You are on page 1of 13

Small Round Cell Tumors of Soft Tissue and Bone

Shi Wei, MD, PhD; Gene P. Siegal, MD, PhD

 Context.—Small round cell tumors of soft tissue and peer-reviewed English-language literature and firsthand
bone constitute a divergent group of neoplasms. These experience from the authors as practicing bone and soft
lesions often demonstrate overlapping clinical and radio- tissue pathologists.
logic characteristics and share histomorphologic and Conclusions.—Immunohistochemistry plays a vital role
sometimes immunophenotypic similarities, but they typi- in rendering a specific diagnosis or narrowing the
cally have diverse prognostic outcomes, thus warranting differential diagnosis in small round cell tumors of soft
different clinical management. Recent advances in molec- tissue and bone. Molecular genetic studies are often
ular and cytogenetic techniques have identified a number
needed, especially for those lesions with unusual histologic
of novel molecular alterations contributing to the diversity
features, an uncommon immunoprofile, and/or unusual
of these lesions. This state-of-the-art knowledge has
enhanced our understanding of these diseases. clinical presentation. Accurate diagnosis of these tumors
Objective.—To provide an overview of the current necessitates recognition of salient histologic features,
concepts in the classification and diagnosis of small round judicious and astute use of ancillary studies, and correla-
cell tumors of soft tissue and bone, focusing on salient tion with the clinical and radiologic characteristics to
histologic features, key immunophenotypic characteristics, guide clinical decision-making.
and recent molecular genetic advancements. (Arch Pathol Lab Med. 2022;146:47–59; doi: 10.5858/
Data Sources.—Data were obtained from pertinent arpa.2020-0773-RA)

S mall round cell tumors (also known as small, blue, round


cell tumors) constitute a divergent group of neoplasms.
These tumors are morphologically alike and characterized as
Recent molecular genetic advances have identified a
growing list of round cell sarcomas, thus having revolu-
tionized the diagnosis of sarcomas and provided insight into
highly aggressive malignant tumors composed of relatively potential therapeutic targets as well as prognostic biomark-
small, round, hyperchromatic and monotonous undifferen- ers. The research to that end has also resulted in the
tiated cells with an increased nuclear to cytoplasmic ratio. reclassification of a number of previously established tumor
The differential diagnosis of these tumors is particularly types, as illustrated in the most recent edition of the World
difficult because of their undifferentiated or primitive Health Organization (WHO) classification of soft tissue and
character. They can occur in any age group but demonstrate bone tumors. Herein, we provide an overview of the current
different age peaks for given tumor types.1 Although there concepts of small round cell tumors of soft tissue and bone,
are always exceptions, some tumors primarily affect the focusing on salient histologic features, key immunopheno-
skeleton, such as Ewing sarcoma and small cell osteosar- typic characteristics, and recent molecular genetic advance-
coma, whereas others typically occur in somatic soft tissue, ments.
including rhabdomyosarcoma and desmoplastic small round
cell tumor. Moreover, the overlapping histologic, immuno- EWING SARCOMA AND SO-CALLED EWING-LIKE
histochemical, and molecular features create diagnostic SARCOMAS
challenges despite significant clinical and prognostic differ-
ences as well as therapeutic indications. Ewing Sarcoma
The Ewing sarcoma/primitive neuroectodermal tumor
Accepted for publication December 11, 2020.
(PNET) family of tumors was historically thought to be 2
Published online February 26, 2021. different entities, with the latter demonstrating neuroecto-
From the Departments of Pathology (Wei, Siegal) and Genetics dermal differentiation. It is now generally recognized that
(Siegal), O’Neal Comprehensive Cancer Center, University of Ewing sarcoma of bone, extraosseous Ewing sarcoma,
Alabama, Birmingham. PNET, and Askin tumor (PNET arising in the chest wall)
This work was funded in part by the Haley’s Hope Memorial
Support Fund and the Thomaus Logan RAID Fund. The authors have represent one and the same neoplastic process. These
no relevant financial interest in the products or companies described tumors share the common genetic abnormalities character-
in this article. ized by fusions involving a member of the TET (TLS/EWSR1/
Presented in part at the Seventh Princeton Integrated Pathology TAF15) or FET (FUS/EWSR1/TAF15) family of genes and a
Symposium; May 16, 2020; Plainsboro, New Jersey. member of the ETS family of transcription factors. The most
Corresponding author: Shi Wei, MD, PhD, Department of
Pathology, University of Alabama at Birmingham, NP 3545, 619 common cytogenetic abnormality is the t(11;22)(q24; q12)
19th Street South, Birmingham, AL 35249-7331 (email: swei@ translocation, which results in EWSR1-FLI1 fusion tran-
uabmc.edu). script (~85%), followed by t(21;22)(q22; q12), which results
Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal 47
Figure 1. Selective Ewing and ‘‘Ewing-like’’ sarcomas. A, Ewing sarcoma of soft tissue demonstrates solid sheets of uniform, small, blue, round cells
with scant cytoplasm and minimal extracellular matrix. B, Atypical Ewing sarcoma arising in the kidney shows larger, atypical epithelioid cells. C,
Admantinoma-like Ewing sarcoma of the thyroid manifests lobules of primitive, small round cells. D through F, CIC-rearranged sarcoma displays
variably sized round cells with focal prominent myxoid stroma (D), moderate nuclear pleomorphism (E), and diffuse WT1 expression (F)
(hematoxylin-eosin, original magnifications 3200 [A through D] and 3400 [E]; original magnification 3400 [F]).

in a EWSR1-ERG transcript (~10%). The remaining cases Ewing sarcoma typically appears as solid sheets of
demonstrate alternative translocations involving either uniform, small blue round cells with scant clear or
EWSR1 or FUS and other ETS family members, including eosinophilic cytoplasm and minimal extracellular matrix
ETV1, ETV4, and FEV, each accounting for ,1% of cases.1 (Figure 1, A). In some cases, the tumor cells are larger and
Ewing sarcoma is the second most common malignant more pleomorphic, known as atypical Ewing sarcoma
tumor of bone in children and young adults, with a peak (Figure 1, B). So-called admantinoma-like Ewing sarcoma
incidence during the second decade of life. Those occurring typically occurs in the head and neck region (Figure 1, C).2,4,5
in patients older than 30 years more often affect soft tissue.2 Diffuse membranous expression of CD99 is characteristic for
When in long bones, it typically arises in the diaphysis or Ewing sarcoma (~95%) but not specific. Focal expression of
metadiaphysis, although any bone in the skeleton can be cytokeratin can also be seen.1 FLI1 and ERG are often
affected. An ill-defined, osteolytic (and rarely osteosclerotic) expressed in those with the corresponding gene fusions,
lesion with an ‘‘onion-skin’’–like multilayered periosteal albeit not specifically, in addition to their utility as
reaction is the most characteristic radiologic presentation. endothelial markers. FLI1 is also expressed in a number of
Extraskeletal Ewing sarcoma (about 12% of all cases) has a lymphomas, including lymphoblastic lymphoma (also
wide distribution of anatomic sites, including visceral CD99þ), anaplastic large cell lymphoma, and angioimmu-
organs.2,3 noblastic T-cell lymphoma, as well as a subset of
48 Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal
melanomas, carcinomas of various organs, and other sar- identified on 10q26, and the fusions of CIS to the DUX4
comas, such as synovial sarcoma.6 ERG is also positive in a gene on 10q26 have also been found subsequently.16 To
number of other malignancies, including acute myeloid date, CIC-DUX4 fusion is the most frequent genetic
leukemia, epithelioid sarcoma, and a subset of prostate car- alteration in EWSR1/FUS-negative undifferentiated small
cinoma.7–9 NKX2.2, a homeodomain-containing transcrip- round cell tumors, whereas a number of other fusion
tion factor that plays a critical role in neuroendocrine/glial partners for CIC have been recently identified, including
differentiation, is a target of EWSR1-FLI1 and has been FOXO4, LEUTX, NUTM1, and NUTM2A.17
shown to be a relatively sensitive marker with a suboptimal CIC-rearranged sarcomas primarily occur in the deep soft
specificity. It is also positive in Ewing sarcoma with an tissue of the limbs and trunk; less commonly affect the head
EWSR1-ERG gene rearrangement, and thus may be helpful and neck, retroperitoneum, pelvis and visceral organs; and
in distinguishing Ewing sarcoma from its histologic mimics.10 rarely arise in bone. There is a wide age distribution at
Genetic analysis is often required for the diagnosis of presentation, ranging from 6 to 81 years, but they more
Ewing sarcoma. Fluorescence in situ hybridization analysis commonly occur in young adults.
using a break-apart probe is highly sensitive in detecting Histologically, CIC-rearranged sarcomas are typically
EWSR1 rearrangements but does not identify its transloca- composed of solid sheets of small round cells, with (at least
tion partner, whereas reverse transcription–polymerase in part) a lobulated growth pattern. Focal myxoid stroma is
chain reaction analysis is a more specific test in identifying reportedly seen in one-third of cases (Figure 1, D). A minor
the EWSR1-FLI1 fusion gene but has suboptimal sensitivity component of spindled or epithelioid cells is not uncom-
(54%) in formalin-fixed, paraffin-embedded tissue.11 Virtu- mon. Most cases show minor nuclear pleomorphism,
ally all current commercial sarcoma fusion panels include although a moderate degree of cytologic atypia may be
EWSR1. rarely present, including coarse chromatin and prominent
Ewing sarcoma displays a high propensity for distant nucleoli (Figure 1, E). These tumors may have variable CD99
metastases, with the most common sites being lung and immunoreactivity, ranging from negative through focal and/
bone. The prognosis of Ewing sarcoma has been improved or weak to diffuse and/or strong. Expression of WT1 and
with current multimodal therapeutic strategies. ETV4 is seen in 90% to 100% of tumors, and thus is useful in
distinguishing CIC-rearranged sarcomas from their histo-
Round Cell Sarcoma With EWSR1–Non-ETS Fusions logic mimickers (Figure 1, F). Other rarely expressed
There is an emerging subgroup of small round cell markers include NUT (CIC-NUTM1 fusion), cytokeratin,
S100 protein, calretinin, ERG, and myogenic makers.17
sarcomas that possess EWSR1 or FUS rearrangements and
Most CIC-rearranged tumors have a dismal response to
share various degrees of similarity with Ewing sarcoma
Ewing sarcoma regiments and have aggressive outcomes
clinically, histomorphologically, and immunophenotypically
when compared to Ewing sarcoma matched for age and
(ie, CD99 and NKX2.2). However, these tumors lack the
stage. Distant metastasis is frequent, most commonly to the
pathognomonic molecular hallmark of Ewing sarcoma
lung.
because their fusion partners are invariably non-ETS
transcription factor family members. The reported cytoge- Sarcoma With BCOR Genetic Alterations
netic alterations include EWSR1-NFATc2, FUS-NFATc2,
Another distinct entity of round cell sarcoma is charac-
EWSR1-PATZ1, EWSR1-SMARCA5, and EWSR1-SP3.12,13 terized by reproducible BCOR genetic alterations. In 2012, a
These tumors have been previously regarded as Ewing-like fusion of the BCOR (BCL6 corepressor) and CCNB3 genes,
sarcomas. which are nonadjacent genes on the X chromosome, was
There is a wide age distribution for this group of tumors, identified in 4 cases of round cell sarcoma of bone lacking
and each of them has a small number of cases. EWSR1- the canonical TET-ETS translocation.18 Additional gene
NFATc2 and FUS-NFATc2 sarcomas predominantly affect fusions involving BCOR have also been subsequently found,
bones, with a strong male predominance (5:1). EWSR1- including BCOR-ITD (internal tandem duplication), BCOR-
PATZ1 sarcomas arise in deep soft tissue, and the same MAML3, ZC3H7B-BCOR, BCOR-KMT2D, and CIITA-
fusion has also been identified in gliomas.14 They are mostly BCOR.19–22
treated similarly to Ewing sarcoma, with generally unfavor- BCOR-CCNB3 sarcomas more frequently arise in bone
able outcomes. Given the limited data, it remains to be than soft tissue, have a striking predilection for children and
determined whether these tumors represent one or more young adults (.90% younger than 20 years), and are male
stand-alone pathologic entities or are better classified as predominant. BCOR-ITD tumors occur preferentially in the
variants of Ewing sarcoma. somatic soft tissue of the trunk, abdomen, and head and
neck, sparing the extremities.23 The BCOR-ITD fusion and,
CIC-Rearranged Sarcoma
less frequently, YWHAE-NUTM2B (both resulting in up-
There have been new emerging undifferentiated round regulation of BCOR), frequently occur in infantile round cell
cell sarcomas that clinically and histologically mimic Ewing sarcoma, clear cell sarcoma of the kidney, and so-called
sarcoma but fail to demonstrate any of the aforementioned primitive myxoid mesenchymal tumor of infancy.19
molecular genetic abnormalities characteristic of Ewing Histologically, sarcomas with BCOR alterations consist of
sarcoma. In 2006, two cases of ‘‘Ewing-like sarcoma’’ were uniform, primitive small round or spindle cells in solid
found to harbor a recurrent t(4;19)(q35;q13) translocation, sheets or a nesting pattern, with varying myxoid or
which resulted in fusion between CIC (Capicua Transcrip- collagenous stroma. Strong and diffuse nuclear BCOR
tional Repressor), a human homolog of Drosophila capicua expression is seen in virtually all tumors but is not specific,
that encodes a high-mobility group box transcription factor, because it can be seen in other tumors, such as synovial
and DUX4, a double homeodomain gene.15 The latter is sarcoma.19 CD99 expression is variable (~50%). SATB2,
located within a D4Z4 repeat array in the subtelomeric TLE1, and cyclin D1 can also be expressed. Cyclin B3 is also
region of 4q35. A similar D4Z4 repeat array has been expressed in BCOR-CCNB3 sarcomas but not other BCOR-
Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal 49
SMALL ROUND CELL SARCOMAS PRODUCING
CARTILAGE OR BONE MATRIX
Small Cell Osteosarcoma
Small cell osteosarcoma has long been considered as a
stand-alone subtype of osteosarcoma.24 It has been now
merged into conventional osteosarcoma as a histologic
variant by the most recent edition of the WHO classification
of soft tissue and bone tumors.25 This rare variant,
reportedly comprising approximately 1.5% of all osteosar-
comas, has a wide age distribution but is more commonly
seen in the second decade of life. Most instances occur in
the metaphysis or diaphysis of long bones. The clinical and
radiologic features of small cell osteosarcoma are similar to
those of conventional osteosarcomas, whereas it reportedly
has a slightly worse prognosis than the latter.24,25
The histomorphologic features of small cell osteosarcoma
are those of combined osteosarcoma and Ewing sarcoma.
The lesional cells are typically round to oval and, less
frequently, short-spindled, and generally have hyperchro-
matic nuclei and scanty cytoplasm (Figure 2, A). The nuclear
size may vary from as small as those of Ewing sarcoma to as
large as those comparable to large cell lymphoma. Nucleoli
are rarely seen and are typically inconspicuous, if present.
The presence of a focal hemangiopericytoma-like growth
pattern is a frequent finding. The diagnosis of small cell
osteosarcoma requires identification of osteoid/bone matrix
production, which is frequently subtle and more commonly
arranged in a lacelike fashion (Figure 2, B).4 Mitotic figures
are variably present. Tumor cells may variably express CD99
and, rarely, keratin and epithelia membranous antigen
(EMA), thus representing a diagnostic pitfall. Special AT-
rich sequence-binding protein 2 (SATB2), a transcription
factor essential for osteoblastogenesis, is a very sensitive
marker but lacks specificity. It can be of help in distinguish-
ing small cell osteosarcoma from Ewing sarcoma in the
absence of unequivocal tumor-producing osteoid/bone, as it
is only rarely (1.3%) expressed in the latter.26
To date, in keeping with other conventional osteosarco-
mas, no recurrent molecular genetic abnormalities have
been detected in small cell osteosarcoma.27 Interestingly, the
t(11; 22)(q24; q12) translocation was reported in 1 case of
small cell osteosarcoma in 1990.28 Subsequently, another
case of small cell osteosarcoma with an EWSR1 rearrange-
ment was also detected by interphase fluorescence in situ
hybridization.29 An EWSR1-CREB3L1 fusion transcript was
identified in a multifocal small cell osteosarcoma, and the
corresponding chimeric gene was confirmed by sequencing
of the genomic breakpoint between EWSR1 and CREB3L1.30
It is not clear if a bone-forming round cell sarcoma
harboring an EWSR1 rearrangement should be classified
as small cell osteosarcoma or Ewing sarcoma. Some
authorities believe that cases with this morphology and
Figure 2. Round cell sarcomas producing bone or cartilage matrix. A, EWSR1 translocation should be considered as Ewing
Small cell osteosarcoma, areas with no identifiable osteoid/bone matrix. sarcoma with divergent differentiation. Investigation of their
B, Small cell osteosarcoma producing lacelike tumor osteoid, with a possible relationship may provide further insight into the
hemangiopericytoma-like growth pattern. C, Mesenchymal chondro- molecular mechanisms of the 2 tumor types.
sarcoma characterized by a biphasic appearance composed of hyaline
cartilage and sheets of small, round, blue cells (hematoxylin-eosin, Mesenchymal Chondrosarcoma
original magnifications 3200 [A], 3100 [B], and 340 [C]).
Mesenchymal chondrosarcoma is a biphasic tumor
comprising primitive round cells and well-differentiated
rearranged tumors.23 The prognosis in BCOR-CCNB3 hyaline cartilage. This high-grade tumor, accounting for 2%
sarcomas is similar to Ewing sarcoma, whereas the to 4% all chondrosarcomas, may occur at any age, with a
outcomes of other tumors in the BCOR family are not well peak incidence in the second and third decades of life. The
characterized. tumor more commonly arises in bone but may also
50 Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal
primarily affect extraskeletal sites. There is a widespread the abdomen, retroperitoneum, and biliary tract, whereas it
skeletal distribution, more frequently occurring in the axial rarely affects the extremities.38
skeleton, including craniofacial bones, ribs, ilium, and The tumor contains primitive mesenchymal cells housed
vertebrae. The meninges are one of the most common within a loose myxoid stroma, admixed with variably
extraskeletal sites. Visceral organs, such as kidney, can also differentiated rhabdomyoblasts, which demonstrate elon-
be rarely involved. Skeletal lesions are primarily lytic and gation, cytoplasmic eosinophilia, and sometimes cross-
destructive, with poor margins on imaging studies, often not striation in cells with terminal differentiation (Figure 3, A).
significantly different from conventional osteosarcoma.31 Poorly differentiated tumors may be composed exclusively
The histologic hallmark of mesenchymal chondrosarcoma of solid sheets of primitive round cells (also called a dense
is a biphasic appearance composed of sheets of primitive pattern), thus resulting in confusion with ARMS.39 Heter-
small round cells admixed with islands of well-differentiated ologous cartilaginous differentiation, albeit rare, is a well-
hyaline cartilage (Figure 2, C). The former simulates Ewing recognized phenomenon (Figure 3, B). Botryoid ERMS
sarcoma or lymphoma, whereas the cartilage nodules may refers to those with a linear distribution of tumor cells
mimic normal chondrogenesis in the growth plate, or those abutting an epithelium-lined surface (Figure 3, C). So-called
from a well-differentiated cartilaginous neoplasm. A hem- anaplastic ERMS is characterized by the presence of
angiopericytoma-like growth pattern can be seen in the markedly atypical cells, not to be confused with pleomor-
phic RMS that typically occurs in elderly individuals.
small cell zones, as can subtle osteoid production. The
Although immunoreactivity may be variable, ERMSs are
transition between the 2 components may be abrupt or
always positive for desmin, a key subunit of the intermediate
gradual.1,32
filament in cardiac, skeletal, and smooth muscles, as well as
The small cells are variably positive for CD99. SOX9, a myogenin and MyoD1, both of which are transcription
master regulator of chondrogenesis, is positive in both small factors involved in myogenesis. It is noteworthy that
cell and cartilaginous components in virtually all cases, but it nonspecific cytoplasmic MyoD1 staining is not infrequent,
is negative in other small cell malignancies, and thus it may and it may be misinterpreted as positive. Aberrant
serve as a useful tool in the differential diagnosis of small expression of other markers, such as cytokeratins and
round cell tumors.33 S100 protein, can also be seen.40 There are no reproducible
The defining molecular signature of mesenchymal chon- molecular genetic alterations identified for ERMS.
drosarcoma is a recurrent HEY1-NCOA2 fusion in almost all
well-characterized cases, representing an in-frame fusion of Alveolar Rhabdomyosarcoma
HEY1 exon 4 to NCOA2 exon 13 at the mRNA level, Alveolar RMS occurs more commonly in the second and
probably arising through intrachromosomal rearrangement third decades of life and more often affects the deep soft
of chromosome 8q.34 This fusion was absent in other tissue of extremities, followed by the head and neck,
subtypes of chondrosarcoma. Interestingly, t(1;5)(q42;q32) paraspinal, and perineal regions.41 The tumor is typically
as the sole karyotypic aberration was reported in a single composed of a monotonous population of primitive,
case of mesenchymal chondrosarcoma lacking a HEY1- hyperchromatic round cells arranged in nests divided by
NCOA2 fusion. This resulted in a novel fusion between the fibrous septa, thus giving rise to an alveolar growth pattern
IRF2BP2 gene and the transcription factor CDX1 gene, (Figure 3, D). The solid variant of ARMS is densely cellular
suggesting the genetic heterogeneity in this tumor.35 but lacks septa and thus may be confused with the dense
Notably, IDH1/2 mutations frequently harbored in central pattern of ERMS.39 Rhabdomyoblastic differentiation may
and periosteal chondromas and chondrosarcomas have not be seen, but not often, and when present it is often a minor
been identified in mesenchymal chondrosarcoma.36,37 component. Other occasional histologic features include
Mesenchymal chondrosarcoma is an aggressive neoplasm multinucleated giant cells and clear cell morphology.
with a poor prognosis. Surgical resection followed by The molecular signature of ARMS is a t(2;13)(q35;q14) or,
adjuvant chemotherapy has been considered the standard less frequently, t(1;13)(p36;q14) translocation, resulting in
of care treatment. Young age and craniofacial bone PAX3-FOXO1 or PAX7-FOXO1 fusion genes, respectively.42
involvement are reportedly associated with a slightly better Compared with ERMS, myogenin expression in ARMS is
prognosis.31 typically stronger and diffuse, whereas MyoD1 expression
may be focal. Expression of focal keratin or neuroendocrine
RHABDOMYOSARCOMA markers can be occasionally seen (Figure 3, E). This should
not be confused with olfactory neuroblastoma when in a
Rhabdomyosarcoma (RMS) constitutes the largest cate- sinonasal location, because the latter typically demonstrates
gory of soft tissue sarcomas in children and young adults. diffuse expression of neuroendocrine markers. ARMS is
The subtypes with prominent small round cell morphology clinically more aggressive than ERMS, thus, it is important
include embryonal RMS (ERMS), alveolar RMS (ARMS), to distinguish the former from the latter.38
and, rarely, spindle cell/sclerosing RMS, whereas pleomor-
phic RMS typically does not demonstrate a small round cell Spindle Cell/Sclerosing Rhabdomyosarcoma
morphology. As its name implies, spindle cell/sclerosing RMS demon-
Embryonal Rhabdomyosarcoma strates a spindle cell and/or sclerosing morphology. This
variant most commonly involves the head/neck region
Embryonal RMS is the most common subtype of RMS, followed by the extremities in adults, but it arises more
typically affecting children younger than 10 years and frequently in the paratesticular region in boys. It may also
occasionally occurring in adolescents, with a predilection rarely occur in visceral organs, the retroperitoneum, and
in white individuals. The head and neck region and bone.43
genitourinary system are the common sites of involvement. The molecular genetic abnormalities identified in this
The tumor may infrequently arise in other sites, including subtype are complex but can be largely categorized into 3
Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal 51
Figure 3. Rhabdomyosarcomas. A, Embryonal rhabdomyosarcoma exhibiting primitive rounded to spindled cells as well as numerous
differentiating rhabdomyoblasts. B, Embryonal rhabdomyosarcoma with cartilaginous differentiation. C, The botryoid pattern of embryonal
rhabdomyosarcoma showing a variably cellular tumor with condensation beneath an epithelium-lined surface. D, Alveolar rhabdomyosarcoma
consisting of monotonous round cells with an ‘‘alveolar’’ growth pattern. E, Focal neuroendocrine differentiation of alveolar rhabdomyosarcoma as
demonstrated by antisynaptophysin immunohistochemistry. F, Sclerosing rhabdomyosarcoma displaying clusters of single cells in a sclerotic
collagenized stroma (hematoxylin-eosin, original magnifications 3200 [A, C, and D], 3100 [B], and 3400 [F]; original magnification 3200 [E]).

groups. The congenital/infantile spindle cell RMS shows coma or fibrosarcoma, with variable degrees of nuclear
VGLL2/NCOA2/CITED2 rearrangements, resulting in gene pleomorphism and mitotic activities. Rhabdomyoblastic
fusions, including SRF-NCOA2, TEAD1-NCOA2, VGLL2- differentiation can be observed occasionally. In sclerosing
NCOA2, and VGLL2-CITED2. Further, the MYOD1 RMS, the tumor cells are arranged in various patterns, such
p.Leu122Arg gene mutation is typically seen in young as nests, cords, trabeculae, or microalveoli, and they are
adults. Important to note is that recurrent genetic alterations embedded in a distinctive sclerotic/hyalinized stroma that
have not been identified in a significant proportion of cases. may simulate tumor osteoid in osteosarcomas. Primitive
Interestingly, EWSR1/FUS-TFCP2 and MEIS1-NCOA2 fu- undifferentiated cells with hyperchromatic, round nuclei
sions have been recently identified in a number of and a high nuclear to cytoplasmic ratio may constitute a
intraosseous RMSs, showing distinct morphology and variable proportion of the tumor (Figure 3, F). This, along
immunophenotype.44 Thus, further classification of this with the lack of prominent sclerotic stroma, may be
subtype is emerging. mistaken for other undifferentiated small round cell
Histologically, spindle cell RMS is characterized by the sarcomas. Moreover, a distinctive epithelioid morphology
proliferation of spindle cells arranged in sheets, fascicles, or has also been observed in intraosseous RMS.44 Spindle cell/
a herringbone pattern, as is commonly seen in leiomyosar- sclerosing RMSs typically demonstrate variable expresion of
52 Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal
desmin, limited myogenin, and strong positivity for MyoD1.
Aberrant expresison of cytokeratin and ALK has been
reportedly seen in intraosseous RMS.
The prognosis of this group of tumors is highly variable.
Congenital/infantile spindle cell RMSs harboring gene
fusions have a favorable clinical course. MYOD1-mutant
tumors are associated with poor survival outcomes.43

ROUND CELL SARCOMAS WITH A PROMINENT


SCLEROTIC STROMA
Desmoplastic Small Round Cell Tumor
Desmoplastic small round cell tumor is a distinct
translocation-related sarcoma characterized by small round
cells with polyphenotypic differentiation, prominent stromal
desmoplasia, and an EWSR1-WT1 gene fusion. It primarily
affects children and young adults, with a striking male
predilection. The tumor more commonly arises in the
abdomen, retroperitoneum, or pelvis, with widespread
serosal implants.45
The neoplastic cells may vary in size and shape, and they
may rarely be spindled or epithelioid. They are typically
arranged in irregularly contoured nests embedded in a
prominent desmoplastic stroma, with occasional glandlike
or rosettelike structures (Figure 4, A). Tumor necrosis,
frequent mitoses, and cystic degeneration are common.
Multiphenotypic differentiation is a distinctive feature of
desmoplastic small round cell tumor. Therefore, markers for
epithelial, muscular, and neural differentiation may be
variably immunoreactive, including antibodies directed
against cytokeratins, EMA, desmin, and neuron-specific
enolase. Myogenin and MyoD1 biomarkers are negative.
Selective nuclear expression of WT1 using antibodies raised
against the C-terminus, but not the N-terminus, is a
characteristic immunophenotype.46,47 Dotlike perinuclear
staining with desmin and coexpression of cytokeratin can
be seen in both desmoplastic small round cell tumor and
Wilms tumor, whereas dual immunoreactivity for the WT1
N-terminus and C-terminus is seen in the latter.40,48
The detection of an EWSR1-WT1 rearrangement resulting
from a t(11;22)(p13;q12) translocation is diagnostic for
desmoplastic small round cell tumor but not always needed
for cases with characteristic histomorphology and immu-
nophenotype. The clinical outcomes are generally poor.
Sclerosing Epithelioid Fibrosarcoma
Sclerosing epithelioid fibrosarcoma (SEF) is a distinctive
fibroblastic neoplasm characterized by epithelioid tumor
cells arranged in nests, cords, or sheets embedded within a
prominent sclerotic collagenized stroma. It most commonly
arises in deep soft tissue of extremities followed by the
shoulder, trunk, and head and neck regions, but it may
rarely occur in the visceral organs or bone.49,50
The tumor is typically composed of small, bland, and
Figure 4. Round cell sarcomas with a prominent sclerotic stroma. A, uniform epithelioid cells with round to ovoid nuclei and
Desmoplastic small round cell tumor showing nests of round cells
separated by a densely collagenized stroma. B, Sclerosing epithelioid eosinophilic or clear cytoplasm. It lacks significant cytologic
fibrosarcoma displaying bland, uniform, small, round, epithelioid cells atypia, thus sometimes closely mimicking lobular carcinoma
lacking significant cytologic atypia resembling lobular carcinoma of the of the breast (Figure 4, B). The lesional cells are arranged in
breast. C, Another example of sclerosing epithelioid fibrosarcoma sheets, nests, cords, or, rarely, fascicles. Pseudoalveolar or
exhibiting a pseudoalveolar growth pattern (hematoxylin-eosin, original pseudoglandular growth may also be seen (Figure 4, C).
magnifications 3100 [A and C] and 3200 [B]). Less cellular areas may have myxoid or fibrous stroma.
Marked nuclear pleomorphism, increased mitotic activities,
and necrosis are uncommon features. There is a small subset
of SEF cases showing overlapping histologic, immunophe-
notypic, and molecular characteristics with low-grade
Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal 53
fibromyxoid sarcoma (LGFMS).51 This observation leads and pelvis.55 Rare cases of primary extraskeletal myxoid
some authorities to propose a potential relationship chondrosarcoma of bone have been described.56
between these 2 tumors. Extraskeletal myxoid chondrosarcoma characteristically
The most distinctive immunophenotype of SEF is the demonstrates a multilobulated growth pattern, in which
expression of mucin 4 (MUC4) (~90%), similar to that in the neoplastic cells are interconnected with each other to
LGFMS. Focal, weak expression of EMA can be seen, form cords, strands, or clusters (Figure 5, C). The tumor is
whereas staining for cytokeratins is typically negative. Most distinctively hypocellular and hypovascular and lacks well-
SEFs harbor EWSR1-CREB3L1 fusions. Less frequently, developed hyaline cartilage. The tumor cells exhibit bland,
EWSR1 may exchange with FUS or PAX5, and CREB3L1 round, oval, or bipolar nuclei, and moderate eosinophilic or
may be replaced by CREB3L2, CREB3L3, or CREM.52 So- vacuolated cytoplasm, and some have prominent rhabdoid
called hybrid SEF/LGFMS cases more frequently display a features. In the rare examples of the hypercellular variant,
FUS-CREB3L2 fusion identical to that observed in most the cells often show epithelioid cytomorphology and an
LGFMS.51 Pure SEFs have a more aggressive clinical course elevated nuclear grade.40
than LGFMSs. There are no specific immunohistochemical markers for
extraskeletal myxoid chondrosarcoma. The tumors may
ROUND CELL TUMORS WITH A PROMINENT MYXOID variably express S100 protein, CD117, and, rarely, cytoker-
STROMA atin and neuroendocrine markers. Those with rhabdoid
features may show loss of INI1. A t(9;22)(q22;q12)
The presence of a prominent myxoid stroma is not
translocation resulting in an EWSR1-NR4A3 fusion has
uncommon in many tumor types, some of which have
been found as the sole anomaly, whereas a number of other
round cell morphology, as described herein.
rare fusion partners for NR4A3 have been recently
Myxoid Liposarcoma identified, including TAF15, TCF12, TFG, FUS, or
HSPA8.57–59 Extraskeletal myxoid chondrosarcoma is usually
Myxoid liposarcoma (which consolidates with the entity
associated with a prolonged survival despite of high rates of
formerly known as round cell liposarcoma) is characteris-
local and distant recurrence.
tically composed of uniform, round/ovoid, nonlipogenic
cells and small lipoblasts. The lesional cells are set in a Mixed Tumors/Myoepithelial Neoplasms
prominent myxoid stroma with a curvilinear capillary Myoepithelial tumors of soft tissue constitute a group of
vascular network that gives rise to a ‘‘chicken-wire’’ neoplasms closely related to their salivary gland counter-
appearance. The tumor most frequently affects young adults, parts, namely myoepithelioma, myoepithelial carcinoma,
with the peak incidence in the fourth to fifth decades of life. and mixed tumor (pleomorphic adenoma). These tumors
It typically arises in the deep soft tissue of extremities, may show variable growth patterns and demonstrate a wide
have synchronous or metachronous multifocal disease, and spectrum of histologic features, including epithelioid,
spreads hematogenously to unusual locations, such as spindled, plasmacytoid, vacuolated (formerly classified as
retroperitoneum or bone.53,54 parachordoma), and undifferentiated round cell morphology
Histologically the tumors are typically lobulated, and the (Figure 5, D). Mixed tumors show a ductal component. The
cellularity is usually moderate but varies from region to stroma may be myxoid or hyalinized. Myoepithelial
region. There may be large mucin pools in paucicellular carcinoma is defined by increased cytologic atypia, often
areas, thus imparting a pulmonary edema-like growth along with high mitotic activity and necrosis. Cartilaginous
pattern. The small round cells lack cytologic atypia. The or osseous dedifferentiation can be seen. Expression of
lipoblasts range from rare to abundant, are usually small, myoepithelial markers is variable, and the most frequently
and are often univacuolated, mimicking signet ring cells expressed markers include cytokeratin, EMA, S100 protein,
(Figure 5, A). The pathognomonic molecular genetic SOX10, calponin, p63, and glial fibrillary acidic protein
alteration of myxoid liposarcoma is a t(12;16)(q13;p11) (GFAP). Loss of INI1 has been observed in a subset of
translocation leading to FUS-DDIT3 fusion transcripts myoepithelial carcinomas. Approximately half of myoepi-
(.95%). A EWSR1-DDIT3 gene fusion resulting from thelial tumors harbor an EWSR1 rearrangement, with a
t(12;22)(q13;q12) substitutes in a very small subset of number of fusion partners identified, including POU5F1,
tumors.53 PBX1, ZNF444, KLF17, ATF1, and PBX3. Rarely, an alternate
High-grade tumors have .5% hypercellularity, less FUS rearrangement with similar partner genes is seen. A
myxoid matrix, and increased nuclear pleomorphism and PLAG1 rearrangement has been identified in mixed tumors
mitotic activity, and are associated with a significantly similar to that seen in their salivary gland counterparts.60
higher rate of distant metastases or death. Pure high-grade
lesions lack myxoid matrix and thus may be indistinguish- Poorly Differentiated Sarcomas With Round Cell
able from other undifferentiated round cell sarcomas, Morphology and Myxoid Stroma
requiring molecular studies for diagnostic confirmation Round cell morphology, along with a spectrum of other
(Figure 5, B). histologic features, is not common in various poorly
differentiated sarcomas. Among them, poorly differentiated
Extraskeletal Myxoid Chondrosarcoma
synovial sarcoma is such an example. The latter may show
Extraskeletal myxoid chondrosarcoma is characterized by variable histologic features, including epithelioid, fascicular
small, uniform cells with round to oval nuclei, set in an growth of spindle cells (reminiscent of malignant peripheral
abundant chondromyxoid matrix. The tumor typically arises nerve sheath tumor), and hyperchromatic small round cells
in the deep soft tissue of proximal extremities and limb resembling Ewing sarcoma (Figure 5, E).61 On the other
girdles in middle-aged adults. Extraskeletal myxoid chon- hand, myxoid stromal changes are not uncommon in
drosarcoma may also involve the soft tissue of other synovial sarcoma. They are usually focal but can occasionally
anatomic sites including the head and neck, abdomen, become a predominant feature, resulting in alternating
54 Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal
Figure 5. Selective common and uncommon round cell tumors with a prominent myxoid stroma. A, Myxoid liposarcoma composed of uniform,
round nonlipogenic cells and small lipoblasts set in a prominent myxoid stroma with a ‘‘chicken-wire’’ vascular network. B, A pure high-grade myxoid
liposarcoma lacking myxoid matrix, and thus indistinguishable from other undifferentiated round cell sarcomas. C, Extraskeletal myxoid
chondrosarcoma characterized by hypocellular and hypovascular bland, round, ovoid, or bipolar cells arranged in cords, strands, or clusters in a
prominent chondromyxoid matrix. D, Myoepithelial carcinoma showing plasmacytoid and round cell morphology, with a prominent myxoid stroma.
E, Poorly differentiated synovial sarcoma consisting of undifferentiated small round cells with a hemangiopericytoma-like growth pattern. F, Poorly
differentiated and myxoid synovial sarcoma with an arrangement of small round cells in retiform cords or microcysts (hematoxylin-eosin, original
magnifications 3200 [A, C through F] and 3200 [B]).

hypocellular and relatively hypercellular areas and arrange- matrix on imaging in middle-aged and older adults.
ments of cells in retiform cords or microcysts.62 The myxoid Although the recognition of plasmacytic origin is mostly
stroma may rarely become prominent, making the diagnosis straightforward given the characteristic cytomorphologic
extremely challenging (Figure 5, F). In such cases, the more features of plasma cells, poorly differentiated plasmacyto-
typical histologic features of synovial sarcoma may be better mas and those with anaplastic morphology may be
recognized in more cellular areas at the periphery of the
problematic (Figure 6, A). Of important note, membranous
lesion.
CD138 reactivity for neoplastic cells in bone is not a
HEMATOLOGIC NEOPLASMS AND METASTASES definitive marker for plasmacytic origin unless a diagnosis of
It is doubtless that hematologic neoplasms frequently hematologic disease has been established.63 Additional
originate in bone. Solitary plasmacytoma of bone and ancillary studies, such as expression of MUM1 and/or j or
plasma cell myeloma, along with metastases, are always top k light chain restriction, may be needed for further
differential diagnoses for lytic lesions without identifiable diagnostic confirmation.
Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal 55
Figure 6. Selective primary hematologic neoplasms of bone and metastasis of nonepithelial round cell neoplasms to bone. A, Poorly differentiated
plasmacytoma. B, Therapy-related myeloid sarcoma in a patient with metastatic lobular breast cancer to the bone. C, Metastatic medulloblastoma. D,
Metastatic melanoma. E, Metastatic well-differentiated neuroendocrine tumor of the pancreas. F, Metastatic epithelioid gastrointestinal tumor
(hematoxylin-eosin, original magnification 3200).

Primary lymphoma of bone is not uncommon, accounting than sarcomas. Virtually all types of carcinomas may relapse
for about 7% of malignant bone tumors. Approximately 5% in bone, some of which are primarily of small round cell
of all extranodal lymphomas originate in bone. The most morphology, such as small cell carcinoma, lobular carcino-
common site is femur, followed by spine and pelvic bones. ma of the breast, and Merkel cell carcinoma. It is
Lymphoma of bone may involve a single anatomic site or noteworthy that adenocarcinoma of some organs (ie,
multiple bones. Most lymphomas of bone are diffuse large prostate) may recur locally or distantly as small cell
B-cell lymphomas. Extranodal Burkitt lymphomas common- carcinoma after treatment.64 When a metastatic lesion
ly involve bone. T-cell lymphomas of bone are rare but do exhibits a round cell morphology, however, the differential
occur, such as anaplastic large cell lymphoma. Rarely, diagnosis is wide-ranging, including (but not limited to)
therapy-related myeloid sarcoma can arise in bone (Figure neuroblastoma, Wilms tumor, retinoblastoma, medulloblas-
6, B). Further detailed descriptions of primary bone toma, hepatoblastoma, melanoma, well-differentiated neu-
lymphomas are beyond the scope of this review. roendocrine tumor, and gastrointestinal stromal tumor,
It is of utmost importance to keep in mind that bone along with the aforementioned entities (Figure 6, C through
represents one of the most common distant sites of tumor F). Thus, pertinent clinical history is crucial in reaching a
metastasis, of which carcinomas are much more common correct diagnosis.
56 Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal
Clinicopathologic Characteristics of Selective Small Round Cell Tumors
Tumor Type Clinical Characteristics Salient Histologic Features Key Immunophenotypes Molecular Genetics
Ewing sarcoma Children and young adults; Uniform small round cells CD99, NKX2.2, FLI1, ERG Gene fusion involving
long bones . other with scant cytoplasm and EWSR1/FUS and a
skeleton; extraskeletal minimal extracellular member of ETS family
involvement more matrix (FLI1 . ERG . others)
common in middle-aged
or elderly
Round cell sarcoma Variable Similar to Ewing sarcoma Variable; CD99 and EWSR1-NFATc2, FUS-
with EWSR1–non- NKX2.2 more common NFATc2, EWSR1-
ETS fusions PATZ1, EWSR1-
SMARCA5, EWSR1-SP3
CIC-rearranged Wide age range but more Round cells (rarely spindled WT1, ETV4, CD99 (þ/) CIC-DUX4 (rarely other
sarcoma common in young adults; or epithelioid) without fusion partners for CIC)
deep soft tissue of the significant nuclear
limbs and trunk but rarely pleomorphism; variably
involving visceral organs myxoid
and bone
Sarcoma with BCOR Children and young adults; Round or spindled cells; BCOR, SATB2, TLE1, BCOR-CCNB3, BCOR-
genetic alterations male predilection; bone solid or nested; variably cyclin D1, cyclin D3 ITD, BCOR-MAML3,
. soft tissue myxoid (BCOR-CCNB3), ZC3H7B-BCOR,
CD99 (þ/) BCOR-KMT2D, CIITA-
BCOR
Small cell Metaphysis or diaphysis of Combined osteosarcoma SATB2, CD99 (þ/) No recurrent molecular
osteosarcoma long bones and Ewing sarcoma; genetic alteration;
hemangiopericytoma-like EWSR1 rearrangement
growth; lacelike osteoid/ rarely reported
bone matrix
Mesenchymal Wide age distribution, peak Biphasic tumor comprising CD99, SOX9 HEY1-NCOA2
chondrosarcoma in the second and third small round cells and
decades; bone . soft hyaline cartilage
tissue
Embryonal Typically first decade, Primitive mesenchymal cells, Desmin, myogenin, MyoD1 No reproducible
rhabdomyosarcoma predilection in white variable rhabdomyoblasts, molecular genetic
individuals; more common loose myxoid stroma alterations
in head/neck region and
genitourinary system
Alveolar Second and third decades, Monotonous small round Desmin, myogenin (strong/ PAX3-FOXO1, PAX7-
rhabdomyosarcoma more commonly affecting cells, alveolar growth diffuse), MyoD1 FOXO1
deep soft tissue of pattern
extremities
Sclerosing More common in head/neck Primitive cells (þ/ Desmin (variable), MyoD1 MYOD1 mutation (small
rhabdomyosarcoma region or extremities rhabdomyoblasts) with (strong), myogenin subset)
(adults) and paratesticular various growth patterns (limited)
location (children) and sclerotic stroma;
epithelioid cells (in bone)
Desmoplastic small Children/young adults, male Small round cells with Multiphenotypic EWSR1-WT1
round cell tumor predilection; commonly prominent stromal differentiation (epithelial/
involving abdomen, pelvis, desmoplasia muscular/neural); desmin
or retroperitoneum (dotlike), WT-1 (C-
terminus)
Sclerosing epithelioid Deep soft tissue and rarely Small, bland, uniform MUC4 EWSR1 (FUS/PAX5)-
fibrosarcoma visceral organs or bone epithelioid cells and CREB3L1 (CREB3L2/
fibrotic stroma CREB3L3/CREM)
Myxoid liposarcoma Young adults, peak in the Uniform round nonlipogenic None FUS-DDIT3 (.95%),
fourth to fifth decades; cells and small lipoblasts; EWSR1-DDIT3
deep soft tissue of myxoid stroma, ‘‘chicken-
extremities wire’’ vascular network
Extraskeletal myxoid Middle-aged adults; deep Small, uniform cells in S100 (variable) EWSR1-NR4A3 (rarely
chondrosarcoma soft tissue cords, strands, or clusters; other fusion partners
multilobulated growth for NR4A3)
pattern; abundant
chondromyxoid matrix

Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal 57
SUMMARY mutational burden, and hypermutation signatures. Oncologist. 2017;22(12):
1478–1490.
Primary small round cell tumors of soft tissue and bone 15. Kawamura-Saito M, Yamazaki Y, Kaneko K, et al. Fusion between CIC and
constitute a diverse group of neoplasms sharing many DUX4 up-regulates PEA3 family genes in Ewing-like sarcomas with t(4;19)(q35;
q13) translocation. Hum Mol Genet. 2006;15(13):2125–2137.
histomorphologic similarities and immunophenotypes, and 16. Italiano A, Sung YS, Zhang L, et al. High prevalence of CIC fusion with
they may have overlapping clinical and radiologic charac- double-homeobox (DUX4) transcription factors in EWSR1-negative undifferen-
teristics but diverse clinical outcomes, as illustrated in the tiated small blue round cell sarcomas. Genes Chromosomes Cancer. 2012;51(3):
207–218.
Table. These lesions may also demonstrate largely overlap- 17. Antonescu CR, Yoshida A. CIC-rearranged sarcoma. In: Bridge J, ed. WHO
ping histomorphologic features with hematologic disorders Classification of Tumors: Soft Tissue and Bone. Lyon, France: International Agency
and metastatic diseases, thus resulting in diagnostic for Research on Cancer; 2020:330–332.
18. Pierron G, Tirode F, Lucchesi C, et al. A new subtype of bone sarcoma
challenges. Although the pathogenesis of a subset of these defined by BCOR-CCNB3 gene fusion. Nat Genet. 2012;44(4):461–466.
tumors remains to be ascertained, state-of-the-art knowl- 19. Kao YC, Sung YS, Zhang L, et al. Recurrent BCOR internal tandem
edge has enhanced our understanding of these tumors. duplication and YWHAE-NUTM2B fusions in soft tissue undifferentiated round
cell sarcoma of infancy: overlapping genetic features with clear cell sarcoma of
Although numerous molecular alterations have contributed kidney. Am J Surg Pathol. 2016;40(8):1009–1020.
to the diversity of these lesions, recent molecular and 20. Specht K, Zhang L, Sung YS, et al. Novel BCOR-MAML3 and ZC3H7B-
genetic advancements have led to the reclassification of BCOR gene fusions in undifferentiated small blue round cell sarcomas. Am J Surg
Pathol. 2016;40(4):433–442.
some of these entities with potential novel therapeutic 21. Kao YC, Owosho AA, Sung YS, et al. BCOR-CCNB3 fusion positive
targets emerging, which have significantly improved treat- sarcomas: a clinicopathologic and molecular analysis of 36 cases with
ment modalities and prognostic outcomes. Immunohisto- comparison to morphologic spectrum and clinical behavior of other round cell
sarcomas. Am J Surg Pathol. 2018;42(5):604–615.
chemical analyses provide useful tools in characterizing 22. Yoshida A, Arai Y, Hama N, et al. Expanding the clinicopathologic and
these lesions and narrowing the differential diagnosis. molecular spectrum of BCOR-associated sarcomas in adults. Histopathology.
However, interpretation of biomarkers requires a cautious 2020;76(4):509–520.
23. Antonescu CR, Puls F, Tirode F. Sarcoma with BCOR genetic alterations. In:
and judicious approach. Molecular genetic studies are often Bridge J, ed. WHO Classification of Tumors: Soft Tissue and Bone. 5th ed. Lyon,
needed, especially for those with an unusual histomorphol- France: International Agency for Research on Cancer; 2020:333–335.
ogy and typical histologic features but unusual clinical 24. Kalil RK, Squire J. Small cell osteosarcoma. In: Fletcher CDM, Bridge JA,
Hodendoorn PCW, Mertens F, eds. WHO Classification of Tumors of Soft Tissue
presentation (ie, age or anatomic site), or with an and Bone. Lyon, France: International Agency for Research on Cancer; 2013:291–
uncommon immunoprofile. Thus, correlation with the 292.
clinical and radiologic characteristics, recognition of salient 25. Baumhoer D, Bohling TO, Cates JMM, et al. Osteosarcoma. In: Bovee
JVMG, Flanagan AM, Lazar AJ, Nielsen GP, Yoshida A, eds. WHO Classification
histologic features, and astute use of ancillary studies are of Tumors: Soft Tissue and Bone. 5th ed. Lyon, France: International Agency for
critical to reach the correct diagnosis and guide clinical Resarch on Cancer; 2020:403–409.
decision-making. 26. Machado I, Navarro S, Picci P, Llombart-Bosch A. The utility of SATB2
immunohistochemical expression in distinguishing between osteosarcomas and
References their malignant bone tumor mimickers, such as Ewing sarcomas and chondro-
1. Wei S, Siegal GP. Round cell tumors of bone: an update on recent sarcomas. Pathol Res Pract. 2016;212(9):811–816.
molecular genetic advances. Adv Anat Pathol. 2014;21(5):359–372. 27. Righi A, Gambarotti M, Longo S, et al. Small cell osteosarcoma:
2. de Alava E, Lessnick SL, Stamenkovic I. Ewing sarcoma. In: Bridge J, ed. clinicopathologic, immunohistochemical, and molecular analysis of 36 cases.
WHO Classification of Tumors: Soft Tissue and Bone. 5th ed. Lyon, France: Am J Surg Pathol. 2015;39(5):691–699.
International Agency for Research on Cancer; 2020:323–325. 28. Noguera R, Navarro S, Triche TJ. Translocation (11;22) in small cell
3. Soni A, Wei S. Primary renal Ewing sarcoma in an adult. Urology. 2017;99: osteosarcoma. Cancer Genet Cytogenet. 1990;45(1):121–124.
e11–e13. 29. Dragoescu E, Jackson-Cook C, Domson G, Massey D, Foster WC. Small cell
4. Small, round cell lesions. In: Wei S, Siegal GP, eds. Atlas of Bone Pathology. osteosarcoma with Ewing sarcoma breakpoint region 1 gene rearrangement
New York, NY: Springer 2013:237–282. detected by interphase fluorescence in situ hybridization. Ann Diagn Pathol.
2013;17(4):377–382.
5. Morlote D, Harada S, Lindeman B, Stevens TM. Adamantinoma-like Ewing
30. Debelenko LV, McGregor LM, Shivakumar BR, Dorfman HD, Raimondi SC.
sarcoma of the thyroid: a case report and review of the literature. Head Neck
A novel EWSR1-CREB3L1 fusion transcript in a case of small cell osteosarcoma.
Pathol. 2019;13(4):618–623.
Genes Chromosomes Cancer. 2011;50(12):1054–1062.
6. Rossi S, Orvieto E, Furlanetto A, Laurino L, Ninfo V, Dei Tos AP. Utility of
31. Fanburg-Smith JC, de Pinieux G, Ladanyi M. Mesenchymal chondrosarco-
the immunohistochemical detection of FLI-1 expression in round cell and
ma. In: Bovee JVMG, Flanagan AM, Lazar AJ, Nielsen GP, Yoshida A, eds. WHO
vascular neoplasm using a monoclonal antibody. Mod Pathol. 2004;17(5):547–
Classification of Tumors: Soft Tissue and Bone. 5th ed. Lyon: International Agency
552.
for Research on Cancer; 2020:385–387.
7. Miettinen M, Wang ZF, Paetau A, et al. ERG transcription factor as an
32. Mesenchymal chondrosarcoma. In: Wei S, Siegal GP, eds. Atlas of Bone
immunohistochemical marker for vascular endothelial tumors and prostatic Pathology. New York, NY: Springer; 2013:152–156.
carcinoma. Am J Surg Pathol. 2011;35(3):432–441. 33. Shakked RJ, Geller DS, Gorlick R, Dorfman HD. Mesenchymal
8. Sotoca AM, Prange KH, Reijnders B, et al. The oncofusion protein FUS-ERG chondrosarcoma: clinicopathologic study of 20 cases. Arch Pathol Lab Med.
targets key hematopoietic regulators and modulates the all-trans retinoic acid 2012;136(1):61–75.
signaling pathway in t(16;21) acute myeloid leukemia. Oncogene. 2016;35(15): 34. Wang L, Motoi T, Khanin R, et al. Identification of a novel, recurrent HEY1-
1965–1976. NCOA2 fusion in mesenchymal chondrosarcoma based on a genome-wide
9. Miettinen M, Wang Z, Sarlomo-Rikala M, Abdullaev Z, Pack SD, Fetsch JF. screen of exon-level expression data. Genes Chromosomes Cancer. 2012;51(2):
ERG expression in epithelioid sarcoma: a diagnostic pitfall. Am J Surg Pathol. 127–139.
2013;37(10):1580–1585. 35. Nyquist KB, Panagopoulos I, Thorsen J, et al. Whole-transcriptome
10. Hung YP, Fletcher CD, Hornick JL. Evaluation of NKX2-2 expression in sequencing identifies novel IRF2BP2-CDX1 fusion gene brought about by
round cell sarcomas and other tumors with EWSR1 rearrangement: imperfect translocation t(1;5)(q42;q32) in mesenchymal chondrosarcoma. PLoS One.
specificity for Ewing sarcoma. Mod Pathol. 2016;29(4):370–380. 2012;7(11):e49705.
11. Bridge RS, Rajaram V, Dehner LP, Pfeifer JD, Perry A. Molecular diagnosis of 36. Meijer D, de Jong D, Pansuriya TC, et al. Genetic characterization of
Ewing sarcoma/primitive neuroectodermal tumor in routinely processed tissue: a mesenchymal, clear cell, and dedifferentiated chondrosarcoma. Genes Chromo-
comparison of two FISH strategies and RT-PCR in malignant round cell tumors. somes Cancer. 2012;51(10):899–909.
Mod Pathol. 2006;19(1):1–8. 37. Damato S, Alorjani M, Bonar F, et al. IDH1 mutations are not found in
12. Wang L, Bhargava R, Zheng T, et al. Undifferentiated small round cell cartilaginous tumours other than central and periosteal chondrosarcomas and
sarcomas with rare EWS gene fusions: identification of a novel EWS-SP3 fusion enchondromas. Histopathology. 2012;60(2):363–365.
and of additional cases with the EWS-ETV1 and EWS-FEV fusions. J Mol Diagn. 38. Rudzinski ER. Embryonal rhabdomyosarcoma. In: Antonescu CR, Bridge JA,
2007;9(4):498–509. Cunha IW, et al, eds. WHO Classification of Tumors: Soft Tissue and Bone. 5th
13. Le Loarer F, Szuhai K, Tirode F. Round cell sarcoma with EWSR1-non-ETS ed. Lyon, France: International Agency for Research on Cancer; 2020:201–204.
fusions. In: Bridge J, ed. WHO Classification of Tumors: Soft Tissue and Bone. 5th 39. Rudzinski ER, Teot LA, Anderson JR, et al. Dense pattern of embryonal
ed. Lyon, France: International Agency for Research on Cancer; 2020:326–329. rhabdomyosarcoma, a lesion easily confused with alveolar rhabdomyosarcoma: a
14. Johnson A, Severson E, Gay L, et al. Comprehensive genomic profiling of report from the Soft Tissue Sarcoma Committee of the Children’s Oncology
282 pediatric low- and high-grade gliomas reveals genomic drivers, tumor Group. Am J Clin Pathol. 2013;140(1):82–90.

58 Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal
40. Wei S, Henderson-Jackson E, Qian X, Bui MM. Soft tissue tumor 51. Arbajian E, Puls F, Magnusson L, et al. Recurrent EWSR1-CREB3L1 gene
immunohistochemistry update: illustrative examples of diagnostic pearls to avoid fusions in sclerosing epithelioid fibrosarcoma. Am J Surg Pathol. 2014;38(6):801–
pitfalls. Arch Pathol Lab Med. 2017;141(8):1072–1091. 808.
41. Kohashi K, Bode-Lesniewska B. Alveolar rhabdomyosarcoma. In: Anto- 52. Arbajian E, Puls F, Antonescu CR, et al. In-depth genetic analysis of
nescu CR, Bridge JA, Cunha IW, et al, eds. WHO Classification of Tumors: Soft sclerosing epithelioid fibrosarcoma reveals recurrent genomic alterations and
Tissue and Bone. 5th ed. Lyon, France: International Agency for Research on potential treatment targets. Clin Cancer Res. 2017;23(23):7426–7434.
Cancer; 2020:205–208. 53. Thway K, Nielsen TO. Myxoid liposarcoma. In: Antonescu CR, Bridge JA,
42. Duan F, Smith LM, Gustafson DM, et al. Genomic and clinical analysis of Cunha IW, et al, eds. WHO Classification of Tumors: Soft Tissue and Bone. 5th
fusion gene amplification in rhabdomyosarcoma: a report from the Children’s ed. Lyon, France: International Agency for Research on Cancer; 2020:42–44.
54. Antonescu CR, Elahi A, Healey JH, et al. Monoclonality of multifocal
Oncology Group. Genes Chromosomes Cancer. 2012;51(7):662–674.
myxoid liposarcoma: confirmation by analysis of TLS-CHOP or EWS-CHOP
43. Agaram NP, Szuhai K. Spindle cell/sclerosing rhabdomyosarcoma. In:
rearrangements. Clin Cancer Res. 2000;6(7):2788–2793.
Antonescu CR, Bridge JA, Cunha IW, et al, eds. WHO Classification of Tumors: 55. Horvai AE, Agaram NP, Lucas DR. Extraskeletal myxoid chondrosarcoma.
Soft Tissue and Bone. 5th ed. Lyon, France: International Agency for Research on In: Antonescu CR, Bridge JA, Cunha IW, et al, eds. WHO Classification of Tumors:
Cancer; 2020:211–213. Soft Tissue and Bone. 5th ed. Lyon, France: International Agency for Research on
44. Agaram NP, Zhang L, Sung YS, et al. Expanding the spectrum of Cancer; 2020:303–305.
intraosseous rhabdomyosarcoma: correlation between 2 distinct gene fusions 56. Finos L, Righi A, Frisoni T, et al. Primary extraskeletal myxoid
and phenotype. Am J Surg Pathol. 2019;43(5):695–702. chondrosarcoma of bone: report of three cases and review of the literature.
45. Agaram NP, Antonescu CR, Ladanyi M. Desmoplastic small round cell Pathol Res Pract. 2017;213(5):461–466.
tumor. In: Antonescu CR, Bridge JA, Cunha IW, et al, eds. WHO Classification of 57. Agaram NP, Zhang L, Sung YS, Singer S, Antonescu CR. Extraskeletal myxoid
Tumors: Soft Tissue and Bone. 5th ed. Lyon, France: International Agency for chondrosarcoma with non-EWSR1-NR4A3 variant fusions correlate with rhabdoid
Research on Cancer; 2020:306–308. phenotype and high-grade morphology. Hum Pathol. 2014;45(5):1084–1091.
46. Hill DA, Pfeifer JD, Marley EF, et al. WT1 staining reliably differentiates 58. Broehm CJ, Wu J, Gullapalli RR, Bocklage T. Extraskeletal myxoid
desmoplastic small round cell tumor from Ewing sarcoma/primitive neuroecto- chondrosarcoma with a t(9;16)(q22;p11.2) resulting in a NR4A3-FUS fusion.
dermal tumor: an immunohistochemical and molecular diagnostic study. Am J Cancer Genet. 2014;207(6):276–280.
Clin Pathol. 2000;114(3):345–353. 59. Urbini M, Astolfi A, Pantaleo MA, et al. HSPA8 as a novel fusion partner of
47. Barnoud R, Sabourin JC, Pasquier D, et al. Immunohistochemical NR4A3 in extraskeletal myxoid chondrosarcoma. Genes Chromosomes Cancer.
expression of WT1 by desmoplastic small round cell tumor: a comparative study 2017;56(7):582–586.
60. Jo VY, Antonescu CR, Horiuchi K, Patel RM. Myoepithelioma, myoepithe-
with other small round cell tumors. Am J Surg Pathol. 2000;24(6):830–836.
lial carcinoma, and mixed tumor. In: Antonescu CR, Bridge JA, Cunha IW, et al,
48. Arnold MA, Schoenfield L, Limketkai BN, Arnold CA. Diagnostic pitfalls of
eds. WHO Classification of Tumors: Soft Tissue and Bone. 5th ed. Lyon, France:
differentiating desmoplastic small round cell tumor (DSRCT) from Wilms tumor International Agency for Research on Cancer; 2020:277–279.
(WT): overlapping morphologic and immunohistochemical features. Am J Surg 61. Bergh P, Meis-Kindblom JM, Gherlinzoni F, et al. Synovial sarcoma:
Pathol. 2014;38(9):1220–1226. identification of low and high risk groups. Cancer. 1999;85(12):2596–2607.
49. Doyle LA, Mertens F. Sclerosing epithelioid fibrosarcoma. In: Antonescu 62. Krane JF, Bertoni F, Fletcher CD. Myxoid synovial sarcoma: an
CR, Bridge JA, Cunha IW, et al, eds. WHO Classification of Tumors: Soft Tissue underappreciated morphologic subset. Mod Pathol. 1999;12(5):456–462.
and Bone. 5th ed. Lyon, France: International Agency for Research on Cancer; 63. Nunez AL, Siegal GP, Reddy VV, Wei S. CD138 (syndecan-1) expression in
2020:130–132. bone-forming tumors. Am J Clin Pathol. 2012;137(3):423–428.
50. Bai S, Jhala N, Adsay NV, Wei S. Sclerosing epithelioid fibrosarcoma of the 64. Nadal R, Schweizer M, Kryvenko ON, Epstein JI, Eisenberger MA. Small
pancreas. Ann Diagn Pathol. 2013;17(2):214–216. cell carcinoma of the prostate. Nat Rev Urol. 2014;11(4):213–219.

Arch Pathol Lab Med—Vol 146, January 2022 Small Round Cell Tumors—Wei & Siegal 59

You might also like