue tumor types that were not found in the

From the *Department of Pathology, University of Maryland School of Medicine, Baltimore, MD; and †Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA.

Conflicts of Interest and Source of Funding: J.L.H. is a consultant to Epizyme, Aadi Biosciences, and TRACON Pharmaceuticals. For

M.E.K. none was declared.

Correspondence: Jason L. Hornick, MD, PhD, Department of Pathology, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115 (e-mail: jhornick@bwh.harvard.edu).

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.


fourth edition (Table 1). As was the case for prior editions, tumors are organized by line of differentiation, such as adipocytic, fibroblastic/myofibroblastic, vascular, and smooth muscle neoplasms. Several emerging tumor types, including the morphologically heterogenous NTRK-rearranged spindle cell neoplasms, are contained within the section on tumors of uncertain differentiation. Epithelioid hemangioendothelioma (EHE) with YAP1-TFE3 fusion will be discussed in the novel


Am J Surg Pathol • Volume 00, Number 00, 』』 2020              www.ajsp.com | 1

TABLE 1. 2020 WHO Classification: New Soft Tissue Tumor Types

Section/Chapter               Tumor Type

Adipocytic tumors                Atypical spindle cell/pleomorphic

lipomatous tumor


differential diagnosis includes spindle cell/pleomorphic lipoma and ALT/WDLPS. Spindle cell/pleomorphic  lipomas have a different anatomic distribution, are well circumscribed, and contain characteristic brightly eosinophilic,  coarse  「ropy」  collagen  fibers. ALT/WDLPS

rarely  involves  the  subcutaneous  compartment  but shows

Fibroblastic/myofibr- oblastic tumors

Vascular tumors Smooth muscle tumors Tumors of uncertain

differentiation Undifferentiated small

round cell sarcomas of bone and soft tissue


Myxoid pleomorphic liposarcoma Angiofibroma of soft tissue

EWSR1-SMAD3-positive fibroblastic tumor (emerging)

Superficial CD34-positive fibroblastic tumor

EHE with YAP1-TFE3

Inflammatory leiomyosarcoma*

NTRK-rearranged spindle cell neoplasm (emerging)

CIC-rearranged sarcoma

Sarcomas with BCOR genetic alterations Round cell sarcomas with EWSR1-non-

ETS fusions


similar histologic appearances as atypical spindle cell lipomatous tumors, although a prominent spindle cell component is uncommon; IHC or fluorescence in situ hybridization (FISH) can be helpful to make this distinction. By IHC, the tumor cells in atypical spindle cell lip- omatous tumors show variable expression of CD34, S100 protein, and desmin, whereas MDM2 and CDK4 are gen- erally negative (rarely, weak or focal staining for the latter markers may be seen). Spindle cell lipomas show diffuse CD34 expression, but by contrast are usually negative for desmin and S100 protein.10 Notably, loss of nuclear RB1 expression is observed in 50% to 70% of atypical spindle cell

*Formerly included as variant of (conventional) leiomyosarcoma; now separate

section.


lipomatous tumors, in keeping with chromosome 13q14

                         deletion, including the RB1 locus, which is also a consistent

finding  in  spindle  cell/pleomorphic  lipomas.11,12  MDM2

genetic changes section below. Also discussed here are 「new」 tumor types within the undifferentiated small round cell sarcoma chapter.

Atypical Spindle Cell/Pleomorphic Lipomatous Tumor

Atypical spindle cell/pleomorphic lipomatous tumor is a benign adipocytic neoplasm, distinct from atypical lipomatous tumor/well-differentiated liposarcoma (ALT/ WDLPS) and spindle cell/pleomorphic lipoma, although it shares some histologic features with both of these other tumor types.3–5 The largest series of atypical spindle cell lipomatous tumors reported a male-to-female ratio of 3:2 and a median age of 54 years.6 Approximately two thirds of tumors occur in the limbs and limb girdles, with a predilection for the hands and feet, and an approximately equal distribution between superficial and deep sites. Less commonly involved are the head and neck, genitals, and trunk, with very rare retroperitoneal involvement. By contrast, ALT/WDLPS occurs equally commonly in the retroperitoneum and limbs, less commonly in the sper- matic cord, mediastinum, and head and neck. Spindle cell/ pleomorphic lipomas characteristically occur in the pos- terior aspect of the neck or upper back, less often the face, orbital region, and oral cavity, with few reports of deep- seated lesions in a broader anatomic distribution.

Histologically, atypical spindle cell lipomatous tumor shows infiltrative margins and demonstrates a range of appearances, with varying proportions of spindle cells, adi- pocytes, and lipoblasts, including hyperchromatic and sometimes pleomorphic cells (atypical pleomorphic lip- omatous tumor).7 The histology may be viewed as a spec- trum flanked by 2 morphologic extremes: a paucicellular pattern with bland spindle cells, minimal nuclear atypia, adipocytes, and abundant (often myxoid) matrix, and a hy- percellular pattern at the other end of the spectrum with mild-to-moderate cytologic atypia, easily identifiable lipoblasts, and less abundant matrix (Fig. 1).6,8,9 The


amplification is absent in atypical spindle cell lipomatous tumor, in distinction from ALT/WDLPS.

Importantly, atypical spindle cell/pleomorphic lip- omatous tumors pursue a benign clinical course with a low rate of nondestructive local recurrence (10% to 15%), lower than the risk for ALT/WDLPS; in contrast to ALT/ WDLPS, atypical spindle cell lipomatous tumor has no potential to dedifferentiate.6

Myxoid Pleomorphic Liposarcoma

Another 「new」 adipocytic tumor type, myxoid pleomorphic liposarcoma is an exceptionally rare and aggressive neoplasm occurring in children and young adults with a predilection for the mediastinum.13 Some cases are associated with Li-Fraumeni syndrome (ie, germline TP53 mutation).14–16 Histologic features include an admixture of bland zones resembling conventional myxoid liposarcoma (including scattered lipoblasts, a delicate capillary vascular network, and myxoid pools) and cellular areas resembling pleomorphic liposarcoma with marked nuclear atypia and pleomorphic lipoblasts (Fig. 2).17 These tumors lack both FUS/EWSR1- DDIT3 fusions (of conventional myxoid liposarcoma) and MDM2 amplification (of well-differentiated and DDLPS).18,19 Myxoid pleomorphic liposarcomas are clinically aggressive, with a high rate of local recurrence and distant metastasis and poor outcomes.

Angiofibroma of Soft Tissue

Angiofibroma of soft tissue is a benign fibroblastic neoplasm that typically arises in the lower extremities, often involving or adjacent to large joints, uncommonly in the back, abdominal wall, pelvic cavity, and breast. A series of 37 cases reported a male predominance, a median age of 49 years, and a slightly more frequent subcutaneous rather than deep/subfascial situation.20 Histologically, angiofibroma of soft tissue contains uniform bland spindle cells, variably myxoid and collagenous stroma, and

2 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

FIGURE 1. Atypical spindle cell lipomatous tumor. A, This tumor shows a variably collagenous to myxoid stroma and is dominated by spindle cells, in areas admixed with adipocytes. B, The tumor cells are relatively uniform, whereas the adipocytes show marked variation in size, including occasional lipoblasts. C,  This  tumor  contains  abundant  myxoid  stroma  with  prominent  adipocytes and scattered spindle cells with hyperchromatic nuclei. D, This example shows dense collagenous stroma, notably pleomorphic    and multinucleated cells, and occasional lipoblasts.

a prominent vascular network comprised of small thin-walled branching blood vessels, similar to (although thicker in caliber than) those in myxoid  liposarcoma  (Fig. 3). Cytologic atypia is generally absent, other than degenerative atypia in occasional cases.

Angiofibroma of soft tissue features the recurrent translocation t(5;8)(p15;q13), resulting in the fusion of aryl hydrocarbon receptor repressor (AHRR) at 5p15 with nuclear receptor coactivator 2 (NCOA2) at 8q13 in most

cases.21 AHRR had not previously been implicated in gene fusions, whereas NCOA2 is the 3′ partner in fusions with MYST3 and ETV6 in leukemias, and with PAX3 and HEY1 in sarcomas.21 The AHRR-NCOA2 fusion is found in 60% to 80% of angiofibromas of soft tissue. Variant GTF2I-NCOA2 and GAB1-ABL1 fusions have been re- ported in few cases.22,23

Of note, the similarly named cellular angiofibroma is entirely different from angiofibroma of soft tissue, in its inguinoscrotal/vulvar location and distinct and less


uniform histologic features.24,25 Genetic findings in cel- lular angiofibroma include losses of RB1, also seen in spindle cell/pleomorphic lipoma, atypical spindle cell/ pleomorphic lipomatous tumor (discussed above), and myofibroblastoma, with overlapping morphologic features shared among these tumor types in the so-called 13q/RB1 family of tumors.26

EWSR1-SMAD3-positive Fibroblastic Tumor (Emerging)

This provisional entity is a benign fibroblastic neoplasm, with recent reports describing a wide age range, female predilection, and distribution largely in the hands and feet, in superficial (dermal or subcutaneous) locations.27,28 Histologic features include nodular growth, a pattern of zonation dem- onstrating peripheral areas of intersecting cellular fascicles comprised of bland spindle cells (Fig. 4), and sometimes central areas of hyalinization and infarction.27 IHC reveals diffuse ERG expression, whereas SMA and CD34 are

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.              www.ajsp.com | 3

FIGURE 2. Myxoid pleomorphic liposarcoma. A, The tumor contains abundant myxoid stroma and 「crow’s feet」 blood vessels, mimicking myxoid liposarcoma; the presence of tumor cells with large, hyperchromatic nuclei would not fit with myxoid liposarcoma. B, In other areas, markedly pleomorphic cells and atypical lipoblasts resemble pleomorphic liposarcoma.

typically negative. This tumor type is defined by fusion of exon 7 of EWSR1 with exon 5 of SMAD3; SMAD3 is an important signal transducer in the TGF-β/SMAD pathway, involved in extracellular matrix synthesis by fibroblasts. This benign neoplasm may recur locally following incomplete excision.

Superficial CD34-positive Fibroblastic Tumor

This distinctive low-grade neoplasm of the skin and subcutis is rare, with few reported cases. Most have oc- curred in middle-aged adults, with a slight male predom- inance, and a predilection for the lower extremities, followed by upper extremities, buttock, and shoulder. Histology demonstrates highly cellular fascicles and sheets of spindled to epithelioid cells with abundant eosinophilic, often glassy cytoplasm and moderate-to-marked pleo- morphism (Fig. 5). There is morphologic overlap reported with PRDM10-rearranged soft tissue tumors.29 By IHC, there is strong CD34 expression (Fig. 5B), and keratins are positive in 70% of cases. The prognosis in reported cases has been excellent.30,31


Inflammatory Leiomyosarcoma

Inflammatory leiomyosarcoma is a rare low-grade sarcoma that was originally buried within the 「inflammatory malignant fibrous histiocytoma」 wastebasket, and previously included as a variant of leiomyosarcoma (in the fourth edition of the WHO Classification); owing to marked differences in clinical behavior and genetics, inflammatory leiomyosarcoma has now been designated a separate entity in the fifth edition. Inflammatory leiomyosarcoma has a peak incidence in young adults and occurs in the soft tissues of the extremities and trunk.32 The histologic features are notable for a prominent inflammatory infiltrate obscuring the neoplastic cells, com- prised of foamy histiocytes and prominent lymphocytes; the tumor cells are arranged in fascicles and are typically rela- tively uniform with mild atypia and eosinophilic cytoplasm (Fig. 6). Cytogenetic studies have identified a distinctive characteristic near-haploid genotype,33,34 with or without subsequent whole genome doubling. Gene expression studies have also identified a distinct signature with prominent expression of genes involved in muscle development and function.35

4 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

FIGURE 3. Angiofibroma of soft tissue. A, The tumor shows a hypocellular appearance with evenly distributed, branching blood vessels and a fine collagenous stroma. B, Some examples contain myxoid stroma. Note the prominent thin-walled blood vessels. C, The uniform ovoid to short spindle cells contain fine chromatin. D, The characteristic blood vessels are generally thicker in caliber than similarly shaped vessels of myxoid liposarcoma.

The differential diagnosis can present a challenge in these cases due to the striking inflammatory infiltrate, with some histologic resemblance to other tumors including inflammatory myofibroblastic tumor and DDLPS, as well as idiopathic mass-forming disorders such as sclerosing mesenteritis, idiopathic retroperitoneal fibrosis, and Ig- G4-related disease (depending upon the anatomic site).

Although only small numbers of cases have been  reported, and follow-up data are limited, the prognosis appears to be favorable, in contrast to the high rate of metastasis and often poor prognosis of conventional leiomyosarcoma.36

NTRK-rearranged Spindle Cell Neoplasm (Emerging)

This provisional category includes a group of rare soft tissue tumors defined by NTRK gene fusions, en- compassing a wide spectrum of histologies and clinical behavior (ranging from benign neoplasms to aggressive sarcomas); infantile fibrosarcoma is excluded from this


category.37 These tumors frequently demonstrate monot- onous spindle cell morphology, infiltrative growth, and co- expression of CD34 and S100 protein by IHC.38 This category includes the recently described lipofibromatosis- like neural tumor39 and tumors that resemble peripheral nerve sheath tumors; many of these tumor subsets most often affect children.40,41 The varied histologic features include haphazard and infiltrative monomorphic spindle cell morphology, a solid pattern with stromal bands and perivascular rings of keloid-like collagen (Fig. 7),42 a pattern with zonation resembling MPNST, and a particularly uncommon pattern with a myopericytoma- like architecture. In addition to CD34 and S100 protein co-expression, tumors retain H3K27me3 (in contrast to many MPNSTs), and the majority are reactive with a pan- TRK monoclonal antibody,43,44 although the latter shows imperfect specificity45; tumors with NTRK1 fusions show the most consistent and strong staining (Fig. 7D).

Most tumors in this category harbor NTRK1 fu- sions, with a variety of partners, although rare NTRK2

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.              www.ajsp.com | 5

FIGURE 4. EWSR1-SMAD3-positive fibroblastic tumor. A, Intersecting fascicles of spindle cells show irregular margins with adjacent subcutaneous adipose tissue. B, The fibroblastic spindle cells contain uniform nuclei and pale eosinophilic cytoplasm.

and NTRK3 fusions have also been reported; those re- sembling peripheral nerve sheath tumors may instead harbor RAF1 or BRAF fusions. NTRK fusions form chi- meric proteins containing aberrant oncogenic receptor tyrosine kinases (TRK-A, TRK-B, or  TRK-C),  which  are therapeutic targets; detection of NTRK fusions is currently mandatory for systemic therapy. The prognosis appears to be related to histologic grade, although many NTRK-rearranged spindle cell neoplasms are clinically benign, and firm criteria for malignancy have yet to be established.

Undifferentiated Small Round Cell Sarcomas of Bone and Soft Tissue

Ewing sarcoma, and the growing family of un- differentiated, predominantly round cell sarcomas that lack EWSR1 (or FUS) rearrangements (formerly des- ignated 「Ewing-like」 sarcomas), are aggressive sarcomas that predominantly affect children and young adults but may also occur in older adults. Recent molecular genetic discoveries and clinicopathologic studies have allowed subclassification of tumors within the former wastebasket


of undifferentiated/unclassified round cell sarcomas into new distinct entities in the 2020 WHO: round cell sarco- mas with EWSR1-non-erythroblast transformation spe- cific (ETS) fusions, CIC-rearranged sarcomas, and sarcomas with BCOR genetic alterations. In addition, the Ewing sarcoma section has been pulled out from the bone tumor chapter of the fourth edition of the WHO Classi- fication, and grouped with the other aforementioned sar- comas in a new chapter 「undifferentiated small round cell sarcomas of bone and soft tissue」 in the 2020 WHO Classification, to reflect a more accurate biological un- derstanding of the complex genomic landscape of this updated category. Finally, there remains a small subset of round cell sarcomas that are as yet unclassifiable.

The substantial overlap in morphology and clinical features, but divergent biological behavior and very low incidence, present unique diagnostic problems for these sarcoma types, and mandate ancillary IHC and/or molecular testing and a multimodal approach that is challenging  outside  of  specialized  referral  centers.  The broad morphologic differential diagnosis for this group of tumors also includes rhabdomyosarcomas

6 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

FIGURE 5. Superficial CD34-positive fibroblastic tumor. A, The tumor is composed of sheets of variably pleomorphic and epithelioid cells with abundant, glassy eosinophilic cytoplasm. Mitotic activity is minimal, in contrast to undifferentiated pleomorphic sarcoma. B, IHC for CD34 is diffusely positive. Keratins are also often positive (not shown).

(especially embryonal and alveolar), as well as poorly differentiated (round cell) synovial sarcoma, small cell  osteosarcoma, mesenchymal chondrosarcoma, schwan- nian stroma-poor or undifferentiated neuroblastomas, Wilms tumor, and desmoplastic small  round  cell  tumor.46 Here we will  discuss  the  main  subcategories  of undifferentiated small round cell sarcomas, with a focus on novel genetic alterations and their histologic/ immunophenotypic differences, as well as clinical im- plications.

Ewing sarcoma is the second most common sarcoma in children,  usually  affecting  young  males  aged  10  to  25 years, although it infrequently occurs in older adults. The entity now encompasses a spectrum of clinical presentations and morphologic appearances, including the now obsolete 「peripheral primitive neuroectodermal tumor」 (PNET; this designation was used when rosettes were apparent histolog- ically).47 Extraskeletal Ewing sarcoma usually occurs in the thigh, followed in descending frequency by pelvis, paraspinal area, and foot. The histology of conventional Ewing sarco- ma demonstrates sheets of closely packed though evenly spaced uniform small round cells, with finely granular


chromatin and inconspicuous nucleoli. The 「large cell」 or 「atypical」 variant of Ewing sarcoma has no im- munophenotypic differences or prognostic implications, al- though a subset of these cases have now been reclassified into CIC-DUX4 sarcomas, as will be further discussed. IHC features include CD99 and FLI1 expression, which are highly sensitive but not specific for Ewing sarcoma (FLI1 in particular is positive in a subset of a wide range of other tumor types); strong, diffuse membranous CD99 is almost invariably seen in Ewing sarcoma. Identification of NKX2-2, a downstream target of EWSR1-FLI1 signaling, provided an IHC marker that is highly sensitive and relatively specific for Ewing sarcoma (although it also labels mesenchymal chon- drosarcoma and olfactory neuroblastoma).48–50

Approximately 85% of Ewing sarcomas have EWSR1-FLI1 fusions, with the remainder showing EWS- R1-ERG fusions (10%), and rare cases harboring fusions between EWSR1 and other members of the ETS tran- scription factor family.51 Rare Ewing sarcoma cases can instead harbor a FUS instead of EWSR1 rearrangement, with an ERG or FEV fusion partner.51 Establishment of one of these alterations by FISH (or alternative methodologies

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.              www.ajsp.com | 7

FIGURE 6. Inflammatory leiomyosarcoma. A, The tumor is composed of relatively uniform, elongated spindle cells with eosinophilic cytoplasm, including scattered atypical forms, admixed with prominent lymphocytes and scattered foam cells. B,  Desmin  is  typically strongly positive.

such as reverse transcription-polymerase chain reaction or next-generation sequencing) may be necessary for selection of appropriate therapy; the prognosis has improved with multimodality therapy to a near 80% 5-year overall survival. CIC rearrangements were identified in primitive mesen- chymal tumors as early as several decades ago52,53; these al- terations are the most common abnormalities in undifferen- tiated round cell sarcomas that lack EWSR1 fusions, com- prising at least 70% of such cases.54 By far the most common CIC rearrangement partner is DUX4 (95% of cases), followed by FOXO4, as well as rare cases with partners including LEUTX, NUTM1, and NUTM2A. Most CIC-rearranged sarcomas arise in young adults and are extraskeletal, favoring the  deep  soft  tissues of  the  extremities  and trunk, with rare tumors presenting in bone and visceral sites. Histologic distinctions from Ewing sarcoma include heterogeneity in nu- clear size and shape; prominent nucleoli; variably abundant, sometimes clear cytoplasm; focal spindle cell or epithelioid features;  and  focally  myxoid stroma  (Fig.   8). By IHC, the majority of cases show patchy CD99 staining; useful findings include extensive ETV4 and WT1 nuclear expression (Figs. 8E, F)50,55–57; these are particularly helpful given the


potential for false negativity with FISH testing using CIC break-apart probes (ie, 15% of cases show cryptic rearrangements).58,59 CIC-rearranged sarcomas pursue an aggressive clinical course,60 with a series of 115 cases reporting a 5-year overall survival of 43%, significantly inferior to that of Ewing sarcoma.56

Sarcomas with BCOR genetic alterations were more recently reported61 and represent ∼5% of non- EWSR1 or FUS rearranged 「Ewing-like」 sarcomas. BCOR fusion partners include CCNB3 (by far the most common), as well as MAML3, ZC3H7B, and unknown

partners62; other cases show BCOR internal tandem du- plication. These tumors share a similar anatomic dis- tribution as Ewing sarcoma, predominantly affecting the long bones or pelvis of adolescents, but show a striking male predominance and appear to pursue a less ag- gressive clinical course than Ewing sarcoma.63,64 The histologic features include variably round to spindled cells, with some cases dominated by spindle cell mor- phology; the individual cells contain uniform nuclei with fine chromatin and scant cytoplasm (Fig. 9). Focal areas of  myxoid  or  collagenous  stroma  are  commonly seen,

8 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

FIGURE 7. NTRK-rearranged spindle cell neoplasms. A, This group of tumors shows a range of histologic appearances and sometimes resembles nerve sheath tumors, ranging from (A) hypocellular tumors with bland spindle cells in a collagenous stroma  to (B) more cellular examples with mitotically active, atypical spindle cells in a myxoid stroma. Note the perivascular hyalinized collagen, similar to the vascular pattern observed in schwannomas. C, Some tumors show a focally nodular or whorled appearance. D, IHC using a pan-TRK antibody is usually positive, most strongly and diffusely in tumors with NTRK1 rearrangements. Weak or moderate staining is not entirely specific for this tumor class.

often with a delicate capillary vasculature. By IHC, nuclear BCOR expression has been shown to be highly sensitive (though only moderately specific), regardless of fusion partner (Fig. 9D), with additional frequently positive markers including cyclin B3 (CCNB3), cyclin D1, SATB2, and CD99 (the latter with  patchy  staining).65 Notable pitfalls include histologic mimics  that share BCOR expression such as poorly differentiated synovial sarcomas66 and SFT,67 as well as positivity in genetically similar tumors including clear cell sarcoma of the kidney and primitive myxoid mesenchymal tumor of infancy. Nuclear CCNB3 expression is seen in cases with BCOR-CCNB3 fusions. In a recent series of 36 cases, unsupervised clustering by RNAseq indicated  that  BCOR altered tumors form a tight genomic group  distinct from Ewing sarcoma and CIC-rearranged sarcomas.68

A final category is that of round cell sarcomas har- boring EWSR1 fusions partnered to non-ETS family


members; emerging categories are the EWSR1-PATZ1 and EWSR1-NFATC2 sarcomas.69 Limited numbers of cases have been reported. Features of EWSR1-PATZ1 sarcomas include an equal sex distribution, broad age range, predi- lection for deep soft tissues of the chest wall, abdomen, and limbs, and potentially aggressive behavior.69–71 Their histology shows rounded to spindle cells forming sheets and nests, with a dense fibrous or myxohyaline stroma, which sometimes mimic myoepithelial tumors (Fig. 10), particularly given S100 protein and SOX10 expression,  or nerve sheath tumors; other IHC findings include desmin, myogenin, and myoD1 positivity in some cases (such tumors have also been referred to as 「polyphenotypic sarcomas」), variable CD99 expression, and keratin negativity. EWSR1-NFATC2 sarcomas have a marked male predilection, affect both children and adults, and generally occur in the metaphysis or diaphysis of long bones72–76; histology demonstrates round, epithelioid, and/ or spindle cells forming cords, nests, and trabeculae, with

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.              www.ajsp.com | 9

A            B

C            D

E            F

FIGURE 8. CIC-rearranged sarcoma. A, This tumor is composed of sheets of round cells somewhat resembling Ewing sarcoma, although the amount of cytoplasm and uneven spacing of nuclei would be unusual. B, The tumor cells often contain prominent nucleoli and abundant pale cytoplasm, helpful clues to the diagnosis. C, Epithelioid tumor cells with clear cytoplasm are a focal finding in many cases. D, This example contains an admixture of  rounded to  epithelioid cells  with amphophilic cytoplasm. Note  the extensive necrosis. The combination of nuclear WT-1 (E) and ETV4 (F) is helpful to support the diagnosis.

hyalinized or myxohyaline stroma. IHC features include CD99 expression in 50% of cases, as well as variable NKX2-2 and PAX7, and focal, often dot-like keratin


expression. Limited outcome data describe some patients cured by resection, but with the potential for late metastasis.

10 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

FIGURE 9. Sarcomas with BCOR genetic alterations. A, This tumor is composed of uniform round cells with fine chromatin in a fibrillary collagenous stroma. B, Some examples contain myxoid stroma and thin-walled blood vessels. C, A component of short spindle cells is often seen. D, Diffuse nuclear staining for BCOR is a characteristic finding, albeit not entirely specific for tumors with BCOR gene alterations.

NEW CONCEPTS IN NOMENCLATURE, GRADING, AND RISK STRATIFICATION

In addition to the new additions to the classification, recent information has also led to clinically significant updates in familiar entities, with prognostic and ther- apeutic implications (Table 2). Examples span the spectrum from common to rare tumors. Although a comprehensive discussion is beyond the scope of this review, selected important examples are included below.

Malignant Melanotic Nerve Sheath Tumor

This tumor type had been widely known as melanotic schwannoma; however, new nomenclature has been in- troduced, given increasing evidence of aggressive clinical behavior. These rare lesions are comprised of Schwann cells with melanocytic differentiation and are frequently asso- ciated with Carney complex, an autosomal dominant tu- mor predisposition syndrome. Malignant melanotic nerve sheath tumor (MMNST) most often arises from spinal or autonomic nerves of adults, with an earlier age of onset


seen in patients with Carney complex. These tumors affect up to 8% of individuals with Carney complex; the majority harbor biallelic inactivating mutations in PRKAR1A on 17q24, which encodes a tumor suppressor. Gene expression analysis has shown a transcriptional signature distinct from both conventional schwannomas and melanomas.77

Histology demonstrates sheets of uniform plump spindled to polygonal or epithelioid cells with eosinophilic to amphophilic cytoplasm, round to ovoid nuclei, nuclear grooves and pseudoinclusions, and coarse to fine melanin pigment which can be sufficiently abundant as to obscure the nuclear morphology; some tumors feature prominent nu- cleoli and a high mitotic rate (Fig. 11). Psammoma bodies are a common finding that impart no clinical significance. By IHC, S100 protein, SOX10, and melanocytic markers including HMB45, melan A, and tyrosinase are positive; these immunophenotypic findings are indistinguishable from melanoma. PRKAR1A expression is often lost, in sporadic tumors and those from patients  with  Carney  complex  (Fig. 11D). Awareness of this lesion can facilitate

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.              www.ajsp.com | 11

FIGURE 10. Round cell sarcomas with EWSR1-PATZ1 fusions. A, This tumor contains an admixture of round cells and epithelioid cells. Note the bland nuclei. B, Some tumors show a reticular growth pattern with short spindle cells in a more myxoid stroma.        C, The histology of these tumors is often reminiscent of myoepithelial neoplasms. Note the perivascular hyalinization. D, IHC for S100 protein is often positive, further mimicking a myoepithelial tumor.

separation from melanoma, which despite the similar immunoprofile, is not generally as uniform  cytologically; the typical anatomic sites of MMNST are also a helpful clue to the diagnosis.

The clinical behavior of MMNST cannot be pre- dicted based on morphologic features.78 Although in the past this tumor type was thought to be generally benign


with occasional cases pursuing an aggressive clinical course, recent evidence has shown a high rate of malignant behavior, including a large series of 40 cases with local recurrences in 35% and metastases in 44% of patients.79

Dedifferentiated Liposarcoma

DDLPS is a relatively common sarcoma type that arises most often in the retroperitoneum, with other sites

                         including spermatic cord, and more rarely the media-

TABLE 2. 2020 WHO Classification: New Concepts in Nomenclature, Grading, and Risk Stratification of Soft Tissue Tumors

Tumor Type               What’s New?              

MMNST                Change in nomenclature (formerly melanotic

schwannoma) to reflect aggressive clinical behavior DDLPS                Recognition of adverse prognostic impact of high

FNCLCC grade, as well as myogenic (particularly rhabdomyoblastic) differentiation

SFT                Prognostic model predicting metastatic risk, using patient age, mitotic rate, tumor size, and necrosis


stinum, head and neck, and trunk. DDLPS shares amplification of MDM2 and CDK4 with ALT/WDLPS, with additional abnormalities more frequently found in DDLPS than in ALT/WDLPS, including amplification of JUN, TERT, CPM, and MAP3K5, and deletion of ATRX, ATM, CHEK1, ABTB16, PPP2R1B, and EI24.80

The histologic hallmark of DDLPS is transition from ALT/WDLPS to a nonlipogenic sarcoma, which is usually but not always high grade, and can be of variable extent within the tumor. The transition between well- differentiated and dedifferentiated areas is usually abrupt,

12 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

FIGURE 11. MMNST. A, Some examples contain abundant melanin pigment,  obscuring the  tumor cells. B,  This tumor contains less prominent melanin. The uniform spindle cells show a fascicular architecture. C, Some tumors show a melanoma-like appearance with epithelioid cells with prominent nucleoli and a high mitotic rate. D, By IHC, many examples show loss of staining  for PRKAR1A (red reaction product).

but can be more gradual, and rarely shows intermingling. A well-differentiated component is not always identifiable. Dedifferentiated areas most frequently resemble un- differentiated pleomorphic sarcoma, or less commonly an intermediate-to-high-grade myxofibrosarcoma, although the histologic spectrum is broad, occasionally including an unusual meningothelial-like whorling pattern.

Cases of 「low-grade」 dedifferentiation have increas- ingly been recognized, most often with uniform spindle cells and mild cytologic atypia, and a cellularity intermediate between WDLPS and conventional DDLPS; in these cases, the distinction between WDLPS and DDLPS may be challenging.80 Although DDLPS is usually nonlipogenic, occasional cases contain scattered lipoblasts in the otherwise undifferentiated component, or sheets of pleomorphic lip- oblasts resembling a pleomorphic liposarcoma; this phe- nomenon is referred to as homologous lipoblastic differen- tiation.81 FISH demonstrating MDM2 amplification (or  IHC showing diffuse nuclear expression of MDM2 and CDK4) is useful in separating these rare cases of DDLPS


from pleomorphic liposarcoma; the latter is associated with much higher metastatic potential. Heterologous differ- entiation may also be seen in DDLPS, including rhabdo- myoblastic and chondro-osseous differentiation, and rarely foci of angiosarcoma.

The prognosis of DDLPS is better than that of other high-grade pleomorphic sarcomas, with an overall mor- tality of 28% to 30% at 5-year follow-up. As reported in the fourth edition of the WHO, the extent of dediffer- entiated areas does not seem to predict outcome. It was historically believed that the morphologic appearances and histologic grade of dedifferentiated areas did not im- pact prognosis; however, the fifth edition includes updated information showing an adverse prognostic impact asso- ciated with high-grade tumors based on the French Féd- ération Nationale des Centres de Lutte Contre le Cancer (FNCLCC) grading system. In a retrospective review of 148 patients with retroperitoneal WDLPS and DDLPS, Mussi et al82 demonstrated a significant negative prognostic impact of high FNCLCC grade on both

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.              www.ajsp.com | 13

disease-specific and event-free survival. In a study of 119 primary retroperitoneal DDLPS cases, Keung et al83  found that FNCLCC grade predicted overall survival, as did extent of resection and tumor integrity. In addition, Jour et al84 identified FNCLCC grade as an independent prognostic factor predicting local recurrence in a study of 50 cases of DDLPS, with grade 3 tumors demonstrating worse local recurrence-free survival than grade 1 and 2 cases. Furthermore, recent studies have indicated that myogenic, particularly rhabdomyoblastic differentiation is associated with a worse outcome in DDLPS.85,86

Solitary Fibrous Tumor

SFT is an anatomically ubiquitous fibroblastic neo- plasm; uniformly cellular examples were formerly known as hemangiopericytoma. SFT occurs most commonly in adults with no gender predilection and is most common in deep somatic soft tissue and at body cavity sites (especially pleura, pelvis, and retroperitoneum); 「extrapleural」 has been removed from the official designation in the 2020


WHO Classification. The histopathology demonstrates a wide spectrum of findings, classically characterized by haphazardly arranged spindled to ovoid cells, with branching, thin-walled staghorn-shaped blood vessels and prominent stromal collagen (Fig. 12). Fat-forming, giant cell-rich, and dedifferentiated variants may be encountered. By IHC, diffuse nuclear STAT6 expression is characteristic of SFT (see below) (Fig. 12D).

A critical genetic update for SFT is the discovery of a pathognomonic NAB2-STAT6 fusion, resulting from a paracentric inversion within chromosome 12q.87–89 The fusion is thought to convert wildtype NAB2 from a transcriptional repressor of EGR1-mediated signaling into a transcriptional activator, via replacement of the C-ter- minal repression domain by the transcriptional activation domain of STAT6, resulting in constitutive activation of proliferation and survival-associated growth factors including IGF2 and FGFR1. The proximity of  NAB2 and STAT6 makes detection of fusions difficult by conventional cytogenetics or FISH testing; however,

FIGURE 12. SFT. A, The tumor contains prominent stromal collagen and shows a haphazard architecture. B, This tumor shows a more cellular appearance. Note the scattered dilated, thin-walled blood vessels. C, A highly cellular example with prominent staghorn vessels. This high-risk tumor had a high mitotic rate (not shown). D, Nuclear immunoreactivity for STAT6 is a consistent finding, reflecting the underlying NAB2-STAT6 fusion that defines this tumor type.

14 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

Am  J Surg  Pathol  •  Volume  00, Number  00, 』』 2020              2020 WHO Classification of Soft Tissue Tumors

STAT6 IHC is both a highly sensitive and specific surro- gate for fusions.90,91 Additional alterations in the TERT promoter and TP53 have been associated with aggressive behavior and dedifferentiation.92,93

The clinical course of SFT has been notoriously difficult to predict based on histologic features.94 Although the majority of tumors pursue a benign course, 5% to 10% recur or metastasize, typically to lungs, liver, and bone, including occasional cases with benign- appearing histology. Traditional criteria for malignancy included  large  size,  dissemination  at presentation, pleo-

morphism, necrosis, and a mitotic rate of ≥ 4 per 10 high power fields, the latter being the most reliable criterion

for malignancy in the past. Overtly malignant SFTs demonstrate a 20% to 30% metastatic rate. The un- predictable  relationship  between  histology  and clinical


RECENTLY IDENTIFIED GENETIC ALTERATIONS

Recent years have seen an explosion of publications describing novel recurrent genetic alterations in soft tissue tumor types, including many that are included for the first time in the 2020 WHO Classification (Table 4). Abnormalities of particular interest are those with immediate practical diagnostic or clinical implications98; examples for endothelial neoplasms will be described here, including FOS and FOSB fusions in epithelioid hemangiomas, YAP1-TFE3 variant of EHE, and SERPINE-FOSB and ACTB-FOSB fusions in pseudomyogenic hemangioendothelioma. A major advance to improve the reproducibility of the diagnosis of MPNST is the discovery of SUZ12 and EED  mutations,  and  resultant loss of H3K27 trimethylation (H3K27me3), which can be demonstrated  by IHC,  as detailed  below.  An important step

behavior has led in recent years to the development of                       

risk stratification models. A 3-tiered model established by Demicco et al95 incorporated age (below 55 or 55 y and

above), tumor size  (< 5  to  ≥ 15 cm  in  increments  of 5 cm), and mitotic count (0, 1 to 3, or  ≥ 4/10 high power


TABLE 4. 2020 WHO Classification: Recently Identified Genetic Alterations in Soft Tissue Tumors

Section               Tumor Type               New Genetic Alterations

fields). This model was subsequently validated in 79 cases

of SFT; the addition  of  necrosis  as  a  fourth  criterion (<< span=""> 10% or ≥ 10%) increased the percentage of patients


Fibroblastic/

myofibroblas- tic tumors


Fibrous hamartoma of

infancy Calcifying fibrous

tumor


EGFR mutations

FN1-EGF

correctly defined as low risk and more precisely identified

a high-risk  group  with rapid  development of metastasis


Lipofibromatosis                FN1-EGF; other receptor

tyrosine kinase or EGFR

(Table 3).96 The French Sarcoma Group has also published a risk calculator, using  patient  age,  tumor  site, mitotic count, and history of radiotherapy to predict local and metastatic recurrences; their findings also confirmed the not infrequent occurrence of delayed relapses 10 to 20 years following primary excision, highlighting the need for long-term follow-up of SFT.97


Pericytic/


ligand fusions Dermatofibrosarcoma COL6A3-PDGFD,

protuberans                EMILIN2-PDGFD

SFT                NAB2-STAT6

Myxoinflammatory                BRAF fusions

fibroblastic sarcoma

Infantile fibrosarcoma EML4-NTRK3; NTRK1,

NTRK2, BRAF, and MET

fusions

Glomus tumor                MIR143-NOTCH1/2/3

TABLE 3. Risk Stratification of Solitary Fibrous Tumor for Development of Metastasis

Risk Factor               Score

Age (y)

<< span=""> 55                0

≥ 55                1

Tumor size (cm)

<< span=""> 5                0

5- << span=""> 10                1

10- << span=""> 15                2

≥ 15                3

Mitotic count (per 10 high power fields)

0                0


perivascular tumors

Vascular tumors

Skeletal muscle tumors


Myopericytoma/ myofibroma

Epithelioid hemangioma

Pseudomyogenic hemangioendothelio- ma

Spindle cell/sclerosing rhabdomyosarcoma


PDGFRB mutations, SRF- RELA (cellular myofibroma)

FOS and FOSB fusions

SERPINE1-FOSB, ACTB-FOSB

MYOD1 mutations (adolescents/adults)

SRF-NCOA2, TEAD1-NCOA2, VGLL2-NCOA2,

VGLL2-CITED2

(congenital/infantile)

EWSR1-TFCP2, FUS-

TFCP2 (intraosseous; spindle cell and epithelioid)

1-3                1


Peripheral nerve Epithelioid


SMARCB1 mutations

≥ 4                2

Tumor necrosis (%)

<< span=""> 10                0


sheath tumors


schwannoma

Granular cell tumor                ATP6AP1 or ATP6AP2

mutations

≥ 10                1

Risk class                Total score


Benign triton tumor (neuromuscular choristoma)


CTNNB1 mutations

Low risk                0-3

Intermediate risk                4-5


MPNST                SUZ12 or EED mutations

(loss of H3K27me3)

High risk                6-7

Reprinted by permission from Springer Nature.96


Tumors of uncertain differentiation


Phosphaturic mesenchymal tumor


FN1-FGFR1, FN1-FGF1

(rare)

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.              www.ajsp.com | 15

forward in our understanding of SFT was the identification of NAB2-STAT6 fusions, and diagnostic utility of STAT6 IHC, as discussed above.

Additional novel genetic abnormalities included in the 2020 WHO are of less immediate practical diagnostic utility, but nevertheless improve our understanding of pathogenesis, and in some cases hold promise for potential tailored therapies. Examples include PDGFD fusions in a small subset of der- matofibrosarcoma protuberans (in addition to the dominant PDGFB fusions),99,100 BRAF fusions in a subset of myxoin- flammatory fibroblastic sarcomas,101 and FN1 fusions in phosphaturic mesenchymal tumors.102,103 Rhabdomyosarco- mas are also undergoing subclassification due to emerging molecular data, as the spindle cell/sclerosing rhabdomyo- sarcoma category has been revealed to harbor recurrent fusions and mutations and show distinct clinical features, analogous to reorganization of the formerly unclassifiable small round cell sarcoma category into new fusion-defined entities.

Epithelioid Hemangioma

Epithelioid hemangioma is a benign vascular neo- plasm that occurs most commonly in young to middle-aged


adults, with a predilection for the head and neck region. Other sites include the distal extremities, as well as rarely the oral mucosa, bone, or penis. Must tumors arise in dermis or subcutaneous tissues. The designation angio- lymphoid hyperplasia with eosinophilia has been used to describe a subset of head and neck cases with obscuring inflammation, although neither prominent lymphocytes nor eosinophils are required for the diagnosis of epithelioid hemangioma.

The histology of epithelioid hemangioma is charac- terized by well-circumscribed lesions with often lobular growth, and vessels that sometimes show a pattern of zo- nation including well-defined peripheral areas and com- pressed central areas; some examples are highly cellular with poorly canalized vascular channels (Fig. 13). The diagnostic cytologic finding is that of plump epithelioid endothelial cells with round nuclei, small nucleoli, and abundant eosinophilic cytoplasm containing occasional vacuoles (Fig. 13). Intimal or mural involvement of large blood vessels may be seen, and rare cases show purely intravascular growth, which is of no prognostic significance.104 By IHC, the lesional cells are positive for vascular markers (CD31 and ERG), as well as

FIGURE 13. Epithelioid hemangioma. A, A cellular example with nests of epithelioid tumor cells with abundant eosinophilic cytoplasm and occasional vacuoles. B, Nuclear staining for FOSB is a helpful diagnostic finding, identified in around 50% of cases.

16 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

FIGURE 14. EHE. A, Conventional EHE composed of uniform epithelioid cells with glassy eosinophilic cytoplasm arranged in cords within a myxohyaline stroma. B, EHE with YAP1-TFE3 fusion often shows a nested appearance; the tumor cells contain abundant pale  eosinophilic  cytoplasm.  C,  A  focally  vasoformative  architecture   is   usually   seen,   in   contrast   to   conventional   EHE. The degree of nuclear atypia in this case is notable. D, IHC for TFE3 shows strong nuclear staining.

FOSB in roughly half of cases (Fig. 13B), including the angiolymphoid hyperplasia with eosinophilia variant105 (although these cases appear to lack FOS family gene fusions; see below).

The 2020 WHO Classification includes the novel genetic discovery of characteristic recurrent fusions in FOS or FOSB, involving as many as half of epithelioid hemangioma cases. FOS has a variety of fusion partners, including LMNA, MBNL1, VIM, and lincRNA, whereas FOSB is often fused to ZFP36, or rarely to WWTR1 or ACTB. The FOS family genes encode leucine zipper pro- teins regulating angiogenesis, differentiation, and cell proliferation. Dysregulation of FOS signaling occurs by fusion partners affecting the C-terminal end of the protein, preventing normal degradation by the 20S proteasome and therefore prolonged FOS activation. The fusion pos- itive tumors are more often found in the extremities, trunk, and bone, and are far less often cutaneous; addi- tionally, these fusions are more often identified in the cellular subtype of epithelioid hemangioma.106–108 Recent


data indicate that the angiolymphoid hyperplasia with eosinophilia variant lacks these gene fusions.106

EHE With YAP1-TFE3

EHE is a malignant vascular neoplasm composed of epithelioid endothelial cells, distinct from epithelioid angio- sarcoma. Although > 90% of cases harbor the translocation t(1;3)(p36;q23-25), resulting in a WWTR1-CAMTA1 gene fusion, a small subset of tumors instead have a novel YAP1-TFE3 fusion; this subset of tumors is reported to occur in younger patients and shows distinctive histology.109 YAP1, similar to WWTR1, is a downstream transcriptional regulator of the Hippo pathway; there is substantial sequence homology between the 2 proteins. The histology of the classic variant includes cords and nests of epithelioid cells with eosinophilic cytoplasm and occasional intracytoplasmic vacuoles in a characteristic myxohyaline stroma (Fig. 14); conventional EHE does not form vascular channels. In contrast, the YAP1-TFE3 variant is composed of large epithelioid cells with voluminous pale cytoplasm and often

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.              www.ajsp.com | 17

FIGURE 15. Pseudomyogenic hemangioendothelioma. A, The tumor shows a loose fascicular architecture with relatively uniform spindle cells with abundant eosinophilic cytoplasm, mimicking a myoid neoplasm. Note the scattered stromal neutrophils. B, This example infiltrates muscle and contains polygonal to spindle cells with amphophilic cytoplasm. C, By IHC, the tumor cells are  diffusely positive for keratins using AE1/AE3 (but not MNF116). D, FOSB is strongly positive, reflecting an underlying FOSB gene rearrangement.

shows a nested growth pattern with a focally vasoformative architecture (Figs. 14B, C). By IHC, YAP1-TFE3 tumors show diffuse nuclear TFE3 expression (Fig. 14D). Recent data suggest that EHE with YAP1-TFE3 is associated with a more favorable outcome than conventional EHE.110

Pseudomyogenic Hemangioendothelioma

Pseudomyogenic hemangioendothelioma is an en- dothelial neoplasm of intermediate biologic potential that often presents as multiple discontiguous nodules in dif- ferent tissue planes, typically involving the limbs, espe- cially the lower extremities.111,112 Most patients present with skin lesions, roughly half have lesions within skeletal muscles, and a minority have bone lesions. This tumor type has a marked male predominance, a peak incidence in young adults, and is rare in adults older than 50 years. Histology demonstrates sheets and loose fascicles of plump spindled to polygonal cells with abundant brightly eosinophilic cytoplasm (Fig. 15), resembling a myoid tumor


or sometimes epithelioid sarcoma. A subset of cases demonstrates a prominent neutrophilic infiltrate. Additional features include infiltrative margins, and, for cutaneous lesi- ons, overlying epidermal hyperplasia. By IHC, pseudom- yogenic hemangioendothelioma is consistently positive for keratins (with AE1/AE3 but not MNF116) (Fig. 15C) and ERG; CD31 expression is less consistent, and CD34, EMA, desmin, and S100 protein are negative. FOSB shows diffuse nuclear expression in nearly all cases (Fig. 15D); this marker is useful to exclude histologic mimics including EHE and epithelioid sarcoma.113,114

The first hallmark genetic finding described in pseu- domyogenic hemangioendothelioma was the balanced translocation t(7;19)(q22;q13), resulting in a SERPINE1- FOSB fusion.115,116 Recently, a recurrent ACTB-FOSB fusion has also been found, in roughly half of cases117,118; these cases appear to demonstrate similar histologic fea- tures and clinical behavior as tumors with SERPINE1- FOSB fusion, except for more common presentation as

18 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

FIGURE 16. MPNST. A, The tumor shows alternating highly cellular, fascicular areas and less cellular areas. B, The tumor cells show loss of nuclear staining for H3K27me3 (histone H3 with lysine 27 trimethylation).

solitary lesions. Both fusions lead to upregulation of FOSB; as mentioned, IHC for FOSB is a useful diagnostic marker. Approximately 60% of patients suffer local recurrences or develop new nodules in the same anatomic region, usually within several years of initial presentation. Lymph node and distant metastases are uncommon.

Malignant Peripheral Nerve Sheath Tumor

MPNST is an aggressive neoplasm showing variably schwannian differentiation; MPNST often arises from a peripheral nerve or from a pre-existing neurofibroma, the latter most often in patients with neurofibromatosis type 1 (NF1).119 Overall, around 50% of patients with MPNST have NF1, 10% arise following therapeutic radiation therapy, and the remainder are sporadic. These tumors usually arise in adults, occur in deep-seated locations, and most commonly affect the proximal lower extremities, the paraspinal region, and proximal upper extremities. NF1 patients carry a 5% to 10% lifetime risk of developing MPNST, with malignant transformation most common in deep plexiform neurofibromas.120 Diagnosis in the spora- dic setting relies on distinctive histology and exclusion of mimics; the conventional Schwann cell markers S100


protein and SOX10 are only positive in 40% to 50% of cases, usually with limited, focal or patchy staining. The diagnosis has recently become more straightforward through the demonstration of loss of H3K27me3 by IHC, as detailed below.

The histology of MPNST is usually characterized by highly cellular fascicles of spindle cells, and often features al- ternating hypocellular/myxoid and hypercellular areas with perivascular accentuation of cellularity (Fig. 16). The spindle cells have hyperchromatic, tapering nuclei and are often relatively uniform, with some degree of nuclear pleomor- phism. Approximately 10% to 15% of tumors show heterologous mesenchymal differentiation, which can include rhabdomyoblastic, chondro-osseous, or rarely angiosarc- omatous elements; MPNST with rhabdomyoblastic differe- ntiation is widely known as malignant triton tumor. Rarely, epithelial (glandular) elements can be seen; glandular MPNST is highly associated with NF1.

In addition to mutations in NF1 and CDKN2A, recent genomic studies have identified inactivating mutations in genes that encode core components of the polycomb re- pressive complex 2 (PRC2), EED or SUZ12.121,122 Sporadic and radiotherapy-associated MPNST most often have

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.              www.ajsp.com | 19

loss-of-function somatic alterations of PRC2 components, identified in the majority of such cases. PRC2 inactivation results in loss of trimethylation of lysine 27 of histone H3 (H3K27me3), and aberrant downstream transcriptional activation.123 Epithelioid MPNST, by contrast, is molecu- larly and immunophenotypically distinct from conventional MPNST, commonly showing SMARCB1 gene inactivation and loss of INI1 by IHC, as well as consistent diffuse ex- pression of S100 protein and SOX10.

These improvements in our understanding of the biol- ogy of MPNST have led to identification of H3K27me3 as a diagnostically useful marker; it is otherwise  often  difficult (or impossible) to support the diagnosis by IHC given the limited (or completely absent) expression of Schwann cell markers. Complete loss of H3K27me3 has been demonstrated

in ∼60% of intermediate-grade and 85% of high-grade MPNST  (Fig.  16B); H3K27me3  loss is uncommon  in low-

grade tumors.124–127 Loss of H3K27me3 is also relatively specific for MPNST in the differential diagnosis with morphologic mimics, including benign nerve sheath tumors (eg, cellular schwannoma and atypical neurofibroma), monophasic synovial sarcoma, and high-risk SFT, among others, although a small subset of spindle cell melanomas and DDLPS also show loss of H3K27me3.128,129

CONCLUSIONS

The 2020 WHO Classification of Tumors of Soft Tis- sue and Bone features 「new」 soft tissue tumor entities, as well as updates in prognostic information for both rare and common sarcomas, and a host of recently described re- current genetic alterations. Increased awareness of rare tu- mor subtypes will facilitate their distinction from more commonly encountered lesions. Despite the rapidly emerging novel molecular alterations that have been identified across a broad array of tumor types, histologic features remain cru- cial to accurate subclassification. The WHO revision reflects the contemporary knowledge of the clinicopathologic fea- tures and molecular underpinnings of soft tissue tumors, updating classification with the goal of an increased under- standing of their biology, accurate diagnosis, and ultimately refined treatment regimens and improved patient outcomes.

REFERENCES

1.WHO Classification of Tumours Editorial Board. WHO Classi- fication of Tumours of Soft Tissue and Bone, 5th ed. Lyon, France: IARC Press; 2020.

2.Fletcher CDM, Bridge JA, Hogendoorn PCW, et al. WHO Classification of Tumours of Soft Tissue and Bone, 4th ed. Lyon, France: IARC Press; 2013.

3.Dei Tos AP, Mentzel T, Newman PL, et  al.  Spindle  cell  liposarcoma, a hitherto unrecognized variant  of  liposarcoma. Analysis of six cases. Am J Surg Pathol. 1994;18:913–921.

4.Mentzel T, Palmedo G, Kuhnen C. Well-differentiated spindle cell liposarcoma (『atypical spindle cell lipomatous tumor』) does not belong to the spectrum of atypical lipomatous tumor but has a close relationship to spindle cell lipoma: clinicopathologic, immunohis- tochemical, and molecular analysis of six cases. Mod Pathol. 2010;23:729–736.

5.Deyrup AT, Chibon F, Guillou L, et al. Fibrosarcoma-like lipomatous neoplasm: a reappraisal of so-called spindle cell lip- osarcoma defining a unique lipomatous tumor unrelated to other liposarcomas. Am J Surg Pathol. 2013;37:1373–1378.

6.
Mariño-Enriquez A, Nascimento AF, Ligon AH, et al. Atypical spindle cell lipomatous tumor: clinicopathologic characterization of 232 cases demonstrating a morphologic spectrum. Am J Surg Pathol. 2017;41:234–244.

7.Creytens D, Mentzel T, Ferdinande L, et al. 「Atypical」 pleomor- phic lipomatous tumor: a clinicopathologic, immunohistochemical and molecular study of 21 cases, emphasizing its relationship to atypical spindle cell lipomatous tumor and suggesting a morpho- logic spectrum (atypical spindle cell/pleomorphic lipomatous tumor). Am J Surg Pathol. 2017;41:1443–1455.

8.Bahadır B, Behzatoğlu K, Hacıhasanoğlu E, et al. Atypical spindle cell/pleomorphic lipomatous tumor: a clinicopathologic, immuno- histochemical, and molecular study of 20 cases. Pathol Int. 2018;68: 550–556.

9.Creytens D. A contemporary review of myxoid adipocytic tumors.

Semin Diagn Pathol. 2019;36:129–141.

10.Van Treeck BJ, Fritchie KJ. Updates in spindle cell/pleomorphic lipomas. Semin Diagn Pathol. 2019;36:105–111.

11.McCarthy AJ, Chetty R. Tumours composed of fat are no longer a simple diagnosis: an overview of fatty tumours with a spindle cell component. J Clin Pathol. 2018;71:483–492.

12.Chen BJ, Mariño-Enríquez A, Fletcher CD, et al. Loss of retinoblastoma protein expression in spindle cell/pleomorphic lipomas and cytogenetically related tumors: an immunohistochem- ical study with diagnostic implications. Am J Surg Pathol. 2012;36: 1119–1128.

13.Alaggio R, Coffin CM, Weiss SW, et al. Liposarcomas in young patients: a study  of  82  cases  occurring  in  patients  younger  than  22 years of age. Am J Surg Pathol. 2009;33:645–658.

14.Zare SY, Leivo M, Fadare O. Recurrent pleomorphic myxoid liposarcoma in a patient with Li-Fraumeni syndrome. Int J Surg Pathol. 2020;28:225–228.

15.Sinclair TJ, Thorson CM, Alvarez E, et al. Pleomorphic myxoid liposarcoma in an adolescent with Li-Fraumeni syndrome. Pediatr Surg Int. 2017;33:631–635.

16.Francom CR, Leoniak SM, Lovell MA, et al. Head and neck pleomorphic myxoid liposarcoma in a child with Li-Fraumeni syndrome. Int J Pediatr Otorhinolaryngol. 2019;123:191–194.

17.Boland JM, Colby TV, Folpe AL. Liposarcomas of the media- stinum and thorax: a clinicopathologic and molecular cytogenetic study of 24 cases, emphasizing unusual and diverse histologic features. Am J Surg Pathol. 2012;36:1395–1403.

18.Hofvander J, Jo VY, Ghanei I, et al. Comprehensive genetic analysis of a paediatric pleomorphic myxoid liposarcoma reveals near-haploidization and loss of the RB1 gene. Histopathology. 2016;69:141–147.

19.Creytens D, van Gorp J, Ferdinande L, et al. Array-based comparative genomic hybridization analysis of a pleomorphic myxoid liposarcoma. J Clin Pathol. 2014;67:834–835.

20.Mariño-Enríquez A, Fletcher CD. Angiofibroma of soft tissue: clinicopathologic characterization of a distinctive benign fibrovas- cular neoplasm in a series of 37 cases. Am J Surg Pathol. 2012;36: 500–508.

21.Jin Y, Möller E, Nord KH, et al. Fusion of the AHRR and NCOA2 genes through a recurrent translocation t(5;8)(p15;q13) in soft tissue angiofibroma results in upregulation of aryl hydrocarbon receptor target genes. Genes Chromosomes Cancer. 2012;51:510–520.

22.Bekers EM, Groenen P, Verdijk MAJ, et al. Soft tissue angiofibroma: clinicopathologic, immunohistochemical and molec- ular analysis of 14 cases. Genes Chromosomes Cancer. 2017;56: 750–757.

23.Arbajian E, Magnusson L, Mertens F, et al. A novel GTF2I/ NCOA2 fusion gene emphasizes the role of NCOA2 in soft tissue angiofibroma development. Genes Chromosomes Cancer. 2013;52: 330–331.

24.Nucci MR, Granter SR, Fletcher CD. Cellular  angiofibroma:  a benign neoplasm distinct from angiomyofibroblastoma and  spindle cell lipoma. Am J Surg Pathol. 1997;21:636–644.

25.Iwasa Y, Fletcher CD. Cellular angiofibroma: clinicopathologic and immunohistochemical analysis of 51 cases. Am J Surg Pathol. 2004;28:1426–1435.

20 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

26.Flucke U, van Krieken JH, Mentzel T. Cellular angiofibroma: analysis of 25 cases emphasizing its relationship to spindle cell lipoma and mammary-type myofibroblastoma. Mod Pathol. 2011; 24:82–89.

27.Kao YC, Flucke U, Eijkelenboom A, et al. Novel EWSR1-SMAD3 gene fusions in a group of acral fibroblastic spindle cell neoplasms. Am J Surg Pathol. 2018;42:522–528.

28.Michal M, Berry RS, Rubin BP, et al. EWSR1-SMAD3-rearranged fibroblastic tumor: an emerging entity in an increasingly more complex group of fibroblastic/myofibroblastic neoplasms. Am J Surg Pathol. 2018;42:1325–1333.

29.Puls F, Pillay N, Fagman H, et al. PRDM10-rearranged soft tissue tumor: a clinicopathologic study of 9 cases. Am J Surg Pathol. 2019; 43:504–513.

30.Carter JM, Weiss SW, Linos K, et al. Superficial CD34-positive fibroblastic tumor: report of 18 cases of a distinctive low-grade mesenchymal neoplasm of intermediate (borderline) malignancy. Mod Pathol. 2014;27:294–302.

31.Lao IW, Yu L, Wang J. Superficial CD34-positive fibroblastic tumour: a clinicopathological and immunohistochemical study of an additional series. Histopathology. 2017;70:394–401.

32.Merchant W, Calonje E, Fletcher CD. Inflammatory leiomyosar- coma: a morphological subgroup within the heterogeneous family of so-called inflammatory malignant fibrous histiocytoma. Histo- pathology. 1995;27:525–532.

33.Dal Cin P, Sciot R, Fletcher CD, et al. Inflammatory leiomyo- sarcoma may be characterized by specific near-haploid chromosome changes. J Pathol. 1998;185:112–115.

34.Chang A, Schuetze SM, Conrad EU III, et al. So-called 「inflammatory leiomyosarcoma」: a series of 3 cases providing additional insights into a rare entity. Int J Surg Pathol. 2005;13: 185–195.

35.Arbajian E, Köster J, Vult von Steyern F, et al. Inflammatory leiomyosarcoma is a distinct tumor characterized by near-haploid- ization, few somatic mutations, and a primitive myogenic gene expression signature. Mod Pathol. 2018;31:93–100.

36.Michal M, Rubin BP, Kazakov DV, et  al.  Inflammatory  leiomyosar- coma shows frequent co-expression of smooth and  skeletal  muscle markers supporting a primitive myogenic phenotype: a report of 9 cases with a proposal for reclassification as low-grade inflammatory myogenic tumor. Virchows Arch. 2020;477:219–230.

37.Antonescu CR, Dickson BC, Swanson D, et al. Spindle cell tumors with RET gene fusions exhibit a morphologic spectrum akin to tumors with NTRK gene fusions. Am J Surg Pathol. 2019;43: 1384–1391.

38.Suurmeijer AJH, Dickson BC, Swanson D, et al. A novel group of spindle cell tumors defined by S100 and CD34 co-expression shows recurrent fusions involving RAF1, BRAF, and NTRK1/2 genes.  Genes Chromosomes Cancer. 2018;57:611–621.

39.Agaram NP, Zhang L, Sung YS, et al. Recurrent NTRK1  gene  fusions define a novel subset of locally aggressive lipofibromatosis- like neural tumors. Am J Surg Pathol. 2016;40:1407–1416.

40.Davis JL, Lockwood CM, Albert CM, et al. Infantile NTRK- associated mesenchymal tumors. Pediatr Dev Pathol. 2018;21: 68–78.

41.Albert CM, Davis JL, Federman N, et al. TRK fusion cancers in children: a clinical review and recommendations for screening. J Clin Oncol. 2019;37:513–524.

42.Suurmeijer AJ, Dickson BC, Swanson D, et al. The histologic spectrum of soft tissue spindle cell tumors with NTRK3 gene rearrangements. Genes Chromosomes Cancer. 2019;58:739–746.

43.Rudzinski ER, Lockwood CM, Stohr BA, et al. Pan-Trk immunohistochemistry identifies NTRK rearrangements in pedia- tric mesenchymal tumors. Am J Surg Pathol. 2018;42:927–935.

44.Hechtman JF, Benayed R, Hyman DM, et al. Pan-Trk immuno- histochemistry is an efficient and reliable screen for the detection of NTRK fusions. Am J Surg Pathol. 2017;41:1547–1551.

45.Hung YP, Fletcher CDM, Hornick JL. Evaluation of pan-TRK immunohistochemistry in infantile fibrosarcoma, lipofibromatosis- like neural tumour and histological mimics. Histopathology. 2018; 73:634–644.

46.
Sbaraglia M, Righi A, Gambarotti M, et al. Ewing sarcoma and Ewing-like tumors. Virchows Arch. 2020;476:109–119.

47.Kilpatrick SE, Reith JD, Rubin B.  Ewing  sarcoma  and  the  history of similar and possibly related small round  cell  tumors:  from whence have we come and where are we going? Adv Anat Pathol. 2018;25:314–326.

48.Smith R, Owen LA, Trem DJ, et al. Expression  profiling of EWS/  FLI identifies NKX2.2 as a critical target gene in Ewing’s sarcoma. Cancer Cell. 2006;9:405–416.

49.Yoshida A, Sekine S, Tsuta K, et al. NKX2.2 is a useful immunohistochemical marker for Ewing sarcoma. Am J Surg Pathol. 2012;36:993–999.

50.Hung YP, Fletcher CD, Hornick JL. Evaluation of NKX2-2 expression in round cell sarcomas and other tumors with EWSR1 rearrangement: imperfect specificity for Ewing sarcoma. Mod Pathol. 2016;29:370–380.

51.De Alava E. Ewing sarcoma, an update on molecular pathology with therapeutic implications. Surg Pathol Clin. 2017;10:575–585.

52.Richkind KE, Romansky SG, Finklestein JZ. t(4;19)(q35;q13.1): a recurrent change in primitive mesenchymal tumors? Cancer Genet Cytogenet. 1996;87:71–74.

53.Kawamura-Saito M, Yamazaki Y,  Kaneko  K,  et  al.  Fusion  between CIC and DUX4 up-regulates PEA3 family genes in Ewing-like sarcomas with t(4;19)(q35;q13) translocation. Hum Mol Genet. 2006;15:2125–2137.

54.Italiano A, Sung YS, Zhang L, et al. High prevalence of CIC fusion with double-homeobox (DUX4) transcription factors in EWSR1- negative undifferentiated small blue round cell sarcomas. Genes Chromosomes Cancer. 2012;51:207–218.

55.Le Guellec S, Velasco V, Pérot G, et al. ETV4 is a useful marker for the diagnosis of CIC-rearranged undifferentiated round-cell sarco- mas: a study of 127 cases including mimicking lesions. Mod Pathol. 2016;29:1523–1531.

56.Antonescu CR, Owosho AA, Zhang L, et al. Sarcomas with CIC- rearrangements are a distinct pathologic entity with aggressive outcome: a clinicopathologic and molecular study of 115 cases. Am J Surg Pathol. 2017;41:941–949.

57.Hung YP, Fletcher CD, Hornick JL. Evaluation of ETV4 and WT1 expression in CIC-rearranged sarcomas and histologic mimics. Mod Pathol. 2016;29:1324–1334.

58.Yoshida A, Arai Y, Kobayashi E, et al. CIC break-apart fluorescence in-situ   hybridization   misses   a   subset   of   CIC-DUX4   sarcomas:   a clinicopathological and molecular study. Histopathology. 2017;71: 461–469.

59.Kao YC, Sung YS, Chen CL, et  al.  ETV  transcriptional  upregulation  is  more  reliable  than  RNA  sequencing  algorithms and FISH in diagnosing round cell sarcomas with CIC gene rearrangements. Genes Chromosomes Cancer. 2017;56:501–510.

60.Yoshida A, Goto K, Kodaira M, et al. CIC-rearranged sarcomas: a study of 20 cases and comparisons with Ewing sarcomas. Am J Surg Pathol. 2016;40:313–323.

61.Pierron G, Tirode F, Lucchesi C, et al. A new subtype of bone sarcoma defined by BCOR-CCNB3 gene fusion. Nat Genet. 2012; 44:461–466.

62.Specht K, Zhang L, Sung YS, et al. Novel BCOR-MAML3 and ZC3H7B-BCOR gene fusions in undifferentiated small blue round cell sarcomas. Am J Surg Pathol. 2016;40:433–442.

63.Puls F, Niblett A, Marland G, et al. BCOR-CCNB3 (Ewing-like) sarcoma: a clinicopathologic analysis of 10 cases, in comparison with conventional Ewing sarcoma. Am J Surg Pathol. 2014;38: 1307–1318.

64.Matsuyama A, Shiba E, Umekita Y, et al. Clinicopathologic diversity of undifferentiated sarcoma with BCOR-CCNB3 fusion: analysis of 11 cases with a reappraisal of the utility of immunohistochemistry for BCOR and CCNB3. Am J Surg Pathol. 2017;41:1713–1721.

65.Kao YC, Sung YS, Zhang L, et al.  BCOR  overexpression  is  a  highly sensitive marker in round cell sarcomas with BCOR genetic abnormalities. Am J Surg Pathol. 2016;40:1670–1678.

66.Kao YC, Sung YS, Zhang L, et al. BCOR upregulation in a poorly differentiated synovial sarcoma with SS18L1-SSX1 fusion—a

Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.              www.ajsp.com | 21

pathologic and molecular pitfall. Genes Chromosomes Cancer. 2017; 56:296–302.

67.Argani P, Kao YC, Zhang L, et al. BCOR overexpression in renal malignant solitary fibrous tumors: a close mimic of  clear  cell  sarcoma of kidney. Am J Surg Pathol. 2019;43:773–782.

68.Kao YC, Owosho AA, Sung YS, et al. BCOR-CCNB3 fusion positive sarcomas: a clinicopathologic and molecular analysis of 36 cases with comparison to morphologic spectrum and clinical behavior of other round cell sarcomas. Am J Surg Pathol. 2018;42: 604–615.

69.Watson S, Perrin V, Guillemot D, et al. Transcriptomic definition of molecular subgroups of small round cell sarcomas. J Pathol. 2018;245:29–40.

70.Chougule A, Taylor MS, Nardi V, et al. Spindle and round cell sarcoma with EWSR1-PATZ1 gene fusion: a sarcoma with polyphenotypic differentiation. Am J Surg Pathol. 2019;43:220–228.

71.Bridge JA, Sumegi J, Druta M, et al. Clinical, pathological, and genomic features of EWSR1-PATZ1 fusion sarcoma. Mod Pathol. 2019;32:1593–1604.

72.Diaz-Perez JA, Nielsen GP, Antonescu C, et al. EWSR1/FUS- NFATc2 rearranged round cell sarcoma: clinicopathological series of 4 cases and literature review. Hum Pathol. 2019;90:45–53.

73.Szuhai K, Ijszenga M, de Jong D, et al. The NFATc2 gene  is  involved in a novel cloned translocation in a Ewing sarcoma variant that couples its function in immunology to oncology. Clin Cancer Res. 2009;15:2259–2268.

74.Koelsche C, Kriegsmann M, Kommoss FKF, et al. DNA methylation profiling distinguishes Ewing-like sarcoma with EWSR1-NFATc2 fusion from Ewing sarcoma. J Cancer Res Clin Oncol. 2019;145:1273–1281.

75.Wang GY, Thomas DG, Davis JL, et al. EWSR1-NFATC2 translocation-associated sarcoma clinicopathologic findings in a rare aggressive primary bone or soft tissue tumor. Am J Surg Pathol. 2019;43:1112–1122.

76.Bode-Lesniewska B, Fritz C, Exner GU, et al. EWSR1-NFATC2 and FUS-NFATC2 gene fusion-associated mesenchymal tumors: clinicopathologic correlation and literature review. Sarcoma. 2019; 2019:9386390.

77.Wang L, Zehir A, Sadowska J, et al. Consistent copy number changes and recurrent PRKAR1A mutations distinguish Melanotic Schwannomas from Melanomas: SNP-array and next generation sequencing analysis. Genes Chromosomes Cancer. 2015;54:463–471.

78.Alexiev BA, Chou PM, Jennings LJ. Pathology of melanotic schwannoma. Arch Pathol Lab Med. 2018;142:1517–1523.

79.Torres-Mora J, Dry S, Li X, et al. Malignant melanotic schwannian tumor: a clinicopathologic, immunohistochemical, and gene expres- sion profiling study of 40 cases, with a proposal for the reclassification of 「melanotic schwannoma」. Am J  Surg  Pathol. 2014;38: 94–105.

80.Thway K. Well-differentiated liposarcoma and dedifferentiated lip- osarcoma: an updated review. Semin Diagn Pathol. 2019;36:112–121.

81.Mariño-Enríquez A, Fletcher CD, Dal Cin P, et al. Dedifferentiated liposarcoma with 「homologous」 lipoblastic (pleomorphic liposar- coma-like) differentiation: clinicopathologic and molecular analysis of a series suggesting revised diagnostic criteria. Am J Surg Pathol. 2010;34:1122–1131.

82.Mussi C, Collini P, Miceli R, et al. The prognostic impact of dedifferentiation in retroperitoneal liposarcoma: a series of surgi- cally treated patients at a single institution. Cancer. 2008;113: 1657–1665.

83.Keung EZ, Hornick JL, Bertagnolli MM, et al. Predictors of outcomes in patients with primary retroperitoneal dedifferentiated liposarcoma undergoing surgery. J Am Coll Surg. 2014;218: 206–217.

84.Jour G, Gullet A, Liu M, et al. Prognostic relevance of Fédération Nationale des Centres de Lutte Contre le Cancer grade and MDM2 amplification levels in dedifferentiated liposarcoma: a study of 50 cases. Mod Pathol. 2015;28:37–47.

85.Gronchi A, Collini P, Miceli R, et al. Myogenic differentiation and histologic grading are major prognostic determinants in retroper- itoneal liposarcoma. Am J Surg Pathol. 2015;39:383–393.

86.
Kurzawa P, Mullen JT, Chen YL, et al. Prognostic value of myogenic differentiation in dedifferentiated liposarcoma. Am J Surg Pathol. 2020;44:799–804.

87.Robinson DR, Wu YM, Kalyana-Sundaram S, et al. Identification of recurrent NAB2-STAT6 gene fusions in solitary fibrous tumor by integrative sequencing. Nat Genet. 2013;45:180–185.

88.Mohajeri A, Tayebwa J, Collin A, et al. Comprehensive genetic analysis identifies a pathognomonic NAB2/STAT6 fusion gene, nonrandom secondary genomic  imbalances,  and  a  characteristic gene expression profile in solitary fibrous tumor. Genes Chromo- somes Cancer. 2013;52:873–886.

89.Chmielecki J, Crago AM, Rosenberg M, et al. Whole-exome sequencing identifies a recurrent NAB2-STAT6 fusion in solitary fibrous tumors. Nat Genet. 2013;45:131–132.

90.Schweizer L, Koelsche C, Sahm F, et al. Meningeal hemangioper- icytoma and solitary fibrous tumors carry the NAB2-STAT6 fusion and can be diagnosed by nuclear expression of STAT6 protein. Acta Neuropathol. 2013;125:651–658.

91.Doyle LA, Vivero M, Fletcher CD, et al. Nuclear expression of STAT6 distinguishes solitary fibrous tumor from histologic mimics. Mod Pathol. 2014;27:390–395.

92.Akaike K, Kurisaki-Arakawa A, Hara K, et al. Distinct clinicopathological features of NAB2-STAT6 fusion gene variants in solitary fibrous tumor with emphasis on the acquisition of highly malignant potential. Hum Pathol. 2015;46:347–356.

93.Machado I, Morales GN, Cruz J, et al. Solitary fibrous tumor: a case series identifying pathological adverse factors-implications for risk stratification and classification. Virchows Arch. 2020;476:597–607.

94.Thway K, Ng W, Noujaim J, et al. The current status of solitary fibrous tumor: diagnostic features, variants, and genetics. Int J Surg Pathol. 2016;24:281–292.

95.Demicco EG, Park MS, Araujo DM, et al. Solitary fibrous tumor: a clinicopathological study of 110 cases and proposed risk assessment model. Mod Pathol. 2012;25:1298–1306.

96.Demicco EG, Wagner MJ, Maki RG, et al. Risk assessment  in solitary fibrous tumors: validation and refinement of a risk stratification model. Mod Pathol. 2017;30:1433–1442.

97.Salas S, Resseguier N, Blay JY, et al. Prediction of local and metastatic recurrence in solitary fibrous tumor: construction of a risk calculator in a multicenter cohort from the French Sarcoma Group (FSG) database. Ann Oncol. 2017;28:1979–1987.

98.Hornick JL. Limited biopsies of soft tissue tumors: the contempo- rary role of immunohistochemistry and molecular diagnostics. Mod Pathol. 2019;32:27–37.

99.Dadone-Montaudié B, Alberti L, Duc A, et al. Alternative PDGFD rearrangements in dermatofibrosarcomas protuberans without PDGFB fusions. Mod Pathol. 2018;31:1683–1693.

100.Dickson BC, Hornick JL, Fletcher CDM, et al. Dermatofibrosar- coma protuberans with a novel COL6A3-PDGFD fusion gene and apparent predilection for breast. Genes Chromosomes Cancer. 2018; 57:437–445.

101.Kao YC, Ranucci V, Zhang L, et al. Recurrent BRAF gene rearrangements in myxoinflammatory fibroblastic sarcomas, but not hemosiderotic fibrolipomatous tumors. Am J Surg Pathol. 2017;41:1456–1465.

102.Lee JC, Jeng YM, Su SY, et al. Identification  of  a  novel  FN1-FGFR1 genetic fusion as a frequent event in phosphaturic mesenchymal tumour. J Pathol. 2015;235:539–545.

103.Lee JC, Su SY, Changou CA, et al.  Characterization  of  FN1-FGFR1 and novel FN1-FGF1 fusion genes in a large series of phosphaturic mesenchymal tumors. Mod Pathol. 2016;29:1335–1346.

104.Luzar B, Ieremia E, Antonescu CR, et al. Cutaneous intravascular epithelioid hemangioma. A clinicopathological and molecular study of 21 cases. Mod Pathol. 2020;33:1527–1536.

105.Ortins-Pina A, Llamas-Velasco M, Turpin S, et al. FOSB immunor- eactivity in endothelia of epithelioid hemangioma (angiolymphoid hyperplasia with eosinophilia). J Cutan Pathol. 2018;45:395–402.

106.Huang SC, Zhang L, Sung YS, et al. Frequent FOS gene rearrangements in epithelioid hemangioma: a molecular study of  58 cases with morphologic reappraisal. Am J Surg Pathol. 2015; 39:1313–1321.

22 | www.ajsp.com              Copyright © 2020 Wolters Kluwer Health, Inc. All rights reserved.

107.Antonescu CR, Chen HW, Zhang L, et al. ZFP36-FOSB fusion defines a subset of epithelioid hemangioma with atypical features. Genes Chromosomes Cancer. 2014;53:951–959.

108.Van IDG, de Jong D, Romagosa C, et al. Fusion events lead to truncation of FOS in epithelioid hemangioma of bone. Genes Chromosomes Cancer. 2015;54:565–574.

109.Antonescu CR, Le Loarer F, Mosquera JM, et al. Novel YAP1-TFE3 fusion defines a distinct subset of epithelioid heman- gioendothelioma. Genes Chromosomes Cancer. 2013;52:775–784.

110.Rosenbaum E, Jadeja B, Xu B, et al. Prognostic stratification of clinical and molecular epithelioid hemangioendothelioma subsets. Mod Pathol. 2020;33:591–602.

111.Hornick JL, Fletcher CD. Pseudomyogenic hemangioendothelio- ma: a distinctive, often multicentric tumor with indolent behavior. Am J Surg Pathol. 2011;35:190–201.

112.Papke DJ Jr., Hornick JL. What is new in endothelial neoplasia?

Virchows Arch. 2020;476:17–28.

113.Sugita S, Hirano H, Kikuchi N, et al. Diagnostic utility of FOSB immunohistochemistry in pseudomyogenic hemangioendothelioma and its histological mimics. Diagn Pathol. 2016;11:75.

114.Hung YP, Fletcher CD, Hornick JL. FOSB is a useful diagnostic marker for pseudomyogenic hemangioendothelioma. Am J Surg Pathol. 2017;41:596–606.

115.Trombetta D, Magnusson L, von Steyern FV, et al. Translocation t (7;19)(q22;q13)—a recurrent chromosome aberration in pseudomyo- genic hemangioendothelioma? Cancer Genet. 2011;204:211–215.

116.Walther C, Tayebwa J, Lilljebjörn H, et al. A novel SERPINE1-FOSB fusion gene results in transcriptional up-regulation of FOSB in pseudomyogenic haemangioendothelioma. J Pathol. 2014;232:534–540.

117.Agaram NP, Zhang L, Cotzia P, et al. Expanding the spectrum of genetic alterations in pseudomyogenic hemangioendothelioma with recurrent novel ACTB-FOSB gene fusions. Am J Surg Pathol. 2018;42:1653–1661.

118.Zhu G, Benayed R, Ho C, et al. Diagnosis of known sarcoma fusions and novel fusion partners by targeted RNA sequencing with identification of a recurrent ACTB-FOSB fusion in pseudomyo- genic hemangioendothelioma. Mod Pathol. 2019;32:609–620.

119.
Martinez AP, Fritchie KJ. Update on peripheral nerve sheath tumors. Surg Pathol Clin. 2019;12:1–19.

120.Miettinen MM, Antonescu CR, Fletcher CDM, et al. Histopatho- logic evaluation of atypical neurofibromatous tumors and their transformation into malignant peripheral nerve sheath tumor in patients with neurofibromatosis 1—a consensus overview. Hum Pathol. 2017;67:1–10.

121.De Raedt T, Beert E, Pasmant E, et al. PRC2 loss amplifies Ras- driven transcription and confers sensitivity to BRD4-based therapies. Nature. 2014;514:247–251.

122.Zhang M, Wang Y, Jones S, et al. Somatic mutations of SUZ12 in malignant peripheral nerve sheath tumors. Nat Genet. 2014;46: 1170–1172.

123.Lee W, Teckie S, Wiesner T, et al. PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors. Nat Genet. 2014;46:1227–1232.

124.Schaefer IM, Fletcher CD, Hornick JL. Loss of H3K27 trimethy- lation distinguishes malignant peripheral nerve sheath tumors from histologic mimics. Mod Pathol. 2016;29:4–13.

125.Prieto-Granada CN, Wiesner T, Messina JL, et al. Loss of H3K27me3 expression is a highly sensitive marker for sporadic and radiation-induced MPNST. Am J Surg Pathol. 2016;40:479–489.

126.Otsuka H, Kohashi K, Yoshimoto M, et al. Immunohistochemical evaluation of H3K27 trimethylation in malignant peripheral nerve sheath tumors. Pathol Res Pract. 2018;214:417–425.

127.Cleven AH, Sannaa GA, Briaire-de Bruijn I, et al. Loss of H3K27 tri-methylation is a diagnostic marker for malignant peripheral nerve sheath tumors and an indicator for an inferior survival. Mod Pathol. 2016;29:582–590.

128.Le Guellec S, Macagno N, Velasco V, et al. Loss of H3K27 trimethylation is not suitable for distinguishing malignant  periph-  eral nerve sheath tumor from melanoma: a study of 387 cases including mimicking lesions. Mod Pathol. 2017;30:1677–1687.

129.Makise N, Sekimizu M, Kubo T, et al. Clarifying the distinction between malignant peripheral nerve sheath tumor and dedifferenti- ated liposarcoma: a critical reappraisal of the diagnostic utility of MDM2 and H3K27me3 status. Am J Surg Pathol. 2018;42:656–664.

相關焦點

  • MacSween's Pathology of the Liver
    Medicine  Books Gray's Anatomy.Gray's Anatomy for StudentsBraunwald’s Heart Disease: A Textbook ofCardiovascular MedicineHurst's the HeartHarrison's Principles of Internal MedicineOxford Handbook of
  • Paper · News | Tissue-sharing in XISU
    Tissue-sharing in XISUChen XiaoqiAfter the sharing economy going famous and popular, such as bike-sharing, XISU has recently put the tissue-sharing into practice on March
  • Crohn's Disease
    This includes your mouth, your food pipe (esophagus), your stomach, the first part of your small intestine or duodenum, your appendix, and your anus.What causes Crohn's disease?
  • FREE LINK: Goljan Rapid Review Pathology 6th edition!
    Edward Goljan is your go-to guide for up-to-date, essential pathology information throughout medical school.
  • The New E-Bike Rules: What You Can Do
    If you are looking for clarification on the new e-bike situation as of April> all gave different answers when asked about what they should do about the new rules and who will be affected by it.
  • New York's Hudson Yards Hope To Revitalise Retail
    ImagesFun, fabulous and future-proof,」 says retail queen Marigay McKee, referring to what others might describe as 『retail’s last hope』.
  • What's Next for Protein Beverages?
    The goal of the product and application will dictate what protein source you use.」「This means it’s on the formulators to explore what is actually happening in the bottle, from batching to processing to one year into the shelf life,」 she said.
  • Post-Workout Nutrition: The Importance of What You Eat
    After exercise, our bodies are tired and are in need of nutrients to replenish what we have lost during the workout in order to replenish the stores and aid muscle recovery and rehydrate.
  • China new long March 8's two boosters "asymmetric"?
    China's new medium-size carrier rocket, the Long March-8, blasted off from the Wenchang Space Launch Center on Dec. 22, carrying five experimental satellites into orbit.
  • New wearable device turns the body into a battery
    It also taps into a person's natural heat -- employing thermoelectric generators to convert the body's internal temperature into electricity.
  • what's what可不是「什麼是什麼」
    今天分享一個很重要的習語「what's what」 它的意思可不是「什麼是什麼」或「」什麼的什麼」。它的含義是:what's what|事情的真相。Donald Trump has been telling people thatthe COVID -19 is nothing and no need to fear of so that lots of people don't know what's what.